WO2022251844A1 - Polymeric nanoparticles comprising chemotherapeutic compounds and related methods - Google Patents

Polymeric nanoparticles comprising chemotherapeutic compounds and related methods Download PDF

Info

Publication number
WO2022251844A1
WO2022251844A1 PCT/US2022/072563 US2022072563W WO2022251844A1 WO 2022251844 A1 WO2022251844 A1 WO 2022251844A1 US 2022072563 W US2022072563 W US 2022072563W WO 2022251844 A1 WO2022251844 A1 WO 2022251844A1
Authority
WO
WIPO (PCT)
Prior art keywords
peg
pla
cancer
composition
block copolymer
Prior art date
Application number
PCT/US2022/072563
Other languages
French (fr)
Inventor
Surender Kharbanda
Harpal Singh
Original Assignee
Hillstream Biopharma, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hillstream Biopharma, Inc. filed Critical Hillstream Biopharma, Inc.
Publication of WO2022251844A1 publication Critical patent/WO2022251844A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This disclosure relates to polymeric nanoparticles comprising an anthracycline chemotherapeutic compound and salinomycin and related methods of using the polymeric nanoparticles, treating of cancer, and making the polymeric nanoparticles.
  • chemotherapeutic agents used in the treatment of cancer can suffer from resistance of the cancer cells to the chemotherapeutic agents or from toxicity induced in healthy cells/tissues. Delivery of anticancer drugs would be more effective if the delivery system was able to effectuate treatment with smaller amounts of drugs and/or new combinations of drugs to mitigate resistance. There is a pressing need for such delivery systems.
  • this disclosure provides a composition comprising polymeric nanoparticles comprising block copolymers comprising a poly(lactic acid)-poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA-PEG-PPG-PEG- PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di block copolymer; salinomycin; and an anthracycline chemotherapeutic compound.
  • the salinomycin and the anthracycline chemotherapeutic compound are associated with the polymeric nanoparticles.
  • the salinomycin and the anthracycline chemotherapeutic compound are both associated substantially with the same polymeric nanoparticles.
  • the anthracycline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof.
  • the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin.
  • the anthracycline chemotherapeutic compound is doxorubicin.
  • the anthracycline chemotherapeutic compound is pirarubicin.
  • the total mass of anthracycline chemotherapeutic compound is greater than or equal to the total mass of the salinomycin.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1.5:1.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 2: 1.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 2.5:1 to 3.5:1.
  • the total mass of anthracycline chemotherapeutic compound is less than or equal to the total mass of the salinomycin.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1:1.5.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1 :2.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 1:2.5 to 1:3.5.
  • the total mass of anthracycline chemotherapeutic compound and is about equal to the total mass of the salinomycin.
  • PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than PLA-PEG di-block copolymer.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:20 to 1:1.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1 : 15 to 1 :2.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:8 to 1:10. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG- PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1 :3 to 1 :5. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:2 to 3:8.
  • the average diameter of the polymeric nanoparticles is between 50 and 170 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 130 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 100 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 80 and 110 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 100 and 170 nm.
  • a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.5. In some embodiments, a PDI of the polymeric nanoparticles is not more than 0.3.
  • a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV.
  • the composition further comprises a PEG-PPG-PEG triblock copolymer.
  • a pharmaceutical composition comprises the composition.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
  • the present disclosure provides a method of reducing proliferation, survival, migration, or colony formation ability of a rapidly proliferating cell in a subject in need thereof, comprising contacting the cell with a composition comprising a therapeutically effective amount of the composition or the pharmaceutical composition.
  • the cell is a cancer cell.
  • the cell is a cancer stem cell.
  • the present disclosure provides a method for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a composition comprising the composition or the pharmaceutical composition.
  • the cancer comprises a solid tumor cancer or a cancer of the blood.
  • the cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, leukemia, lymphoma, osteosarcoma, gastric cancer, prostate cancer, colon cancer, non-small cell lung cancer and small cell lung cancer, liver cancer, kidney cancer, head and neck cancer, cervical cancer, acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, and combinations thereof.
  • the cancer or breast cancer comprises triple negative breast cancer (TNBC).
  • TNBC triple negative breast cancer
  • the cancer is metastatic.
  • the method further comprises administering an additional anti-cancer therapy to the subject.
  • the additional anti-cancer therapy comprises surgery, chemotherapy, radiation, hormone therapy, immunotherapy, or a combination thereof.
  • the cancer is resistant or refractory to a chemotherapeutic agent.
  • the subject is a human.
  • the composition or pharmaceutical composition is administered intravenously, intratumorally, or subcutaneously.
  • the present disclosure provides a method of manufacturing polymeric nanoparticles comprising: a) mixing penta-block PLA-PEG-PPG-PEG-PLA and di-block PLA-PEG hybrid block copolymers dissolved in acetonitrile with salinomycin and at least one anthracycline compound to form a first mixture; b) mixing the first mixture with a PEG-PPG-PEG triblock copolymer dissolved in water to form a second mixture; c) stirring the second mixture and evaporating the acetonitrile; and d) filtering the stirred and evaporated second mixture, thereby manufacturing the polymeric nanoparticles.
  • the salinomycin and at least one anthracycline chemotherapeutic compound are dissolved in DMSO when used in step a).
  • the PEG-PPG-PEG triblock copolymer comprises poloxamer 407.
  • the method further comprises adding triethylamine during step b).
  • the anthracycline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof.
  • the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin.
  • the anthracycline chemotherapeutic compound is doxorubicin.
  • the anthracycline chemotherapeutic compound is pirarubicin.
  • the total mass of anthracycline chemotherapeutic compound is greater than or equal to the total mass of the salinomycin.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1.5:1.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 2:1.
  • the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 2.5:1 to 3.5:1.
  • the total mass of anthracycline chemotherapeutic compound is less than or equal to the total mass of the salinomycin. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1:1.5. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1 :2. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 1:2.5 to 1:3.5. In some embodiments, the total mass of anthracycline chemotherapeutic compound is about equal to the total mass of the salinomycin.
  • PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than PLA-PEG di-block copolymer.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:20 to 1:1.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:15 to 1:2.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:8 to 1:10. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG- PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1 :3 to 1 :5. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:2 to 3:8.
  • the average diameter of the polymeric nanoparticles is between 50 and 170 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 130 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 100 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 80 and 110 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 100 and 170 nm.
  • a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.5. In some embodiments, a PDI of the polymeric nanoparticles is not more than 0.3.
  • a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV.
  • FIG. 1A shows a chemical structure of epirubicin (EPI).
  • FIG. IB shows a chemical structure of idarubicin (IDA).
  • FIG. 1C shows a chemical structure of daunorubicin (DNR).
  • FIG. ID shows a chemical structure of doxorubicin (DOX).
  • FIG. IE shows a chemical structure of pirarubicin (PIRA).
  • FIG. IF shows a chemical structure of nemorubicin (NEMO).
  • FIG. 2 is a plot showing data from an in vitro release assay for NPs containing the indicated drugs.
  • FIG. 3 is a plot showing data from an in vitro cell proliferation assay for NPs containing the indicated drugs.
  • FIG. 4 is a plot showing tumor volume data over time from a tumor regression study that treated mice using NPs containing the indicated drugs.
  • FIG. 5 is a plot showing data from an in vitro cell proliferation assay for free pirarubicin and doxorubicin with no NPs included.
  • FIG. 6A is a plot showing data from an in vitro release assay for NPs containing pirarubicin.
  • FIG. 6B is a plot showing data from an in vitro release assay for NPs containing pirarubicin at a pH of 7.4 and a pH of 5.
  • FIG. 7A is a plot showing data from an in vitro cell proliferation assay performed with SUM149 cells. Either NPs containing pirarubicin or free pirarubicin were used.
  • FIG. 7B is a plot showing data from an in vitro cell proliferation assay performed with MDA-MB 468 cells. Either NPs containing pirarubicin or free pirarubicin were used.
  • FIG. 8A is a plot showing data from an in vitro cell proliferation assay using SEIM149 cells and NPs containing the indicated drugs.
  • FIG. 8B is a plot showing data from an in vitro cell proliferation assay using MDA-MB 468 cells and NPs containing the indicated drugs.
  • FIG. 9A is a plot showing data from an in vitro cell proliferation assay using SEIM149 cells and NPs containing the indicated drugs.
  • FIG. 9B is a plot showing data from an in vitro cell proliferation assay using MDA-MB 468 cells and NPs containing the indicated drugs.
  • the articles “a” and “an” refer to one or to more than one (i.e., to at least one) of the grammatical object of the article.
  • an element means one element or more than one element.
  • use of the term “including” as well as other forms, such as “include,” “includes,” and “included,” is not limiting.
  • the term “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term “about” is meant to encompass variations of ⁇ 5% from the specified value, as such variations are appropriate to perform the disclosed methods.
  • the term “comprising” may include the embodiments “consisting of’ and “consisting essentially of.”
  • the terms “comprise(s),” “include(s),” “having,” “has,” “may,” “contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that require the presence of the named ingredients/steps and permit the presence of other ingredients/steps.
  • such description should be construed as also describing compositions or processes as “consisting of’ and “consisting essentially of’ the enumerated compounds, which allows the presence of only the named compounds, along with any pharmaceutically acceptable carriers, and excludes other compounds.
  • nanoparticle refers to particles in the range between 10 nm to 1000 nm in diameter, wherein diameter refers to the diameter of a perfect sphere having the same volume as the particle.
  • the term “nanoparticle” is used interchangeably with “nanoparti cle(s).” In some cases, a population of particles may be present. As used herein, the “diameter” of the nanoparticles is an average of a distribution in a particular population.
  • polymer is given its ordinary meaning as used in the art, i.e., a molecular structure comprising one or more repeat units (monomers), connected by covalent bonds. The repeat units may all be identical, or in some cases, there may be more than one type of repeat unit present within the polymer.
  • a "chemotherapeutic agent,” “therapeutic agent,” and/or “drug” is a biological (large molecule) or chemical (small molecule) compound useful in the treatment of cancer, regardless of mechanism of action.
  • Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, proteins, antibodies, photosensitizers, and kinase inhibitors.
  • combination refers to the combined administration of two or more therapeutic agents (e g., co-delivery).
  • Components of a combination therapy may be administered simultaneously or sequentially, i.e., at least one component of the combination is administered at a time temporally distinct from the other component(s).
  • a component(s) is administered within one month, one week, 1-6 days, 18, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2 , 1 hour, or 30, 20, 15, 10, or 5 minutes of the other component(s).
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues a warm-blooded animal, e.g., a mammal or human, without excessive toxicity, irritation allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
  • a "therapeutically effective amount" of a polymeric nanoparticle comprising one or more therapeutic agents is an amount sufficient to provide an observable or clinically significant improvement over the baseline clinically observable signs and symptoms of the disorders treated with the combination.
  • subject or "patient” as used herein is intended to include animals, which are capable of suffering from or afflicted with a cancer or any disorder involving, directly or indirectly, a cancer.
  • subjects include mammals, e.g., humans, apes, monkeys, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non human animals.
  • the subject is a human, e.g., a human suffering from cancer.
  • treating comprises a treatment relieving, reducing or alleviating at least one symptom in a subject or producing a delay in the progression of a disease.
  • treatment can be the diminishment of one or several symptoms of a disorder or complete eradication of a disorder, such as cancer.
  • the term “treat” also denotes to arrest and/or reduce the risk of worsening a disease.
  • prevent comprises the prevention of at least one symptom associated with or caused by the state, disease or disorder being prevented.
  • human equivalent dose refers to a dose of a composition to be administered to a human that is calculated from a specific dose used in an animal study.
  • rapidly proliferating cells refers to cells having the capacity for autonomous growth (e.g., cancer cells).
  • cancer stem cell refers to a cancer cell that has characteristics of a stem cell, such as giving rise to all cell types within a particular tumor type and the ability to self-renew.
  • the cancer stem cell is resistant or refractory to chemotherapy.
  • the term “associated substantially with” in the context of a nanoparticle means a substance is encapsulated or stably interacting with a nanoparticle.
  • the nanoparticle is loaded with both substances, as opposed to an embodiment where one substance is loaded into a first set of nanoparticles and a second substance is loaded into a second set of nanoparticles.
  • a substance is associated substantially with a nanoparticle, at least 80%, at least 90%, at least 95%, or at least 99% of the mass of the substance is encapsulated or stably interacting with the nanoparticle.
  • Nanoparticles can be produced as nanocapsules or nanospheres.
  • Drug loading in the nanoparticle can be performed by either an adsorption process or an encapsulation process (Spada et al., 2011; Protein delivery of polymeric nanoparticles; World Academy of Science, Engineering and Technology: 76, incorporated herein, by reference, in its entirety).
  • Nanoparticles, by using both passive and active targeting strategies, can enhance the intracellular concentration of drugs in cancer cells while avoiding toxicity in normal cells.
  • nanoparticles When nanoparticles bind to specific receptors and enter the cell, they are usually enveloped by endosomes via receptor-mediated endocytosis, thereby bypassing the recognition of P-glycoprotein, one of the main drug resistance mechanisms (Cho et al., 2008, Therapeutic Nanoparticles for Drug Delivery in Cancer, Clin. Cancer Res., 2008, 14: 1310-1316, incorporated herein, by reference, in its entirely).
  • Nanoparticles are removed from the body by opsonization and phagocytosis (Sosnik et al., 2008; Polymeric Nanocarriers: New Endeavors for the Optimization of the Technological Aspects of Drugs; Recent Patents on Biomedical Engineering, 1: 43-59, incorporated herein, by reference, in its entirety).
  • Nanocarrier based systems can be used for effective drug delivery with the advantages of improved intracellular penetration, localized delivery, protection of drugs against premature degradation, controlled pharmacokinetic and drug tissue distribution profile, lower dose requirement, and cost effectiveness (Farokhzad OC, et al.; Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc. Natl. Acad. Sci.
  • Nanoparticles are indirectly proportional to their small dimensions. Due to their small size, the polymeric nanoparticles have been found to evade recognition and uptake by the reticulo-endothelial system (RES), and can thus circulate in the blood for an extended period (Borchard et al., 1996, Pharm. Res. 7: 1055-1058, incorporated herein, by reference, in its entirety). Nanoparticles are also able to extravasate at the pathological site like the leaky vasculature of a solid tumor, providing a passive targeting mechanism. Due to the higher surface area leading to faster solubilization rates, nano-sized structures usually show higher plasma concentrations and area under the curve (AUC) values.
  • AUC area under the curve
  • Nanoparticle size affects drug release. Larger particles have slower diffusion of drugs into the system. Smaller particles offer larger surface area but lead to last drug release. Smaller particles tend to aggregate during storage and transportation of nanoparticle dispersions. Hence, a compromise between a small size and maximum stability of nanoparticles is desired.
  • the size of nanoparticles used in a drug delivery system should be large enough to prevent their rapid leakage into blood capillaries but small enough to escape capture by fixed macrophages that are lodged in the reticuloendothelial system, such as the liver and spleen.
  • Nanoparticles In addition to their size, the surface characteristics of nanoparticles are also an important factor in determining the life span during circulation. Nanoparticles should ideally have a hydrophilic surface to escape macrophage capture. Nanoparticles formed from block copolymers with hydrophilic and hydrophobic domains meet these criteria. Controlled polymer degradation also allows for increased levels of agent delivery to a diseased state. Polymer degradation can also be affected by the particle size. Degradation rates increase with increase in particle size in vitro (Biopolymeric nanoparticles; Sundar et al., 2010, Science and Technology of Advanced Materials; doi: 10.1088/1468-6996/11/1/014104, incorporated herein, by reference, in its entirety).
  • Poly(lactic acid) (PL A) has been approved by the US FDA for applications in tissue engineering, medical materials and drug carriers.
  • US2006/0165987A1 incorporated herein, by reference, in its entirety, describes a stealthy polymeric biodegradable nanosphere comprising poly(ester)-poly(ethylene) multiblock copolymers and optional components for imparting rigidity to the nanospheres and incorporating pharmaceutical compounds.
  • US2008/0081075A1 incorporated herein, by reference, in its entirety, discloses a novel mixed micelle structure with a functional inner core and hydrophilic outer shells, self- assembled from a graft macromolecule and one or more block copolymer.
  • US2010/0004398A1 incorporated herein, by reference, in its entirety, describes a polymeric nanoparticle of shell/core configuration with an interphase region and a process for producing the same.
  • polymeric nanoparticles for the delivery of chemotherapeutic compounds.
  • the inventors of the present disclosure have developed polymeric nanoparticles comprising formulations of chemotherapeutic compounds.
  • the polymeric nanoparticles are useful for the delivery of drugs.
  • the nanoparticles can find use in treatment of diseases exhibiting rapid cell division such as various cancers by delivering appropriate chemotherapeutic agents.
  • composition comprising: a) polymeric nanoparticles comprising hybrid block copolymers comprising a poly(lactic acid)- poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA- PEG-PPG-PEG-PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer; b) salinomycin; and c) an anthracycline chemotherapeutic compound. Both the salinomycin and the anthracycline chemotherapeutic compound are associated with the polymeric nanoparticles.
  • the salinomycin and the anthracycline chemotherapeutic compound can be associated with the polymeric nanoparticles by being contained within an enclosed region of a shell of polymer.
  • the drugs can be interspersed within the polymer that forms the shell, or the drugs can adhere to an outside surface of the shell.
  • the drugs can be associated with the polymeric nanoparticle in any manner suitable to carry and deliver the drugs to locations of disease in need of treatment.
  • the salinomycin and the anthracycline chemotherapeutic compound can both be associated substantially with the same polymeric nanoparticles. In some embodiments, the salinomycin and the anthracycline chemotherapeutic compound are encapsulated by the nanoparticle.
  • the polymeric nanoparticles can comprise both hydrophobic and hydrophilic block copolymers.
  • the polymeric nanoparticles provided herein comprise hybrid block copolymers comprising a poly(lactic acid)-poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA-PEG-PPG-PEG-PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer.
  • the PLA-PEG-PPG-PEG-PLA penta-block copolymer can be formed from PEG-PPG-PEG triblock copolymer and PLA via ring opening polymerization of the lactide.
  • the molecular weight of the penta- block copolymer can range from 5,000 g/mol to 40,000 g/mol.
  • the molecular weight range of di-block copolymer can be from 2,000 g/mol to 15,000 g/mol.
  • Poly(lactic acid) (PLA) is a hydrophobic polymer and can be a component of the polymeric nanoparticles.
  • poly(glycolic acid) (PGA) and block copolymer of poly lactic acid-co-glycolic acid (PLGA) may also be used.
  • the hydrophobic polymer can also comprise a biologically derived polymer or a biopolymer.
  • the molecular weight of the PLA used is generally in the range of about 2,000 g/mol to 80,000 g/mol.
  • the PLA used is in the range of about 10,000 g/mol to 80,000 g/mol.
  • the average molecular weight of PLA may also be about 70,000 g/mol.
  • one g/mol is equivalent to one “dalton” (i.e., dalton and g/mol are interchangeable when referring to the molecular weight of a polymer).
  • “Kilodalton” (or “kDa”) as used herein refers to 1,000 dal tons.
  • Polyethylene glycol) (PEG) is another preferred component of the polymer used to form the polymeric nanoparticles.
  • PEG can impart hydrophilicity, reduce phagocytosis by macrophages, and/or reduce immunological recognition.
  • Block copolymers like polyethylene glycol)-poly(propylene glycol)-poly(ethylene glycol) (PEG-PPG-PEG) are hydrophilic or hydrophilic-hydrophobic copolymers that can be components of the polymeric nanoparticles of the present disclosure.
  • the PLA-PEG-PPG-PEG-PLA penta-block copolymer can be formed from ring opening polymerization using lactide and also by using mPEG for the di-block.
  • block copolymers of the present disclosure may have two, three, four, five, or more distinct blocks.
  • the polymeric nanoparticles provided herein comprise a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer.
  • a first block copolymer of the instant disclosure consists essentially of two segments of poly(lactic acid) (PLA), separated by a segment of poly(ethylene glycol)-polypropylene glycol)-poly(ethylene glycol) (PEG-PPG-PEG), to form the PLA-PEG-PPG-PEG-PLA penta-block copolymer.
  • PLA poly(lactic acid)
  • PEG-PPG-PEG poly(ethylene glycol)-polypropylene glycol)-poly(ethylene glycol)
  • a second block copolymer of the instant disclosure consists essentially of a PLA-PEG di-block copolymer.
  • the first and second block copolymers of the instant disclosure can be combined to form the polymeric nanoparticles of the instant disclosure.
  • the process described in Example 1 of the present disclosure can be used to accomplish the combination.
  • the polymeric nanoparticles of the instant disclosure can be biodegradable.
  • the nanoparticles comprise QUATRAMERTM reagent, which comprises a PLA-PEG-PPG-PEG-PLA penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer.
  • QUATRAMERTM reagent is available from Hillstream Biopharma; Bridgewater, NJ, USA.
  • the PLA-PEG-PPG-PEG-PLA penta-block copolymer and the PLA-PEG di block copolymer may optionally be combined in specific ratios. As used herein, such ratios are expressed in the form of Mass pe nta-biock:Massdi-biock, unless stated otherwise. In some embodiments, less PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than the PLA-PEG di -block copolymer. In some embodiments, a mass ratio of PLA-PEG- PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be from 1 :20 to 1:1.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer can be from 1:15 to 1:2. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer can be from 1 : 10 to 1:2. In some embodiments, a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer can be from 1 :8 to 1:10 or can be about 1 :9.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta- block copolymer to PLA-PEG di-block copolymer can be from 1 :3 to 1 :5 or can be about 1 :4. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be from 1 :2 to 3 :8 or can be about 3:7.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be at least 1 :20 such as, for example, at least 1 : 19, at least 1 : 18, at least 1 : 17, at least 1 : 16, at least 1 : 15, at least 1 : 14, at least 1 : 13, at least 1 : 12, at least 1 : 11, at least 1 : 10, at least 1 :9, at least 1 :8, at least 1 :7, at least 1 :6, at least 1:5, at least 1 :4, at least 1 :3, or at least 1 :2.
  • a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be not more than 1 : 1 such as, for example, not more than 1 :2, not more than 1:3, not more than 1 :4, not more than 1:5, not more than 1 :6, not more than 1 :7, not more than 1 :8, not more than 1 :9, not more than 1:10, not more than 1:11, not more than 1:12, not more than 1:13, not more than 1:14, not more than 1:15, not more than 1:16, not more than 1:17, not more than 1:18, or not more than 1:19.
  • the polymeric nanoparticles of the instant disclosure have, in various embodiments, a diameter that is an average of a distribution of nanoparticles in a particular population.
  • the polymeric nanoparticles have dimensions that can be measured using a transmission electron microscope, or another suitable technique that can allow for measurements of the diameters of a sample of a population of polymeric nanoparticles.
  • the diameter of the nanoparticles can be at least 50 nm such as, for example, at least 60 nm, at least 70 nm, at least 80 nm, at least 90 nm, at least 100 nm, at least 110 nm, at least 120 nm, at least 130 nm, at least 140 nm, at least 150 nm, or at least 160 nm.
  • the diameter of the nanoparticles can be not more than 170 nm such as, for example, not more than 160 nm, not more than 150 nm, not more than 140 nm, not more than 130 nm, not more than 120 nm, not more than 110 nm, not more than 100 nm, not more than 90 nm, not more than 80 nm, not more than 70 nm, or not more than 60 nm.
  • the diameter of the nanoparticles can range from 50 nm to 170 nm such as, for example, from 60 nm to 130 nm, from 60 nm to 100 nm, from 80 nm to 110 nm, from 90 to 130 nm, from 100 to 170 nm, or any other suitable range, based on the properties of the polymeric nanoparticles (e.g., the precise drugs associated therewith).
  • a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.50 such as, for example, not more than 0.45, not more than 0.40, not more than 0.35, not more than 0.30, not more than 0.25, not more than 0.20, not more than 0.15, not more than 0.10, or not more than 0.05. In some embodiments, the PDI is from 0.05 to 0.2. As used herein, the PDI is a ratio of the mass average molar mass of the penta- and di-block copolymers in the polymeric nanoparticles to the number average molar mass of the penta- and di-block copolymers in the polymeric nanoparticles. PDI may also be referred to simply as, “dispersity.”
  • Number average molar mass is defined as below: where Ni is the number of molecules of molecular mass Mu
  • Mass average molar mass is defined as below: where Ni is the number of molecules of molecular mass Mu
  • mass average molar mass and number average molar mass can be measured by any suitable process such as, for example, gel permeation chromatography, viscometry via the Mark-Houwink equation, or colligative methods (for number average molar mass); or static light scattering, small angle neutron scattering, X-ray scattering, or sedimentation velocity (for number average molar mass).
  • a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV such as, for example, -5 mV to -30 mV, -5 to -25 mV, or -5 to -15 mV.
  • zeta potential is a measure of the electrical potential difference at the slipping plane.
  • the slipping plane is the interface of mobile fluid around a particle (e.g., a polymeric nanoparticle of the present disclosure) with fluid components that remain attached to the particle surface (e.g., via adsorption and/or electrostatic interaction).
  • the zeta potential and PDI (Polydispersity Index) of the nanoparticles may be calculated (see U.S. patent number 9,149,426, incorporated herein by reference, in its entirety).
  • compositions provided herein can comprise one or more chemotherapeutic compounds.
  • the polymeric nanoparticles can associate with the chemotherapeutic compounds.
  • the polymeric nanoparticles can encapsulate the chemotherapeutic compounds and/or adsorb to the chemotherapeutic compounds.
  • the polymeric nanoparticles can associate with the chemotherapeutic compounds in any manner suitable to carry the chemotherapeutic compounds throughout a subject’s body and deliver the chemotherapeutic compounds to a diseased cell (e.g., a rapidly dividing cell such as a cancer cell).
  • the chemotherapeutic compounds comprise salinomycin and an anthracycline chemotherapeutic compound.
  • the anthracycline chemotherapeutic compound can comprise doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin, or a combination thereof. Suitable examples of anthracycline chemotherapeutic compounds are shown with their structures in FIG. 1 A-FIG. IF.
  • the abbreviations refer to: epirubicin (EPI, FIG. 1A), idarubicin (IDA, FIG. IB), daunorubicin (DNR, FIG. 1C), doxorubicin (DOX, FIG. ID), pirarubicin (PIRA, FIG. IE), and nemorubicin (NEMO, FIG. IF).
  • the inventors of the present disclosure have determined that when the salinomycin and the anthracycline chemotherapeutic compound are both included in a composition of the present disclosure (e.g ., associated with polymeric nanoparticles of the present disclosure), improved performance (e.g., improved growth inhibition of diseased cells) can be obtained, compared to the activity of the individual drugs.
  • the total mass of anthracycline chemotherapeutic compound can be greater than or equal to the total mass of the salinomycin.
  • the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of at least 1.1:1, at least 1.2:1, at least 1.3:1, at least 1.4:1, at least 1.5:1, at least 2:1, at least 2.5:1, or at least 3:1.
  • the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of 2.5:1 to 3.5:1, or a mass ratio of 2:1 to 4:1.
  • the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of about 3:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of not more than 4:1, not more than 5:1, or not more than 6:1. [0106] In some embodiments, the total mass of anthracycline chemotherapeutic compound can be less than or equal to the total mass of the salinomycin.
  • the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of not more than 1:1.5, not more than 1:1.6, not more than 1:1.7, not more than 1:1.8, not more than 1:1.9, not more than 1 :2, not more than 1 :2.5, or not more than 1:3.
  • the anthracy cline chemotherapeutic compound and the salinomycin can be present in a mass ratio of 1 :2.5 to 1 :3.5, or a mass ratio of 1 :2 to 1 :4.
  • the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of about 1:3.
  • the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of at least 1:4, at least 1:5, or at least 1:6.
  • the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of about 1:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of 1:1.
  • preparation of polymeric nanoparticles can optionally involve addition of a PEG-PPG-PEG triblock copolymer.
  • the inventors of the present disclosure have found that including such a triblock copolymer can improve the stability of the polymeric nanoparticles and/or can serve as an emulsifier for other components.
  • the triblock copolymer may be associated with or associated substantially with the polymeric nanoparticles. In some embodiments, the triblock copolymer may not be associated with the polymeric nanoparticles.
  • the triblock copolymer comprises a central polypropylene glycol) block flanked by two poly(ethylene glycol) blocks. In some embodiments, the triblock copolymer comprises poloxamer 407.
  • the present disclosure provides a method of manufacturing nanoparticles comprising: a) mixing penta-block PLA-PEG-PPG-PEG-PLA and di-block PLA-PEG hybrid block copolymers dissolved in acetonitrile with salinomycin and at least one anthracycline compound to form a first mixture; b) mixing the first mixture with a PEG-PPG-PEG tri-block copolymer dissolved in water to form a second mixture; c) stirring the second mixture and evaporating the acetonitrile; and d) filtering the stirred and evaporated second mixture, thereby manufacturing the nanoparticles.
  • the salinomycin and at least one anthracycline chemotherapeutic compound are dissolved in DMSO when used in step a).
  • the DMSO may be evaporated during step c).
  • the PEG- PPG-PEG triblock copolymer comprises poloxamer 407.
  • the method further comprises adding triethylamine during step b).
  • the penta-block and di -block copolymers are present in the acetonitrile at 10 mg to 40 mg per mL of acetonitrile. In some embodiments, the penta-block and di-block copolymers are present in the acetonitrile at 15 mg to 30 mg per mL of acetonitrile. In some embodiments, the penta-block and di-block copolymers are present in the acetonitrile at about 20 mg per mL of acetonitrile.
  • the at least one anthracycline compound and salinomycin are dissolved in DMSO such that the total mass of drugs is dissolved at 0.05 mg per pL of DMSO to 0.2 mg per pL of DMSO. In some embodiments, the at least one anthracycline compound and salinomycin are dissolved in DMSO such that the total mass of drugs is dissolved at about 0.1 mg per pL of DMSO.
  • the PEG-PPG-PEG tri -block copolymer is dissolved in the water at 2.5 mg of PEG-PPG-PEG tri -block copolymer per mL of water to 10 mg of PEG- PPG-PEG tri-block copolymer per mL of water. In some embodiments, the PEG-PPG-PEG tri-block copolymer is dissolved in the water at 3.5 mg of PEG-PPG-PEG tri-block copolymer per mL of water to 7.5 mg of PEG-PPG-PEG tri -block copolymer per mL of water. In some embodiments, the PEG-PPG-PEG tri-block copolymer is dissolved in the water at about 5 mg per mL of water.
  • the PEG-PPG-PEG tri -block copolymer comprises poloxamer 407.
  • the PEG-PPG-PEG tri-block copolymer comprises PLURONIC® F127.
  • PLURONIC® F127 and poloxamer 407 both comprise a triblock copolymer comprising a central polypropylene glycol) block flanked by two poly(ethylene glycol) blocks. The approximate lengths of the two PEG blocks can be 101 repeat units, while the approximate length of the propylene glycol block can be 56 repeat units.
  • PLURONIC® F127 is available from BASF SE, Ludwigshafen, Germany.
  • both PLURONIC® F127 and poloxamer 407 comprise the same tri-block copolymer, but they may vary from each other based on their respective molecular weights and/or the number of monomers in each of their blocks.
  • the PLURONIC® F 127 and/or poloxamer 407 can comprise a molecular weight of from 10,500 g/mol to 14,500 g/mol such as, for example a molecular weight of about 12,600 g/mol.
  • the triethylamine is added in an amount of 0.5 pL to 2 pL for every 4 mL of water.
  • the triethylamine is added in an amount of about 0.5 pL for every 4 mL of water. In some embodiments, the triethylamine is added in an amount of about 1 pL for every 4 mL of water. In some embodiments, the triethylamine is added in an amount of about 2 mL for every 4 mL of water.
  • the anthracy cline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof.
  • the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin.
  • the anthracycline chemotherapeutic compound is doxorubicin.
  • the anthracycline chemotherapeutic compound is pirarubicin.
  • the penta- block and di-block copolymers may be prepared and used as reagents for preparation of the polymeric nanoparticles. These can be prepared as described in Example 1 from a poloxamer copolymer such as poloxamer 181 (for the penta-block copolymer), methoxypoly(ethylene glycol) (mPEG) (for the di-block copolymer), initiator, and PLA.
  • a poloxamer copolymer such as poloxamer 181 (for the penta-block copolymer), methoxypoly(ethylene glycol) (mPEG) (for the di-block copolymer), initiator, and PLA.
  • mPEG methoxypoly(ethylene glycol)
  • initiator for the di-block copolymer
  • PLA a ring-opening polymerization of lactide in the presence of a Sn-catalyst can be employed, or any other suitable technique as determined by the skilled artisan.
  • the poloxamer 181 can comprise a molecular weight of 1,000 g/mol to 3,000 g/mol. In some embodiments, the poloxamer 181 can comprise a molecular weight of about 2,000 g/mol. The appropriate molecular weight can be selected in order to, for example, improve the properties ( e.g ., stability) of the polymeric nanoparticles.
  • a pharmaceutical composition comprising the polymeric nanoparticle compositions described herein for use in medicine and in other fields that use a carrier system or a reservoir or depot of nanoparticles.
  • the polymeric nanoparticles can be used in prognostic, therapeutic, diagnostic and/or theranostic compositions.
  • the nanoparticles of the present disclosure are used for drug and agent delivery (e.g., within a tumor cell), as well as for disease diagnosis and medical imaging in human and animals.
  • the instant disclosure provides a method for the treatment of disease using the nanoparticles, further comprising a chemotherapeutic agent, as described herein.
  • the nanoparticles of the present disclosure can also be use in other applications such as chemical or biological reactions where a reservoir or depot is required, as biosensors, as agents for immobilized enzymes and the like.
  • a pharmaceutical composition comprising a) polymeric nanoparticles comprising hybrid block copolymers comprising a PLA-PEG-PPG- PEG-PLA penta-block copolymer with a PLA-PEG di-block copolymer; b) salinomycin; and c) an anthracycline chemotherapeutic compound. Both the salinomycin and the anthracycline chemotherapeutic compound are associated with the polymeric nanoparticles.
  • Suitable pharmaceutical compositions or formulations can contain, for example, from about 0.1% to about 99.9%, preferably from about 1% to about 60%, of the active ingredient(s).
  • Pharmaceutical formulations for enteral or parenteral administration are, for example, those in unit dosage forms, such as sugar-coated tablets, tablets, capsules or suppositories, or ampoules. If not indicated otherwise, these are prepared in a manner known per se, for example by means of conventional mixing, granulating, sugar-coating, dissolving or lyophilizing processes. It will be appreciated that the unit content of a combination partner contained in an individual dose of each dosage form need not in itself constitute an effective amount since the necessary effective amount may be reached by administration of a plurality of dosage units.
  • the pharmaceutical compositions can contain, as the active ingredient, one or more of nanoparticles in combination with one or more pharmaceutically acceptable carriers (excipients).
  • the active ingredient is typically mixed with an excipient, diluted by an excipient, or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • excipients include lactose (e.g. lactose monohydrate), dextrose, sucrose, sorbitol, mannitol, starches (e.g. sodium starch glycolate), gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, colloidal silicon dioxide, microcrystalline cellulose, polyvinylpyrrolidone (e.g. povidone), cellulose, water, syrup, methyl cellulose, and hydroxypropyl cellulose.
  • lactose e.g. lactose monohydrate
  • dextrose sucrose
  • sorbitol sorbitol
  • mannitol starches
  • gum acacia calcium phosphate
  • alginates alginates
  • tragacanth gelatin
  • calcium silicate colloidal silicon dioxide
  • microcrystalline cellulose e.g. povidone
  • polyvinylpyrrolidone e.g. povidone
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy- benzoates; sweetening agents; and flavoring agents.
  • lubricating agents such as talc, magnesium stearate, and mineral oil
  • wetting agents such as talc, magnesium stearate, and mineral oil
  • emulsifying and suspending agents such as methyl- and propylhydroxy- benzoates
  • preserving agents such as methyl- and propylhydroxy- benzoates
  • sweetening agents such as methyl- and propylhydroxy- benzoates
  • liquid forms in which the compounds and compositions of the present disclosure can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • polymeric nanoparticles and pharmaceutical compositions disclosed herein can be used to treat or prevent any condition or disorder which is known to or suspected of benefitting from treatment with salinomycin and anthracy cline chemotherapeutic compounds.
  • the polymeric nanoparticles and/or pharmaceutical compositions disclosed herein can be used to reduce proliferation, survival, migration, or colony formation ability of a rapidly proliferating cell in a subject in need thereof. This can be accomplished by contacting the cell with a therapeutically effective amount of the polymeric nanoparticles and/or pharmaceutical compositions. Such a method can be conducted in vivo (e.g, in a cancer patient), in vitro , or ex vivo.
  • the cell can be a cancer cell or a cancer stem cell.
  • the polymeric nanoparticles and/or pharmaceutical compositions disclosed herein can be used to treat or prevent cancer or a precancerous condition.
  • the cancer can be, a cancer cell or a cancer stem cell.
  • the cancer can be a solid tumor cancer or a cancer of the blood.
  • the cancer can be selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, leukemia, lymphoma, osteosarcoma, gastric cancer, prostate cancer, colon cancer, non-small cell lung cancer and small cell lung cancer, liver cancer, kidney cancer, head and neck cancer, cervical cancer, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, multiple myeloma, and combinations thereof.
  • the cancer can comprise triple negative breast cancer (TNBC).
  • TNBC triple negative breast cancer
  • the cancer can be metastatic cancer.
  • the cancer may be an affliction of a subject.
  • the subject may be a human.
  • the treatment using the polymeric nanoparticles or pharmaceutical composition comprising them can comprise administration of an additional anti-cancer therapy.
  • the additional anti-cancer therapy can comprise any medically suitable therapy that could be combined with the polymeric nanoparticles disclosed herein. Such combinations of therapies can increase the overall effectiveness of cancer treatments.
  • the additional anti-cancer therapy can comprise surgery; chemotherapy, radiation, hormone therapy, immunotherapy, or a combination thereof.
  • Additional anti-cancer therapies that may be combined with the polymeric- nanoparticle-based therapies disclosed herein include: lenalidomide, crizotinib or a histone deacetylase (HD AC) inhibitor , such as those disclosed in US Patent No. 8,883,842, incorporated by reference herein, in its entirety.
  • HD AC histone deacetylase
  • Additional anti-cancer therapies that may be combined with the polymeric-nanoparticle-based therapies disclosed herein include: gleevec, herceptin, avstin, PD-1 checkpoint inhibitors, PDL-1 checkpoint inhibitors, CTLA-4 checkpoint inhibitors, tamoxifen, trastuzamab, raloxifene, fluorouracil/5-fu, pamidronate disodium, anastrozole, exemestane, cyclophos-phamide, letrozole, toremifene, fulvestrant, fluoxymester-one, trastuzumab, methotrexate, megastrol acetate, docetaxel, paclitaxel, testolactone, aziridine, vinblastine, capecitabine, goselerin acetate, zoledronic acid, taxol, vinblastine, and/or vincristine.
  • the cancer can be resistant to certain chemotherapeutic agents.
  • Administration of the of the polymeric nanoparticles of the present disclosure can be an alternative therapy when a different therapy, vulnerable to resistance, has been attempted unsuccessfully.
  • the therapies of the instant disclosure can offer alternative forms or administration of chemotherapeutic drugs that can reduce the effect of resistance to the drugs.
  • composition or pharmaceutical composition comprising the polymeric nanoparticles can be administered to the subject via an administration route.
  • the composition or pharmaceutical composition can be administered intravenously, intratumorally, or subcutaneously.
  • the composition can be administered at least once per day, once every other day, once per week, twice per week, once per month, or twice per month. In an embodiment of the methods, the composition is administered at least once per day. In an embodiment of the methods, the composition is administered at least once every other day. In an embodiment of the methods, the composition is administered at least once per week. In an embodiment of the methods, the composition is administered at least twice per week. In an embodiment of the methods, the composition is administered at least once per month. In an embodiment of the methods, the composition is administered at least twice per month. In another embodiment, the composition is administered more than once per day.
  • the composition is administered over a period of three weeks. In other embodiments of the methods, the composition is administered over a period of 30 days. In other embodiments of the methods, the composition is administered over a period of 60 days. In other embodiments of the methods, the composition is administered over a period of 90 days. In other embodiments of the methods, the composition is administered over a period of 120 days. In other embodiments of the methods, the composition is administered over a period of 150 days. In other embodiments of the methods, the composition is administered over a period of 6 months. In other embodiments of the methods, the composition is administered over a period of about 6 months to about 1 year. In other embodiments of the methods, the composition is administered over a period of about 1 year to about 2 years.
  • the effective dosage of the polymeric nanoparticles provided herein may vary depending on the chemotherapeutic agent(s) used, the mode of administration, the condition being treated, and the severity of the condition being treated.
  • the dosage regimen of the polymeric nanoparticle can be selected in accordance with a variety of factors including the route of administration and the renal and hepatic function of the patient.
  • the therapeutically effective amount can be a human equivalent dose that is determined from an animal experiment.
  • Di-block and penta-block copolymers can serve as starting materials for the manufacture of polymeric nanoparticles of the present disclosure.
  • An exemplary method that was performed to prepare these starting is provided below.
  • the penta-block copolymer is synthesized from initiator PLURONIC® L-61.
  • PLURONIC® L-61 comprises polyoxypropylene with a molecular mass of 1800 g/mol and comprises a 10% polyoxyethylene content (poloxamer 181).
  • PLURONIC® L-61 is available from BASF SE, Ludwigshafen, Germany.
  • the di-block copolymer is synthesized from initiator mPEG5000. Both syntheses occur by ring-opening polymerization of lactide in the presence of a Sn-catalyst in tetraglyme as the solvent. Purification is performed through multiple rounds of precipitation to remove the catalyst, solvent, and lactic acid impurities to yield both polymers that are released according to the release criteria separately.
  • the polymers may be stored as starting materials for manufacturing polymeric nanoparticles.
  • Polymeric nanoparticles of the present disclosure associated with salinomycin and an anthracycline chemotherapeutic agent can be prepared as follows.
  • PLA-based block copolymers penta-block PLA-PEG-PPG-PEG-PLA and di-block PLA-PEG hybrid block copolymers
  • 100 mg PLURONIC® F127 was dissolved in 20 mL double distilled (dd) water, followed by addition of 5pL triethylamine to create a second mixture.
  • the inventors of the present disclosure have found that addition of the triethylamine (or other pH-raising compounds) can aid in association of the anthracycline chemotherapeutic compounds with the polymeric nanoparticles.
  • anthracycline chemotherapeutic compounds and salinomycin were dissolved in 100 pL DMSO to create a third mixture.
  • Mixtures of either salinomycin and doxorubicin in 1:3, 1:1, and 3:1 ratios or pirarubicin and salinomycin in 1:3, 1:1, and 1:3 ratios were used.
  • nanoparticles containing either only doxorubicin or only pirarubicin were also prepared.
  • the third mixture was added to the first mixture in order to create the various combinations of drugs and nanoparticle polymer ratios (and a fourth mixture).
  • the fourth mixture was added slowly to the second mixture using a 26-gauge needle and syringe. The resulting emulsion was allowed to stir, and the organic solvent was removed by evaporation.
  • the chemotherapeutic drug- loaded polymeric nanoparticles were filtered using AMICON® filters (ultracentrifuge filters available from MilliporeSigma, with a molecular weight cut off of 3,000 Da) at 4,000 rpm for 30 min to separate the unencapsulated drug from the nanoparticles.
  • the polymeric nanoparticles were redispersed in 10 mL dd water, with 50 mg glucose (dissolved in 5 mL dd water) added to the nanoparticles and gently mixed (total volume of 15mL). The polymeric nanoparticles were stored at -20°C until further use.
  • Tables 1 and 2 summarize various physical properties of the resulting nanoparticles prepared in this manner.
  • these NP formulations contained 40 mg of penta- and di -block copolymers, 40 mg of FI 27 polymer, and 20 mg of glucose.
  • Polymeric nanoparticles comprising doxorubicin alone, salinomycin alone, or both salinomycin and doxorubicin (at ratio of SAL:DOX of 1 :3) were suspended in phosphate buffered saline at a pH of 7.4 at 37°C, with shaking at 120 rpm. Release percentages for both drugs from each of the three polymeric nanoparticle formulations were measured over time at 1, 2, 3, 4, 5, 6, 7, 8, 9, and 12 days. Results are shown in FIG. 2 and Table 3.
  • FIG. 2 shows the cumulative percentage of the drugs released by the various nanoparticle compositions that were tested. Data from the NPs containing salinomycin and doxorubicin in a ratio of 1 :3 are shown individually for each of salinomycin and doxorubicin. Table 3 Drag Release Data
  • SUM149 cells are a triple negative breast cancer (TNBC) cell line.
  • TNBC triple negative breast cancer
  • Cells were grown in cell culture and treated with one of five different NP formulations (SAL-loaded NPs, DOX-loaded NPs, NPs loaded with a 3 : 1 SAL:DOX ratio, NPs loaded with a 1 : 1 SAL:DOX ratio, and NPs loaded with a 1 :3 SAL:DOX ratio).
  • Tables 4 and 5 and FIG. 3 show the results of the cell proliferation assay. Surprisingly, the NPs loaded with a 1:3 SAL:DOX ratio showed synergistic activity, with a particularly small IC 50 for SAL. Additionally, the combination index was particularly favorable for the 1 :3 SAL:DOX formulation. All SAL/DOX combination NP formulations showed increased and surprising performance, relative to the single drug NPs.
  • EAC tumor regression in a syngeneic mouse model An experiment was conducted to examine the ability of NPs to reduce the growth of tumors in mice. Specifically, syngeneic mice that develop Ehrlic Ascites Carcinoma (EAC) tumors were used. EAC is a spontaneous murine mammary adenocarcinoma mouse model in tumor biology. Mice were treated with NPs associated with either SAL (administered at 3 mg/kg), DOX (administered at 3 mg/kg), or a 1:3 ratio of SAL:DOX (administered with DOX at 3 mg/kg and SAL at 1 mg/kg). Details of administration and dosing are described in Table 6. Administration of NPs was accomplished intravenously. Doses were administered twice per week, for four weeks, with a dose administered on days 1, 4, 8, 11, 15, 18, 22, and 25.
  • FIG. 4 shows the results of the experiment. Tumor volume grew when mice were treated with a vehicle control and grew more slowly with treatment with either SAL NPs or DOX NPs. Surprisingly however, tumor growth was inhibited (or prevented) most effectively when mice were treated with NPs associated with a 1 :3 ratio of SAL:DOX.
  • FIG. 5 shows the percent of MDA-MB 231 cells that survived treatment with the drugs at various concentrations. Based on the data, free doxorubicin showed an IC50 of 0.242 mM and free pirarubicin showed an IC50 of 0.207 mM.
  • FIG. 6A shows the cumulative percentage of pirarubicin released by the nanoparticle composition. Specifically, cumulative release of pirarubicin from nanoparticles over 15 days was approximately 22% of total encapsulated pirarubicin concentration.
  • release was again measured from pirarubicin-containing NPs.
  • release was measured at both a pH of 7.4 and a pH of 5 at 0, 1, 2, 3, 6, 8, 11, 13, and 15 days. Both measurements were conducted at 37°C, with shaking at 150 rpm.
  • FIG. 6B shows that release of pirarubicin was markedly increased in the lower pH environment. Specifically, cumulative release of pirarubicin from nanoparticles over 15 days at pH 7.4 was approximately 22% of the total encapsulated pirarubicin concentration. Cumulative release of pirarubicin from nanoparticles over 15 days at pH 5 was approximately 89% of the total encapsulated pirarubicin concentration.
  • FIG. 7A and FIG. 7B show the results of the cell proliferation assay.
  • the NPs loaded with pirarubicin showed a lower IC 50 than the treatment with free pirarubicin.
  • the IC 50 was 3.245 mM for free pirarubicin and 0.2283 pM for the NPs loaded with pirarubicin.
  • the IC50 was 0.6007 mM for free pirarubicin and 0.1504 mM for the NPs loaded with pirarubicin.
  • FIG. 8A and Table 7 show the results of the cell proliferation assay in SUM149 cells. Unexpectedly, The NPs loaded with a 1:3 ratio of pirarubicin to salinomycin showed the lowest IC50 and combination index (Cl).
  • FIG. 8B and Table 8 show the results of the cell proliferation assay in MDA-MB 468 cells. Similar to the data seen in SUM149 cells, the NPs loaded with a 1:3 ratio of pirarubicin to salinomycin showed the lowest IC50 and combination index (Cl).
  • FIG. 9A and Table 9 show the results of the cell proliferation assay in SUM149 cells. Unexpectedly, The NPs loaded with a 1:3 ratio of pirarubicin to salinomycin showed the lowest IC50 and greatest reduction in IC50.
  • FIG. 9B and Table 10 show the results of the cell proliferation assay in MDA-MB 468 cells. Similar to the data seen in SUM149 cells, the NPs loaded with a 1 :3 ratio of pirarubicin to salinomycin showed the lowest IC50 and the greatest reduction in IC50.

Abstract

The present disclosure relates to a composition comprising: a) polymeric nanoparticles comprising hybrid block copolymers comprising a poly(lactic acid)-poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA-PEG-PPG-PEG-PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer; b) salinomycin; and c) an anthracycline chemotherapeutic compound. Both the salinomycin and the anthracycline chemotherapeutic compound are associated with the same polymeric nanoparticle. Methods related to the use of the polymeric nanoparticles are also disclosed.

Description

POLYMERIC NANOPARTICLES COMPRISING CHEMOTHERAPEUTIC COMPOUNDS AND RELATED METHODS
CROSS REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of U.S. Provisional Application No. 63/192,828, filed on May 25, 2021, the contents of which is incorporated herein by reference in its entirety.
FIELD
[0002] This disclosure relates to polymeric nanoparticles comprising an anthracycline chemotherapeutic compound and salinomycin and related methods of using the polymeric nanoparticles, treating of cancer, and making the polymeric nanoparticles.
BACKGROUND
[0003] Conventional chemotherapeutic agents used in the treatment of cancer can suffer from resistance of the cancer cells to the chemotherapeutic agents or from toxicity induced in healthy cells/tissues. Delivery of anticancer drugs would be more effective if the delivery system was able to effectuate treatment with smaller amounts of drugs and/or new combinations of drugs to mitigate resistance. There is a pressing need for such delivery systems.
SUMMARY
[0004] In one aspect, this disclosure provides a composition comprising polymeric nanoparticles comprising block copolymers comprising a poly(lactic acid)-poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA-PEG-PPG-PEG- PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di block copolymer; salinomycin; and an anthracycline chemotherapeutic compound. The salinomycin and the anthracycline chemotherapeutic compound are associated with the polymeric nanoparticles.
[0005] In some embodiments, the salinomycin and the anthracycline chemotherapeutic compound are both associated substantially with the same polymeric nanoparticles.
[0006] In some embodiments, the anthracycline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof. In some embodiments, the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin. In some embodiments, the anthracycline chemotherapeutic compound is doxorubicin. In some embodiments, the anthracycline chemotherapeutic compound is pirarubicin.
[0007] In some embodiments, the total mass of anthracycline chemotherapeutic compound is greater than or equal to the total mass of the salinomycin. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1.5:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 2: 1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 2.5:1 to 3.5:1.
[0008] In some embodiments, the total mass of anthracycline chemotherapeutic compound is less than or equal to the total mass of the salinomycin. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1:1.5. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1 :2. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 1:2.5 to 1:3.5.
[0009] In some embodiments, the total mass of anthracycline chemotherapeutic compound and is about equal to the total mass of the salinomycin.
[0010] In some embodiments, less PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than PLA-PEG di-block copolymer. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:20 to 1:1. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1 : 15 to 1 :2. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:8 to 1:10. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG- PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1 :3 to 1 :5. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:2 to 3:8.
[0011] In some embodiments, the average diameter of the polymeric nanoparticles is between 50 and 170 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 130 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 100 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 80 and 110 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 100 and 170 nm.
[0012] In some embodiments, a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.5. In some embodiments, a PDI of the polymeric nanoparticles is not more than 0.3.
[0013] In some embodiments, a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV.
[0014] In some embodiments, the composition further comprises a PEG-PPG-PEG triblock copolymer.
[0015] In some embodiments, a pharmaceutical composition comprises the composition. The pharmaceutical composition further comprises a pharmaceutically acceptable carrier. [0016] In another aspect, the present disclosure provides a method of reducing proliferation, survival, migration, or colony formation ability of a rapidly proliferating cell in a subject in need thereof, comprising contacting the cell with a composition comprising a therapeutically effective amount of the composition or the pharmaceutical composition. In some embodiments, the cell is a cancer cell. In some embodiments, the cell is a cancer stem cell.
[0017] In another aspect, the present disclosure provides a method for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a composition comprising the composition or the pharmaceutical composition. In some embodiments, the cancer comprises a solid tumor cancer or a cancer of the blood.
[0018] In some embodiments, the cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, leukemia, lymphoma, osteosarcoma, gastric cancer, prostate cancer, colon cancer, non-small cell lung cancer and small cell lung cancer, liver cancer, kidney cancer, head and neck cancer, cervical cancer, acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, and combinations thereof. In some embodiments, the cancer or breast cancer comprises triple negative breast cancer (TNBC). In some embodiments, the cancer is metastatic.
[0019] In some embodiments, the method further comprises administering an additional anti-cancer therapy to the subject. In some embodiments, the additional anti-cancer therapy comprises surgery, chemotherapy, radiation, hormone therapy, immunotherapy, or a combination thereof. In some embodiments, the cancer is resistant or refractory to a chemotherapeutic agent.
[0020] In some embodiments, the subject is a human.
[0021] In some embodiments, the composition or pharmaceutical composition is administered intravenously, intratumorally, or subcutaneously.
[0022] In another aspect, the present disclosure provides a method of manufacturing polymeric nanoparticles comprising: a) mixing penta-block PLA-PEG-PPG-PEG-PLA and di-block PLA-PEG hybrid block copolymers dissolved in acetonitrile with salinomycin and at least one anthracycline compound to form a first mixture; b) mixing the first mixture with a PEG-PPG-PEG triblock copolymer dissolved in water to form a second mixture; c) stirring the second mixture and evaporating the acetonitrile; and d) filtering the stirred and evaporated second mixture, thereby manufacturing the polymeric nanoparticles.
[0023] In some embodiments, the salinomycin and at least one anthracycline chemotherapeutic compound are dissolved in DMSO when used in step a). In some embodiments, the PEG-PPG-PEG triblock copolymer comprises poloxamer 407. In some embodiments, the method further comprises adding triethylamine during step b).
[0024] In some embodiments, the anthracycline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof. In some embodiments, the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin. In some embodiments, the anthracycline chemotherapeutic compound is doxorubicin. In some embodiments, the anthracycline chemotherapeutic compound is pirarubicin.
[0025] In some embodiments, the total mass of anthracycline chemotherapeutic compound is greater than or equal to the total mass of the salinomycin. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1.5:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 2:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 2.5:1 to 3.5:1.
[0026] In some embodiments, the total mass of anthracycline chemotherapeutic compound is less than or equal to the total mass of the salinomycin. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1:1.5. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1 :2. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 1:2.5 to 1:3.5. In some embodiments, the total mass of anthracycline chemotherapeutic compound is about equal to the total mass of the salinomycin.
[0027] In some embodiments, less PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than PLA-PEG di-block copolymer. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:20 to 1:1. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:15 to 1:2. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:8 to 1:10. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG- PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1 :3 to 1 :5. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:2 to 3:8.
[0028] In some embodiments, the average diameter of the polymeric nanoparticles is between 50 and 170 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 130 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 60 and 100 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 80 and 110 nm. In some embodiments, the average diameter of the polymeric nanoparticles is between 100 and 170 nm.
[0029] In some embodiments, a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.5. In some embodiments, a PDI of the polymeric nanoparticles is not more than 0.3.
[0030] In some embodiments, a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV.
BRIEF DESCRIPTION OF THE DRAWINGS [0031] Aspects, features, benefits, and advantages of the embodiments described herein will be apparent with regard to the following description, appended claims, and accompanying drawings. The drawings depict exemplary embodiments of the disclosure and are not intended to be limiting.
[0032] FIG. 1A shows a chemical structure of epirubicin (EPI).
[0033] FIG. IB shows a chemical structure of idarubicin (IDA). [0034] FIG. 1C shows a chemical structure of daunorubicin (DNR).
[0035] FIG. ID shows a chemical structure of doxorubicin (DOX).
[0036] FIG. IE shows a chemical structure of pirarubicin (PIRA).
[0037] FIG. IF shows a chemical structure of nemorubicin (NEMO).
[0038] FIG. 2 is a plot showing data from an in vitro release assay for NPs containing the indicated drugs.
[0039] FIG. 3 is a plot showing data from an in vitro cell proliferation assay for NPs containing the indicated drugs.
[0040] FIG. 4 is a plot showing tumor volume data over time from a tumor regression study that treated mice using NPs containing the indicated drugs.
[0041] FIG. 5 is a plot showing data from an in vitro cell proliferation assay for free pirarubicin and doxorubicin with no NPs included.
[0042] FIG. 6A is a plot showing data from an in vitro release assay for NPs containing pirarubicin.
[0043] FIG. 6B is a plot showing data from an in vitro release assay for NPs containing pirarubicin at a pH of 7.4 and a pH of 5.
[0044] FIG. 7A is a plot showing data from an in vitro cell proliferation assay performed with SUM149 cells. Either NPs containing pirarubicin or free pirarubicin were used.
[0045] FIG. 7B is a plot showing data from an in vitro cell proliferation assay performed with MDA-MB 468 cells. Either NPs containing pirarubicin or free pirarubicin were used. [0046] FIG. 8A is a plot showing data from an in vitro cell proliferation assay using SEIM149 cells and NPs containing the indicated drugs.
[0047] FIG. 8B is a plot showing data from an in vitro cell proliferation assay using MDA-MB 468 cells and NPs containing the indicated drugs.
[0048] FIG. 9A is a plot showing data from an in vitro cell proliferation assay using SEIM149 cells and NPs containing the indicated drugs.
[0049] FIG. 9B is a plot showing data from an in vitro cell proliferation assay using MDA-MB 468 cells and NPs containing the indicated drugs.
DETAILED DESCRIPTION
[0050] It will be appreciated that for clarity, the following disclosure will describe various aspects of embodiments. It should be noted that the specific embodiments are not intended as an exhaustive description or as a limitation to the broader aspects discussed herein. One aspect described in conjunction with a particular embodiment is not necessarily limited to that embodiment and can be practiced with any other embodiment(s). Reference throughout this specification to “one embodiment,” “an embodiment,” “an example embodiment,” “some embodiments,” or the like means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present disclosure. Thus, appearances of the phrases “in one embodiment,” “in an embodiment,” “an example embodiment,” or “some embodiments” in various places throughout this specification are not necessarily all referring to the same embodiment, but may. Furthermore, the particular features, structures or characteristics may be combined in any suitable manner, as would be apparent to a person skilled in the art from this disclosure, in one or more embodiments. Furthermore, while some embodiments described herein include some but not other features included in other embodiments, combinations of features of different embodiments are meant to be within the scope of the disclosure. For example, in the appended claims, any of the claimed embodiments can be used in any combination.
Definitions
[0051] Listed below are definitions of various terms used herein. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.
[0052] Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Generally, the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, and organic and polymer chemistry, are those well-known and commonly employed in the art.
[0053] As used herein, the articles “a” and “an” refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element. Furthermore, use of the term “including” as well as other forms, such as “include,” “includes,” and “included,” is not limiting.
[0054] As used herein, the term “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term “about” is meant to encompass variations of ±5% from the specified value, as such variations are appropriate to perform the disclosed methods. [0055] As used in the specification and in the claims, the term “comprising” may include the embodiments “consisting of’ and “consisting essentially of.” The terms “comprise(s),” “include(s),” “having,” “has,” “may,” “contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that require the presence of the named ingredients/steps and permit the presence of other ingredients/steps. However, such description should be construed as also describing compositions or processes as “consisting of’ and “consisting essentially of’ the enumerated compounds, which allows the presence of only the named compounds, along with any pharmaceutically acceptable carriers, and excludes other compounds.
[0056] All ranges disclosed herein are inclusive of the recited endpoint and independently combinable (for example, the range of “from 50 mg to 500 mg” is inclusive of the endpoints, 50 mg and 500 mg, and all the intermediate values). The endpoints of the ranges and any values disclosed herein are not limited to the precise range or value; they are sufficiently imprecise to include values approximating these ranges and/or values.
[0057] As used herein, the term "nanoparticle" refers to particles in the range between 10 nm to 1000 nm in diameter, wherein diameter refers to the diameter of a perfect sphere having the same volume as the particle. The term "nanoparticle" is used interchangeably with "nanoparti cle(s)." In some cases, a population of particles may be present. As used herein, the “diameter” of the nanoparticles is an average of a distribution in a particular population. [0058] As used herein, the term "polymer" is given its ordinary meaning as used in the art, i.e., a molecular structure comprising one or more repeat units (monomers), connected by covalent bonds. The repeat units may all be identical, or in some cases, there may be more than one type of repeat unit present within the polymer.
[0059] A "chemotherapeutic agent," "therapeutic agent," and/or "drug" is a biological (large molecule) or chemical (small molecule) compound useful in the treatment of cancer, regardless of mechanism of action. Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, proteins, antibodies, photosensitizers, and kinase inhibitors.
[0060] The term "combination," "therapeutic combination," or "pharmaceutical combination" as used herein refer to the combined administration of two or more therapeutic agents (e g., co-delivery). Components of a combination therapy may be administered simultaneously or sequentially, i.e., at least one component of the combination is administered at a time temporally distinct from the other component(s). In embodiments, a component(s) is administered within one month, one week, 1-6 days, 18, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2 , 1 hour, or 30, 20, 15, 10, or 5 minutes of the other component(s).
[0061] The term "pharmaceutically acceptable" as used herein refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues a warm-blooded animal, e.g., a mammal or human, without excessive toxicity, irritation allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
[0062] A "therapeutically effective amount" of a polymeric nanoparticle comprising one or more therapeutic agents is an amount sufficient to provide an observable or clinically significant improvement over the baseline clinically observable signs and symptoms of the disorders treated with the combination.
[0063] The term "subject" or "patient" as used herein is intended to include animals, which are capable of suffering from or afflicted with a cancer or any disorder involving, directly or indirectly, a cancer. Examples of subjects include mammals, e.g., humans, apes, monkeys, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non human animals. In an embodiment, the subject is a human, e.g., a human suffering from cancer.
[0064] The term "treating" or 'treatment" as used herein comprises a treatment relieving, reducing or alleviating at least one symptom in a subject or producing a delay in the progression of a disease. For example, treatment can be the diminishment of one or several symptoms of a disorder or complete eradication of a disorder, such as cancer. Within the meaning of the present disclosure, the term "treat" also denotes to arrest and/or reduce the risk of worsening a disease. The term "prevent", "preventing" or "prevention" as used herein comprises the prevention of at least one symptom associated with or caused by the state, disease or disorder being prevented.
[0065] As used herein, the term "human equivalent dose" refers to a dose of a composition to be administered to a human that is calculated from a specific dose used in an animal study.
[0066] As used herein, the term "rapidly proliferating cells" refers to cells having the capacity for autonomous growth (e.g., cancer cells).
[0067] As used herein, the term "cancer stem cell" refers to a cancer cell that has characteristics of a stem cell, such as giving rise to all cell types within a particular tumor type and the ability to self-renew. In some embodiments, the cancer stem cell is resistant or refractory to chemotherapy.
[0068] As used herein, the term “associated substantially with” in the context of a nanoparticle means a substance is encapsulated or stably interacting with a nanoparticle. For example, when a combination of substances are associated substantially with a nanoparticle, in some embodiments, the nanoparticle is loaded with both substances, as opposed to an embodiment where one substance is loaded into a first set of nanoparticles and a second substance is loaded into a second set of nanoparticles. In some embodiments, when a substance is associated substantially with a nanoparticle, at least 80%, at least 90%, at least 95%, or at least 99% of the mass of the substance is encapsulated or stably interacting with the nanoparticle.
Polymeric nanoparticles
[0069] Nanoparticles (also referred to herein as “NPs”) can be produced as nanocapsules or nanospheres. Drug loading in the nanoparticle can be performed by either an adsorption process or an encapsulation process (Spada et al., 2011; Protein delivery of polymeric nanoparticles; World Academy of Science, Engineering and Technology: 76, incorporated herein, by reference, in its entirety). Nanoparticles, by using both passive and active targeting strategies, can enhance the intracellular concentration of drugs in cancer cells while avoiding toxicity in normal cells. When nanoparticles bind to specific receptors and enter the cell, they are usually enveloped by endosomes via receptor-mediated endocytosis, thereby bypassing the recognition of P-glycoprotein, one of the main drug resistance mechanisms (Cho et al., 2008, Therapeutic Nanoparticles for Drug Delivery in Cancer, Clin. Cancer Res., 2008, 14: 1310-1316, incorporated herein, by reference, in its entirely).
[0070] Nanoparticles are removed from the body by opsonization and phagocytosis (Sosnik et al., 2008; Polymeric Nanocarriers: New Endeavors for the Optimization of the Technological Aspects of Drugs; Recent Patents on Biomedical Engineering, 1: 43-59, incorporated herein, by reference, in its entirety). Nanocarrier based systems can be used for effective drug delivery with the advantages of improved intracellular penetration, localized delivery, protection of drugs against premature degradation, controlled pharmacokinetic and drug tissue distribution profile, lower dose requirement, and cost effectiveness (Farokhzad OC, et al.; Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc. Natl. Acad. Sci. USA 2006,103 (16): 6315-20; Fonseca C, et al., Paclitaxel-loaded PLGA nanoparticles: preparation, physicochemical characterization and in vitro anti -turn oral activity. J. Controlled Release 2002; 83 (2): 273-86; Hood et al., Nanomedicine, 2011, 6(7): 1257-1272, incorporated herein, by reference, in their entireties).
[0071] The uptake of nanoparticles is indirectly proportional to their small dimensions. Due to their small size, the polymeric nanoparticles have been found to evade recognition and uptake by the reticulo-endothelial system (RES), and can thus circulate in the blood for an extended period (Borchard et al., 1996, Pharm. Res. 7: 1055-1058, incorporated herein, by reference, in its entirety). Nanoparticles are also able to extravasate at the pathological site like the leaky vasculature of a solid tumor, providing a passive targeting mechanism. Due to the higher surface area leading to faster solubilization rates, nano-sized structures usually show higher plasma concentrations and area under the curve (AUC) values. Lower particle size helps in evading the host defense mechanism and increase the blood circulation time. Nanoparticle size affects drug release. Larger particles have slower diffusion of drugs into the system. Smaller particles offer larger surface area but lead to last drug release. Smaller particles tend to aggregate during storage and transportation of nanoparticle dispersions. Hence, a compromise between a small size and maximum stability of nanoparticles is desired. The size of nanoparticles used in a drug delivery system should be large enough to prevent their rapid leakage into blood capillaries but small enough to escape capture by fixed macrophages that are lodged in the reticuloendothelial system, such as the liver and spleen. [0072] In addition to their size, the surface characteristics of nanoparticles are also an important factor in determining the life span during circulation. Nanoparticles should ideally have a hydrophilic surface to escape macrophage capture. Nanoparticles formed from block copolymers with hydrophilic and hydrophobic domains meet these criteria. Controlled polymer degradation also allows for increased levels of agent delivery to a diseased state. Polymer degradation can also be affected by the particle size. Degradation rates increase with increase in particle size in vitro (Biopolymeric nanoparticles; Sundar et al., 2010, Science and Technology of Advanced Materials; doi: 10.1088/1468-6996/11/1/014104, incorporated herein, by reference, in its entirety).
[0073] Poly(lactic acid) (PL A) has been approved by the US FDA for applications in tissue engineering, medical materials and drug carriers. US2006/0165987A1, incorporated herein, by reference, in its entirety, describes a stealthy polymeric biodegradable nanosphere comprising poly(ester)-poly(ethylene) multiblock copolymers and optional components for imparting rigidity to the nanospheres and incorporating pharmaceutical compounds. US2008/0081075A1, incorporated herein, by reference, in its entirety, discloses a novel mixed micelle structure with a functional inner core and hydrophilic outer shells, self- assembled from a graft macromolecule and one or more block copolymer.
[0074] US2010/0004398A1, incorporated herein, by reference, in its entirety, describes a polymeric nanoparticle of shell/core configuration with an interphase region and a process for producing the same.
[0075] Provided herein are polymeric nanoparticles for the delivery of chemotherapeutic compounds. The inventors of the present disclosure have developed polymeric nanoparticles comprising formulations of chemotherapeutic compounds. The polymeric nanoparticles are useful for the delivery of drugs. For example, the nanoparticles can find use in treatment of diseases exhibiting rapid cell division such as various cancers by delivering appropriate chemotherapeutic agents.
[0076] Accordingly, provided herein is a composition comprising: a) polymeric nanoparticles comprising hybrid block copolymers comprising a poly(lactic acid)- poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA- PEG-PPG-PEG-PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer; b) salinomycin; and c) an anthracycline chemotherapeutic compound. Both the salinomycin and the anthracycline chemotherapeutic compound are associated with the polymeric nanoparticles.
[0077] For example, the salinomycin and the anthracycline chemotherapeutic compound can be associated with the polymeric nanoparticles by being contained within an enclosed region of a shell of polymer. Alternatively or additionally, the drugs can be interspersed within the polymer that forms the shell, or the drugs can adhere to an outside surface of the shell. The drugs can be associated with the polymeric nanoparticle in any manner suitable to carry and deliver the drugs to locations of disease in need of treatment.
[0078] In certain embodiments, the salinomycin and the anthracycline chemotherapeutic compound can both be associated substantially with the same polymeric nanoparticles. In some embodiments, the salinomycin and the anthracycline chemotherapeutic compound are encapsulated by the nanoparticle.
[0079] In certain embodiments, the polymeric nanoparticles can comprise both hydrophobic and hydrophilic block copolymers. In an embodiment, the polymeric nanoparticles provided herein comprise hybrid block copolymers comprising a poly(lactic acid)-poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA-PEG-PPG-PEG-PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer. The PLA-PEG-PPG-PEG-PLA penta-block copolymer can be formed from PEG-PPG-PEG triblock copolymer and PLA via ring opening polymerization of the lactide. In certain embodiments, the molecular weight of the penta- block copolymer can range from 5,000 g/mol to 40,000 g/mol. In certain embodiments, the molecular weight range of di-block copolymer can be from 2,000 g/mol to 15,000 g/mol. [0080] Poly(lactic acid) (PLA), is a hydrophobic polymer and can be a component of the polymeric nanoparticles. As alternative to PLA or in addition to PLA, poly(glycolic acid) (PGA) and block copolymer of poly lactic acid-co-glycolic acid (PLGA) may also be used. The hydrophobic polymer can also comprise a biologically derived polymer or a biopolymer. The molecular weight of the PLA used is generally in the range of about 2,000 g/mol to 80,000 g/mol. Thus, in an embodiment, the PLA used is in the range of about 10,000 g/mol to 80,000 g/mol. The average molecular weight of PLA may also be about 70,000 g/mol. As used herein, one g/mol is equivalent to one “dalton” (i.e., dalton and g/mol are interchangeable when referring to the molecular weight of a polymer). “Kilodalton” (or “kDa”) as used herein refers to 1,000 dal tons.
[0081] Polyethylene glycol) (PEG) is another preferred component of the polymer used to form the polymeric nanoparticles. PEG can impart hydrophilicity, reduce phagocytosis by macrophages, and/or reduce immunological recognition. Block copolymers like polyethylene glycol)-poly(propylene glycol)-poly(ethylene glycol) (PEG-PPG-PEG) are hydrophilic or hydrophilic-hydrophobic copolymers that can be components of the polymeric nanoparticles of the present disclosure. For example, the PLA-PEG-PPG-PEG-PLA penta-block copolymer can be formed from ring opening polymerization using lactide and also by using mPEG for the di-block. In general, block copolymers of the present disclosure may have two, three, four, five, or more distinct blocks.
[0082] In a further embodiment, the polymeric nanoparticles provided herein comprise a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer.
[0083] In some embodiments, a first block copolymer of the instant disclosure consists essentially of two segments of poly(lactic acid) (PLA), separated by a segment of poly(ethylene glycol)-polypropylene glycol)-poly(ethylene glycol) (PEG-PPG-PEG), to form the PLA-PEG-PPG-PEG-PLA penta-block copolymer.
[0084] In some embodiments, a second block copolymer of the instant disclosure consists essentially of a PLA-PEG di-block copolymer. [0085] In some embodiments, the first and second block copolymers of the instant disclosure can be combined to form the polymeric nanoparticles of the instant disclosure. In some embodiments, the process described in Example 1 of the present disclosure can be used to accomplish the combination.
[0086] In some embodiments, the polymeric nanoparticles of the instant disclosure can be biodegradable.
[0087] In various embodiments, the nanoparticles comprise QUATRAMER™ reagent, which comprises a PLA-PEG-PPG-PEG-PLA penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer. QUATRAMER™ reagent is available from Hillstream Biopharma; Bridgewater, NJ, USA.
[0088] The PLA-PEG-PPG-PEG-PLA penta-block copolymer and the PLA-PEG di block copolymer may optionally be combined in specific ratios. As used herein, such ratios are expressed in the form of Masspenta-biock:Massdi-biock, unless stated otherwise. In some embodiments, less PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than the PLA-PEG di -block copolymer. In some embodiments, a mass ratio of PLA-PEG- PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be from 1 :20 to 1:1. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer can be from 1:15 to 1:2. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer can be from 1 : 10 to 1:2. In some embodiments, a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer can be from 1 :8 to 1:10 or can be about 1 :9. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta- block copolymer to PLA-PEG di-block copolymer can be from 1 :3 to 1 :5 or can be about 1 :4. In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be from 1 :2 to 3 :8 or can be about 3:7.
[0089] In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be at least 1 :20 such as, for example, at least 1 : 19, at least 1 : 18, at least 1 : 17, at least 1 : 16, at least 1 : 15, at least 1 : 14, at least 1 : 13, at least 1 : 12, at least 1 : 11, at least 1 : 10, at least 1 :9, at least 1 :8, at least 1 :7, at least 1 :6, at least 1:5, at least 1 :4, at least 1 :3, or at least 1 :2.
[0090] In some embodiments, a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer can be not more than 1 : 1 such as, for example, not more than 1 :2, not more than 1:3, not more than 1 :4, not more than 1:5, not more than 1 :6, not more than 1 :7, not more than 1 :8, not more than 1 :9, not more than 1:10, not more than 1:11, not more than 1:12, not more than 1:13, not more than 1:14, not more than 1:15, not more than 1:16, not more than 1:17, not more than 1:18, or not more than 1:19.
[0091] The polymeric nanoparticles of the instant disclosure have, in various embodiments, a diameter that is an average of a distribution of nanoparticles in a particular population. The polymeric nanoparticles have dimensions that can be measured using a transmission electron microscope, or another suitable technique that can allow for measurements of the diameters of a sample of a population of polymeric nanoparticles.
[0092] In some embodiments, the diameter of the nanoparticles can be at least 50 nm such as, for example, at least 60 nm, at least 70 nm, at least 80 nm, at least 90 nm, at least 100 nm, at least 110 nm, at least 120 nm, at least 130 nm, at least 140 nm, at least 150 nm, or at least 160 nm.
[0093] In some embodiments, the diameter of the nanoparticles can be not more than 170 nm such as, for example, not more than 160 nm, not more than 150 nm, not more than 140 nm, not more than 130 nm, not more than 120 nm, not more than 110 nm, not more than 100 nm, not more than 90 nm, not more than 80 nm, not more than 70 nm, or not more than 60 nm.
[0094] In some embodiments, the diameter of the nanoparticles can range from 50 nm to 170 nm such as, for example, from 60 nm to 130 nm, from 60 nm to 100 nm, from 80 nm to 110 nm, from 90 to 130 nm, from 100 to 170 nm, or any other suitable range, based on the properties of the polymeric nanoparticles (e.g., the precise drugs associated therewith).
[0095] In some embodiments, a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.50 such as, for example, not more than 0.45, not more than 0.40, not more than 0.35, not more than 0.30, not more than 0.25, not more than 0.20, not more than 0.15, not more than 0.10, or not more than 0.05. In some embodiments, the PDI is from 0.05 to 0.2. As used herein, the PDI is a ratio of the mass average molar mass of the penta- and di-block copolymers in the polymeric nanoparticles to the number average molar mass of the penta- and di-block copolymers in the polymeric nanoparticles. PDI may also be referred to simply as, “dispersity.”
[0096] Number average molar mass is defined as below:
Figure imgf000016_0001
where Ni is the number of molecules of molecular mass Mu [0097] Mass average molar mass is defined as below:
Figure imgf000017_0001
where Ni is the number of molecules of molecular mass Mu
[0098] In some embodiments, mass average molar mass and number average molar mass can be measured by any suitable process such as, for example, gel permeation chromatography, viscometry via the Mark-Houwink equation, or colligative methods (for number average molar mass); or static light scattering, small angle neutron scattering, X-ray scattering, or sedimentation velocity (for number average molar mass).
[0099] In some embodiments, a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV such as, for example, -5 mV to -30 mV, -5 to -25 mV, or -5 to -15 mV. As used herein, zeta potential is a measure of the electrical potential difference at the slipping plane. The slipping plane is the interface of mobile fluid around a particle (e.g., a polymeric nanoparticle of the present disclosure) with fluid components that remain attached to the particle surface (e.g., via adsorption and/or electrostatic interaction).
[0100] The zeta potential and PDI (Polydispersity Index) of the nanoparticles may be calculated (see U.S. patent number 9,149,426, incorporated herein by reference, in its entirety).
[0101] In addition to the polymer components described herein, the compositions provided herein can comprise one or more chemotherapeutic compounds. The polymeric nanoparticles can associate with the chemotherapeutic compounds. For example, in some embodiments, the polymeric nanoparticles can encapsulate the chemotherapeutic compounds and/or adsorb to the chemotherapeutic compounds. The polymeric nanoparticles can associate with the chemotherapeutic compounds in any manner suitable to carry the chemotherapeutic compounds throughout a subject’s body and deliver the chemotherapeutic compounds to a diseased cell (e.g., a rapidly dividing cell such as a cancer cell).
[0102] In some embodiments, the chemotherapeutic compounds comprise salinomycin and an anthracycline chemotherapeutic compound. The anthracycline chemotherapeutic compound can comprise doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin, or a combination thereof. Suitable examples of anthracycline chemotherapeutic compounds are shown with their structures in FIG. 1 A-FIG. IF. The abbreviations refer to: epirubicin (EPI, FIG. 1A), idarubicin (IDA, FIG. IB), daunorubicin (DNR, FIG. 1C), doxorubicin (DOX, FIG. ID), pirarubicin (PIRA, FIG. IE), and nemorubicin (NEMO, FIG. IF).
[0103] The inventors of the present disclosure have determined that when the salinomycin and the anthracycline chemotherapeutic compound are both included in a composition of the present disclosure ( e.g ., associated with polymeric nanoparticles of the present disclosure), improved performance (e.g., improved growth inhibition of diseased cells) can be obtained, compared to the activity of the individual drugs.
[0104] Additionally, it has been determined that the ratio of the anthracycline chemotherapeutic compound to the salinomycin can impact the performance of the chemotherapeutic compounds. Unless stated otherwise, all ratios disclosed herein for the anthracycline chemotherapeutic compound compared to the salinomycin are written with the number referring to the relative amount anthracycline chemotherapeutic compound first and the number referring to the relative amount of salinomycin second, e.g., Massanthracycime chemotherapeutic compoundMasssaiinomycin. However, it is noted that the opposite order is used occasionally and explicitly in the Examples. It is understood that, for example, a 1 :3 ratio of anthracycline chemotherapeutic compound to salinomycin is equivalent to, and inherently discloses, a 3:1 ratio of salinomycin to anthracycline chemotherapeutic compound. Additionally, unless stated otherwise, the ratios herein are based on measurements of the masses of the respective chemical species.
[0105] In some embodiments, the total mass of anthracycline chemotherapeutic compound can be greater than or equal to the total mass of the salinomycin. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of at least 1.1:1, at least 1.2:1, at least 1.3:1, at least 1.4:1, at least 1.5:1, at least 2:1, at least 2.5:1, or at least 3:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of 2.5:1 to 3.5:1, or a mass ratio of 2:1 to 4:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of about 3:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of not more than 4:1, not more than 5:1, or not more than 6:1. [0106] In some embodiments, the total mass of anthracycline chemotherapeutic compound can be less than or equal to the total mass of the salinomycin. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of not more than 1:1.5, not more than 1:1.6, not more than 1:1.7, not more than 1:1.8, not more than 1:1.9, not more than 1 :2, not more than 1 :2.5, or not more than 1:3. In some embodiments, the anthracy cline chemotherapeutic compound and the salinomycin can be present in a mass ratio of 1 :2.5 to 1 :3.5, or a mass ratio of 1 :2 to 1 :4. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of about 1:3. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of at least 1:4, at least 1:5, or at least 1:6.
[0107] In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of about 1:1. In some embodiments, the anthracycline chemotherapeutic compound and the salinomycin can be present in a mass ratio of 1:1.
[0108] As described below, preparation of polymeric nanoparticles can optionally involve addition of a PEG-PPG-PEG triblock copolymer. The inventors of the present disclosure have found that including such a triblock copolymer can improve the stability of the polymeric nanoparticles and/or can serve as an emulsifier for other components. In some embodiments, the triblock copolymer may be associated with or associated substantially with the polymeric nanoparticles. In some embodiments, the triblock copolymer may not be associated with the polymeric nanoparticles. In some embodiments, the triblock copolymer comprises a central polypropylene glycol) block flanked by two poly(ethylene glycol) blocks. In some embodiments, the triblock copolymer comprises poloxamer 407.
Methods for Preparing Polymeric Nanoparticles [0109] The present disclosure provides a method of manufacturing nanoparticles comprising: a) mixing penta-block PLA-PEG-PPG-PEG-PLA and di-block PLA-PEG hybrid block copolymers dissolved in acetonitrile with salinomycin and at least one anthracycline compound to form a first mixture; b) mixing the first mixture with a PEG-PPG-PEG tri-block copolymer dissolved in water to form a second mixture; c) stirring the second mixture and evaporating the acetonitrile; and d) filtering the stirred and evaporated second mixture, thereby manufacturing the nanoparticles.
[0110] In some embodiments, the salinomycin and at least one anthracycline chemotherapeutic compound are dissolved in DMSO when used in step a). In some embodiments, the DMSO may be evaporated during step c). In some embodiments, the PEG- PPG-PEG triblock copolymer comprises poloxamer 407. In some embodiments, the method further comprises adding triethylamine during step b).
[0111] In some embodiments, the penta-block and di -block copolymers are present in the acetonitrile at 10 mg to 40 mg per mL of acetonitrile. In some embodiments, the penta-block and di-block copolymers are present in the acetonitrile at 15 mg to 30 mg per mL of acetonitrile. In some embodiments, the penta-block and di-block copolymers are present in the acetonitrile at about 20 mg per mL of acetonitrile.
[0112] In some embodiments, the at least one anthracycline compound and salinomycin are dissolved in DMSO such that the total mass of drugs is dissolved at 0.05 mg per pL of DMSO to 0.2 mg per pL of DMSO. In some embodiments, the at least one anthracycline compound and salinomycin are dissolved in DMSO such that the total mass of drugs is dissolved at about 0.1 mg per pL of DMSO.
[0113] In some embodiments, the PEG-PPG-PEG tri -block copolymer is dissolved in the water at 2.5 mg of PEG-PPG-PEG tri -block copolymer per mL of water to 10 mg of PEG- PPG-PEG tri-block copolymer per mL of water. In some embodiments, the PEG-PPG-PEG tri-block copolymer is dissolved in the water at 3.5 mg of PEG-PPG-PEG tri-block copolymer per mL of water to 7.5 mg of PEG-PPG-PEG tri -block copolymer per mL of water. In some embodiments, the PEG-PPG-PEG tri-block copolymer is dissolved in the water at about 5 mg per mL of water.
[0114] In some embodiments, the PEG-PPG-PEG tri -block copolymer comprises poloxamer 407. In some embodiments, the PEG-PPG-PEG tri-block copolymer comprises PLURONIC® F127. PLURONIC® F127 and poloxamer 407 both comprise a triblock copolymer comprising a central polypropylene glycol) block flanked by two poly(ethylene glycol) blocks. The approximate lengths of the two PEG blocks can be 101 repeat units, while the approximate length of the propylene glycol block can be 56 repeat units. PLURONIC® F127 is available from BASF SE, Ludwigshafen, Germany.
[0115] It is understood that both PLURONIC® F127 and poloxamer 407 comprise the same tri-block copolymer, but they may vary from each other based on their respective molecular weights and/or the number of monomers in each of their blocks. In some embodiments, the PLURONIC® F 127 and/or poloxamer 407 can comprise a molecular weight of from 10,500 g/mol to 14,500 g/mol such as, for example a molecular weight of about 12,600 g/mol. [0116] In some embodiments, the triethylamine is added in an amount of 0.5 pL to 2 pL for every 4 mL of water. In some embodiments, the triethylamine is added in an amount of about 0.5 pL for every 4 mL of water. In some embodiments, the triethylamine is added in an amount of about 1 pL for every 4 mL of water. In some embodiments, the triethylamine is added in an amount of about 2 mL for every 4 mL of water.
[0117] In some embodiments, the anthracy cline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof. In some embodiments, the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin. In some embodiments, the anthracycline chemotherapeutic compound is doxorubicin. In some embodiments, the anthracycline chemotherapeutic compound is pirarubicin.
[0118] Before the preparation of polymeric nanoparticles as described above, the penta- block and di-block copolymers may be prepared and used as reagents for preparation of the polymeric nanoparticles. These can be prepared as described in Example 1 from a poloxamer copolymer such as poloxamer 181 (for the penta-block copolymer), methoxypoly(ethylene glycol) (mPEG) (for the di-block copolymer), initiator, and PLA. A ring-opening polymerization of lactide in the presence of a Sn-catalyst can be employed, or any other suitable technique as determined by the skilled artisan. In some embodiments, the poloxamer 181 can comprise a molecular weight of 1,000 g/mol to 3,000 g/mol. In some embodiments, the poloxamer 181 can comprise a molecular weight of about 2,000 g/mol. The appropriate molecular weight can be selected in order to, for example, improve the properties ( e.g ., stability) of the polymeric nanoparticles.
Pharmaceutical Compositions
[0119] Also provided herein is a pharmaceutical composition comprising the polymeric nanoparticle compositions described herein for use in medicine and in other fields that use a carrier system or a reservoir or depot of nanoparticles. The polymeric nanoparticles can be used in prognostic, therapeutic, diagnostic and/or theranostic compositions. Suitably, the nanoparticles of the present disclosure are used for drug and agent delivery (e.g., within a tumor cell), as well as for disease diagnosis and medical imaging in human and animals.
Thus, the instant disclosure provides a method for the treatment of disease using the nanoparticles, further comprising a chemotherapeutic agent, as described herein. The nanoparticles of the present disclosure can also be use in other applications such as chemical or biological reactions where a reservoir or depot is required, as biosensors, as agents for immobilized enzymes and the like.
[0120] Thus, in an aspect, provided herein is a pharmaceutical composition comprising a) polymeric nanoparticles comprising hybrid block copolymers comprising a PLA-PEG-PPG- PEG-PLA penta-block copolymer with a PLA-PEG di-block copolymer; b) salinomycin; and c) an anthracycline chemotherapeutic compound. Both the salinomycin and the anthracycline chemotherapeutic compound are associated with the polymeric nanoparticles.
[0121] Suitable pharmaceutical compositions or formulations can contain, for example, from about 0.1% to about 99.9%, preferably from about 1% to about 60%, of the active ingredient(s). Pharmaceutical formulations for enteral or parenteral administration are, for example, those in unit dosage forms, such as sugar-coated tablets, tablets, capsules or suppositories, or ampoules. If not indicated otherwise, these are prepared in a manner known per se, for example by means of conventional mixing, granulating, sugar-coating, dissolving or lyophilizing processes. It will be appreciated that the unit content of a combination partner contained in an individual dose of each dosage form need not in itself constitute an effective amount since the necessary effective amount may be reached by administration of a plurality of dosage units.
[0122] The pharmaceutical compositions can contain, as the active ingredient, one or more of nanoparticles in combination with one or more pharmaceutically acceptable carriers (excipients). In making the compositions of the disclosure, the active ingredient is typically mixed with an excipient, diluted by an excipient, or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
[0123] Some examples of suitable excipients include lactose (e.g. lactose monohydrate), dextrose, sucrose, sorbitol, mannitol, starches (e.g. sodium starch glycolate), gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, colloidal silicon dioxide, microcrystalline cellulose, polyvinylpyrrolidone (e.g. povidone), cellulose, water, syrup, methyl cellulose, and hydroxypropyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy- benzoates; sweetening agents; and flavoring agents.
[0124] The liquid forms in which the compounds and compositions of the present disclosure can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
Methods comprising use of polymeric nanoparticles [0125] The polymeric nanoparticles and pharmaceutical compositions disclosed herein can be used to treat or prevent any condition or disorder which is known to or suspected of benefitting from treatment with salinomycin and anthracy cline chemotherapeutic compounds. [0126] In one aspect, the polymeric nanoparticles and/or pharmaceutical compositions disclosed herein can be used to reduce proliferation, survival, migration, or colony formation ability of a rapidly proliferating cell in a subject in need thereof. This can be accomplished by contacting the cell with a therapeutically effective amount of the polymeric nanoparticles and/or pharmaceutical compositions. Such a method can be conducted in vivo (e.g, in a cancer patient), in vitro , or ex vivo.
[0127] In some embodiments, the cell can be a cancer cell or a cancer stem cell.
[0128] In another aspect, the polymeric nanoparticles and/or pharmaceutical compositions disclosed herein can be used to treat or prevent cancer or a precancerous condition. In some embodiments, the cancer can be, a cancer cell or a cancer stem cell. In some embodiments, the cancer can be a solid tumor cancer or a cancer of the blood. In some embodiments, the cancer can be selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, leukemia, lymphoma, osteosarcoma, gastric cancer, prostate cancer, colon cancer, non-small cell lung cancer and small cell lung cancer, liver cancer, kidney cancer, head and neck cancer, cervical cancer, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, multiple myeloma, and combinations thereof.
[0129] In some embodiments, the cancer can comprise triple negative breast cancer (TNBC).
[0130] In some embodiments, the cancer can be metastatic cancer. [0131] In some embodiments, the cancer may be an affliction of a subject. In some embodiments, the subject may be a human.
[0132] In some embodiments, the treatment using the polymeric nanoparticles or pharmaceutical composition comprising them can comprise administration of an additional anti-cancer therapy. The additional anti-cancer therapy can comprise any medically suitable therapy that could be combined with the polymeric nanoparticles disclosed herein. Such combinations of therapies can increase the overall effectiveness of cancer treatments.
[0133] In some embodiments, the additional anti-cancer therapy can comprise surgery; chemotherapy, radiation, hormone therapy, immunotherapy, or a combination thereof.
[0134] Additional anti-cancer therapies that may be combined with the polymeric- nanoparticle-based therapies disclosed herein include: lenalidomide, crizotinib or a histone deacetylase (HD AC) inhibitor , such as those disclosed in US Patent No. 8,883,842, incorporated by reference herein, in its entirety. Additional anti-cancer therapies that may be combined with the polymeric-nanoparticle-based therapies disclosed herein include: gleevec, herceptin, avstin, PD-1 checkpoint inhibitors, PDL-1 checkpoint inhibitors, CTLA-4 checkpoint inhibitors, tamoxifen, trastuzamab, raloxifene, fluorouracil/5-fu, pamidronate disodium, anastrozole, exemestane, cyclophos-phamide, letrozole, toremifene, fulvestrant, fluoxymester-one, trastuzumab, methotrexate, megastrol acetate, docetaxel, paclitaxel, testolactone, aziridine, vinblastine, capecitabine, goselerin acetate, zoledronic acid, taxol, vinblastine, and/or vincristine.
[0135] In some embodiments, the cancer can be resistant to certain chemotherapeutic agents. Administration of the of the polymeric nanoparticles of the present disclosure can be an alternative therapy when a different therapy, vulnerable to resistance, has been attempted unsuccessfully. Alternatively or additionally, the therapies of the instant disclosure can offer alternative forms or administration of chemotherapeutic drugs that can reduce the effect of resistance to the drugs.
[0136] In some embodiments, the composition or pharmaceutical composition comprising the polymeric nanoparticles can be administered to the subject via an administration route. For example, the composition or pharmaceutical composition can be administered intravenously, intratumorally, or subcutaneously.
[0137] In some embodiments of the methods, the composition can be administered at least once per day, once every other day, once per week, twice per week, once per month, or twice per month. In an embodiment of the methods, the composition is administered at least once per day. In an embodiment of the methods, the composition is administered at least once every other day. In an embodiment of the methods, the composition is administered at least once per week. In an embodiment of the methods, the composition is administered at least twice per week. In an embodiment of the methods, the composition is administered at least once per month. In an embodiment of the methods, the composition is administered at least twice per month. In another embodiment, the composition is administered more than once per day.
[0138] In some embodiments of the methods, the composition is administered over a period of three weeks. In other embodiments of the methods, the composition is administered over a period of 30 days. In other embodiments of the methods, the composition is administered over a period of 60 days. In other embodiments of the methods, the composition is administered over a period of 90 days. In other embodiments of the methods, the composition is administered over a period of 120 days. In other embodiments of the methods, the composition is administered over a period of 150 days. In other embodiments of the methods, the composition is administered over a period of 6 months. In other embodiments of the methods, the composition is administered over a period of about 6 months to about 1 year. In other embodiments of the methods, the composition is administered over a period of about 1 year to about 2 years.
[0139] The effective dosage of the polymeric nanoparticles provided herein may vary depending on the chemotherapeutic agent(s) used, the mode of administration, the condition being treated, and the severity of the condition being treated. The dosage regimen of the polymeric nanoparticle can be selected in accordance with a variety of factors including the route of administration and the renal and hepatic function of the patient. In certain embodiments, the therapeutically effective amount can be a human equivalent dose that is determined from an animal experiment.
Examples
[0140] The disclosure will now be illustrated with working examples, which are intended to illustrate the working of disclosure and not intended to restrict the scope of the present disclosure. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice of the disclosed methods and compositions, these exemplary methods, devices and materials are described herein. Example 1
Synthesis of di-block and penta-block copolymers [0141] Di-block and penta-block copolymers can serve as starting materials for the manufacture of polymeric nanoparticles of the present disclosure. An exemplary method that was performed to prepare these starting is provided below.
[0142] The penta-block copolymer is synthesized from initiator PLURONIC® L-61. PLURONIC® L-61 comprises polyoxypropylene with a molecular mass of 1800 g/mol and comprises a 10% polyoxyethylene content (poloxamer 181). PLURONIC® L-61 is available from BASF SE, Ludwigshafen, Germany. The di-block copolymer is synthesized from initiator mPEG5000. Both syntheses occur by ring-opening polymerization of lactide in the presence of a Sn-catalyst in tetraglyme as the solvent. Purification is performed through multiple rounds of precipitation to remove the catalyst, solvent, and lactic acid impurities to yield both polymers that are released according to the release criteria separately. The polymers may be stored as starting materials for manufacturing polymeric nanoparticles.
Example 2
Preparation of polymeric nanoparticles
[0143] Polymeric nanoparticles of the present disclosure associated with salinomycin and an anthracycline chemotherapeutic agent can be prepared as follows.
[0144] 100 mg of PLA-based block copolymers (penta-block PLA-PEG-PPG-PEG-PLA and di-block PLA-PEG hybrid block copolymers) were dissolved in 5ml acetonitrile to create a first mixture. Otherwise equivalent replicates were formulated to comprise various ratios of penta- to di -block copolymers (Masspenta-biock:Massdi-biock). Separately, 100 mg PLURONIC® F127 was dissolved in 20 mL double distilled (dd) water, followed by addition of 5pL triethylamine to create a second mixture. The inventors of the present disclosure have found that addition of the triethylamine (or other pH-raising compounds) can aid in association of the anthracycline chemotherapeutic compounds with the polymeric nanoparticles.
[0145] Next, anthracycline chemotherapeutic compounds and salinomycin were dissolved in 100 pL DMSO to create a third mixture. Mixtures of either salinomycin and doxorubicin in 1:3, 1:1, and 3:1 ratios or pirarubicin and salinomycin in 1:3, 1:1, and 1:3 ratios were used. Additionally, nanoparticles containing either only doxorubicin or only pirarubicin were also prepared.
[0146] The third mixture was added to the first mixture in order to create the various combinations of drugs and nanoparticle polymer ratios (and a fourth mixture). [0147] The fourth mixture was added slowly to the second mixture using a 26-gauge needle and syringe. The resulting emulsion was allowed to stir, and the organic solvent was removed by evaporation.
[0148] After complete evaporation of the organic solvent, the chemotherapeutic drug- loaded polymeric nanoparticles were filtered using AMICON® filters (ultracentrifuge filters available from MilliporeSigma, with a molecular weight cut off of 3,000 Da) at 4,000 rpm for 30 min to separate the unencapsulated drug from the nanoparticles. The polymeric nanoparticles were redispersed in 10 mL dd water, with 50 mg glucose (dissolved in 5 mL dd water) added to the nanoparticles and gently mixed (total volume of 15mL). The polymeric nanoparticles were stored at -20°C until further use.
[0149] All of the above steps were performed in a darkened workspace, to limit exposure of the various chemical products to light.
[0150] Tables 1 and 2, below, summarize various physical properties of the resulting nanoparticles prepared in this manner. Regarding Tables 1 and 2, these NP formulations contained 40 mg of penta- and di -block copolymers, 40 mg of FI 27 polymer, and 20 mg of glucose.
Table 1 Polymeric Nanoparticle Properties (SAL is salinomycin, DOX is doxorubicin)
Figure imgf000027_0001
Figure imgf000028_0002
Table 2 Polymeric Nanoparticle Properties (SAL is salinomycin, PIRA is pirarubicin)
Figure imgf000028_0001
Example 3
In vitro release assay
[0151] An experiment was conducted to measure the release of various chemotherapeutic drugs from polymeric nanoparticles according to the present disclosure. The polymeric nanoparticles were prepared according to Example 1.
[0152] Polymeric nanoparticles comprising doxorubicin alone, salinomycin alone, or both salinomycin and doxorubicin (at ratio of SAL:DOX of 1 :3) were suspended in phosphate buffered saline at a pH of 7.4 at 37°C, with shaking at 120 rpm. Release percentages for both drugs from each of the three polymeric nanoparticle formulations were measured over time at 1, 2, 3, 4, 5, 6, 7, 8, 9, and 12 days. Results are shown in FIG. 2 and Table 3. FIG. 2 shows the cumulative percentage of the drugs released by the various nanoparticle compositions that were tested. Data from the NPs containing salinomycin and doxorubicin in a ratio of 1 :3 are shown individually for each of salinomycin and doxorubicin. Table 3 Drag Release Data
Figure imgf000029_0001
Example 4
In vitro cell proliferation inhibition assay
[0153] An experiment was conducted to measure the ability of polymeric nanoparticles of the present disclosure to inhibit the proliferation of SUM149 cells. SUM149 cells are a triple negative breast cancer (TNBC) cell line. Cells were grown in cell culture and treated with one of five different NP formulations (SAL-loaded NPs, DOX-loaded NPs, NPs loaded with a 3 : 1 SAL:DOX ratio, NPs loaded with a 1 : 1 SAL:DOX ratio, and NPs loaded with a 1 :3 SAL:DOX ratio).
[0154] Tables 4 and 5 and FIG. 3 show the results of the cell proliferation assay. Surprisingly, the NPs loaded with a 1:3 SAL:DOX ratio showed synergistic activity, with a particularly small IC50 for SAL. Additionally, the combination index was particularly favorable for the 1 :3 SAL:DOX formulation. All SAL/DOX combination NP formulations showed increased and surprising performance, relative to the single drug NPs.
Table 4 IC50 for SAL and DOX in various NP formulations
Figure imgf000029_0002
Figure imgf000030_0001
Table 5 Performance of various NP formulations
Figure imgf000030_0002
Example 5
EAC tumor regression in a syngeneic mouse model [0155] An experiment was conducted to examine the ability of NPs to reduce the growth of tumors in mice. Specifically, syngeneic mice that develop Ehrlic Ascites Carcinoma (EAC) tumors were used. EAC is a spontaneous murine mammary adenocarcinoma mouse model in tumor biology. Mice were treated with NPs associated with either SAL (administered at 3 mg/kg), DOX (administered at 3 mg/kg), or a 1:3 ratio of SAL:DOX (administered with DOX at 3 mg/kg and SAL at 1 mg/kg). Details of administration and dosing are described in Table 6. Administration of NPs was accomplished intravenously. Doses were administered twice per week, for four weeks, with a dose administered on days 1, 4, 8, 11, 15, 18, 22, and 25.
[0156] FIG. 4 shows the results of the experiment. Tumor volume grew when mice were treated with a vehicle control and grew more slowly with treatment with either SAL NPs or DOX NPs. Surprisingly however, tumor growth was inhibited (or prevented) most effectively when mice were treated with NPs associated with a 1 :3 ratio of SAL:DOX.
Table 6 Dosing calculations for NP treatments
Figure imgf000031_0001
Example 6
In vitro cell proliferation inhibition assay
[0157] An experiment was conducted to measure the ability of NP-free, unencapsulated pirarubicin and doxorubicin to inhibit growth of cancer cells. MDA-MB 231 cancer cells were grown in culture and exposed to either NP-free, unencapsulated doxorubicin or pirarubicin at various concentrations.
[0158] FIG. 5 shows the percent of MDA-MB 231 cells that survived treatment with the drugs at various concentrations. Based on the data, free doxorubicin showed an IC50 of 0.242 mM and free pirarubicin showed an IC50 of 0.207 mM.
Example 7 In vitro release assay
[0159] An experiment was conducted to measure the release of pirarubicin from polymeric nanoparticles according to the present disclosure. Release was measured at various pH levels.
[0160] Polymeric nanoparticles comprising pirarubicin were suspended in phosphate buffered saline at a pH of 7.4 at 37°C, with shaking at 150 rpm. The release percentage for pirarubicin was measured over time at 1, 2, 3, 6, 8, 11, 13, and 17 days. Results are shown in FIG. 6A. FIG. 6A shows the cumulative percentage of pirarubicin released by the nanoparticle composition. Specifically, cumulative release of pirarubicin from nanoparticles over 15 days was approximately 22% of total encapsulated pirarubicin concentration.
[0161] In a similar experiment, release was again measured from pirarubicin-containing NPs. In this experiment, release was measured at both a pH of 7.4 and a pH of 5 at 0, 1, 2, 3, 6, 8, 11, 13, and 15 days. Both measurements were conducted at 37°C, with shaking at 150 rpm. FIG. 6B shows that release of pirarubicin was markedly increased in the lower pH environment. Specifically, cumulative release of pirarubicin from nanoparticles over 15 days at pH 7.4 was approximately 22% of the total encapsulated pirarubicin concentration. Cumulative release of pirarubicin from nanoparticles over 15 days at pH 5 was approximately 89% of the total encapsulated pirarubicin concentration.
Example 8
In vitro cell proliferation inhibition assay
[0162] An experiment was conducted to measure the ability of polymeric nanoparticles of the present disclosure to inhibit the proliferation of SUM149 and MDA-MB 468 cancer cell lines. Cells lines were grown separately in cell culture and each was treated independently with free pirarubicin and pirarubicin-loaded NPs.
[0163] FIG. 7A and FIG. 7B show the results of the cell proliferation assay. The NPs loaded with pirarubicin showed a lower IC50 than the treatment with free pirarubicin. For the SUM149 cell line (FIG. 7A), the IC50 was 3.245 mM for free pirarubicin and 0.2283 pM for the NPs loaded with pirarubicin. For the MDA-MB 468 cell line (FIG. 7B), the IC50 was 0.6007 mM for free pirarubicin and 0.1504 mM for the NPs loaded with pirarubicin.
Example 9
In vitro cell proliferation inhibition assay
[0164] An experiment was conducted to measure the ability of polymeric nanoparticles of the present disclosure to inhibit the proliferation of SUM149 and MDA-MB 468 cancer cell lines. 4 replicates of each cell line were grown separately in cell culture and each replicate was treated independently with one of pirarubicin-loaded NPs, or NPs loaded with one of three combinations of pirarubicin and salinomycin in a mass ratio (P IRA: SAL) of 3:1, 1:1, or 1:3.
[0165] FIG. 8A and Table 7 show the results of the cell proliferation assay in SUM149 cells. Unexpectedly, The NPs loaded with a 1:3 ratio of pirarubicin to salinomycin showed the lowest IC50 and combination index (Cl). FIG. 8B and Table 8 show the results of the cell proliferation assay in MDA-MB 468 cells. Similar to the data seen in SUM149 cells, the NPs loaded with a 1:3 ratio of pirarubicin to salinomycin showed the lowest IC50 and combination index (Cl).
Table 7 NP performance against SUM149 cells
Figure imgf000033_0001
Table 8 NP performance against MDA-MB 468 cells
Figure imgf000033_0002
Figure imgf000034_0001
Example 10
In vitro cell proliferation inhibition assay
[0166] An experiment was conducted to measure the ability of polymeric nanoparticles of the present disclosure to inhibit the proliferation of SUM149 and MDA-MB 468 cancer cell lines. 5 replicates of each cell line were grown separately in cell culture and each replicate was treated independently with one of pirarubicin-loaded NPs, salinomycin-loaded NPs, or NPs loaded with one of three combinations of pirarubicin and salinomycin in a mass ratio (PHL SAL) of 3:1, 1:1, or 1:3.
[0167] FIG. 9A and Table 9 show the results of the cell proliferation assay in SUM149 cells. Unexpectedly, The NPs loaded with a 1:3 ratio of pirarubicin to salinomycin showed the lowest IC50 and greatest reduction in IC50. FIG. 9B and Table 10 show the results of the cell proliferation assay in MDA-MB 468 cells. Similar to the data seen in SUM149 cells, the NPs loaded with a 1 :3 ratio of pirarubicin to salinomycin showed the lowest IC50 and the greatest reduction in IC50.
Table 9 Proliferation assay results in SUM149 cells
Figure imgf000034_0002
Table 10 Proliferation assay results in MDA-MB 468 cells
Figure imgf000035_0001
[0168] One skilled in the art will appreciate further features and advantages of the invention based on the above-described embodiments. Accordingly, the invention is not to be limited by what has been particularly shown and described, except as indicated by the appended claims. All publications and references cited herein are expressly incorporated herein by reference in their entirety.

Claims

CLAIMS What is claimed is:
1. A composition comprising: a) polymeric nanoparticles comprising block copolymers comprising a poly(lactic acid)-poly(ethylene glycol)-poly(propylene glycol)-poly(ethylene glycol)-poly(lactic acid) (PLA-PEG-PPG-PEG-PLA) penta-block copolymer with a poly(lactic acid)-poly(ethylene glycol) (PLA-PEG) di-block copolymer; b) salinomycin; and c) an anthracycline chemotherapeutic compound, wherein both the salinomycin and the anthracycline chemotherapeutic compound are associated with the polymeric nanoparticles.
2. The composition of claim 1, wherein the salinomycin and the anthracycline chemotherapeutic compound are both associated substantially with the same polymeric nanoparticles.
3. The composition of claim 1 or 2, wherein the anthracycline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof.
4. The composition of claim 1 or 2, wherein the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin.
5. The composition of claim 1 or 2, wherein the anthracycline chemotherapeutic compound is doxorubicin.
6. The composition of claim 1 or 2, wherein the anthracycline chemotherapeutic compound is pirarubicin.
7. The composition of any one of claims 1-6, wherein the total mass of anthracycline chemotherapeutic compound is greater than or equal to the total mass of the salinomycin.
8. The composition of claim 7, wherein the anthracy cline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1.5:1.
9. The composition of claim 7 or 8, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 2:1.
10. The composition of any one of claims 7-9, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 2.5:1 to 3.5:1.
11. The composition of any one of claims 1-6, wherein the total mass of anthracycline chemotherapeutic compound is less than or equal to the total mass of the salinomycin.
12. The composition of claim 11, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1:1.5.
13. The composition of claim 11 or 12, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1 :2.
14. The composition of any one of claims 11-13, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 1 :2.5 to 1 :3.5.
15. The composition of any one of claims 1-6, wherein the total mass of anthracycline chemotherapeutic compound is about equal to the total mass of the salinomycin.
16. The composition of any one of claims 1-15, wherein less PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than PLA-PEG di-block copolymer.
17. The composition of claim 16, wherein a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1 :20 to 1:1.
18. The composition of claim 16 or 17, wherein a mass ratio of PLA-PEG-PPG-PEG- PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:15 to 1:2.
19. The composition of any one of claims 16-18, wherein a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1 :8 to 1:10.
20. The composition of any one of claims 16-18, wherein a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:3 to 1:5.
21. The composition of any one of claims 16-18, wherein a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1 :2 to 3 :8.
22. The composition of any one of claims 1-21, wherein the average diameter of the polymeric nanoparticles is between 50 and 170 nm.
23. The composition of any one of claims 1-22, wherein the average diameter of the polymeric nanoparticles is between 60 and 130 nm.
24. The composition of any one of claims 1-23, wherein the average diameter of the polymeric nanoparticles is between 60 and 100 nm.
25. The composition of any one of claims 1-23, wherein the average diameter of the polymeric nanoparticles is between 80 and 110 nm.
26. The composition of any one of claims 1-22, wherein the average diameter of the polymeric nanoparticles is between 100 and 170 nm.
27. The composition of any one of claims 1-26, wherein a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.5.
28. The composition of any one of claims 1-27, wherein a PDI of the polymeric nanoparticles is not more than 0.3.
29. The composition of any one of claims 1-28, wherein a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV.
30. The composition of any one of claims 1-29, further comprising a PEG-PPG-PEG triblock copolymer.
31. A pharmaceutical composition comprising the composition of any one of claims 1-30, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
32. A method of reducing proliferation, survival, migration, or colony formation ability of a rapidly proliferating cell in a subject in need thereof, comprising contacting the cell with a therapeutically effective amount of a composition comprising: the composition of any one of claims 1-30 or the pharmaceutical composition of claim 31.
33. The method of claim 32, wherein the cell is a cancer cell.
34. The method of claim 32 or 33, wherein the cell is a cancer stem cell.
35. A method for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a composition comprising: the composition of any one of claims 1-30 or the pharmaceutical composition of claim 31.
36. The method of claim 35, wherein the cancer comprises a solid tumor cancer or a cancer of the blood.
37. The method of any one of claims 35 or 36, wherein the cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, leukemia, lymphoma, osteosarcoma, gastric cancer, prostate cancer, colon cancer, non-small cell lung cancer and small cell lung cancer, liver cancer, kidney cancer, head and neck cancer, cervical cancer, acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, and combinations thereof.
38. The method of any one of claims 35-37, wherein the cancer or breast cancer comprises triple negative breast cancer (TNBC).
39. The method of any one of claims 35-38, wherein the cancer is metastatic.
40. The method of any one of claims 35-39, further comprising administering an additional anti-cancer therapy to the subject.
41. The method of claim 40, wherein the additional anti-cancer therapy comprises surgery, chemotherapy, radiation, hormone therapy, immunotherapy, or a combination thereof.
42. The method of any one of claims 35-41, wherein the cancer is resistant or refractory to a chemotherapeutic agent.
43. The method of any one of claims 35-42, wherein the subject is a human.
44. The method of any one of claims 35-43, wherein the composition or pharmaceutical composition is administered intravenously, intratumorally, or subcutaneously.
45. A method of manufacturing polymeric nanoparticles comprising: a) mixing penta-block PLA-PEG-PPG-PEG-PLA and di-block PLA-PEG block copolymers dissolved in acetonitrile with salinomycin and at least one anthracycline compound to form a first mixture; b) mixing the first mixture with a PEG-PPG-PEG triblock copolymer dissolved in water to form a second mixture; c) stirring the second mixture and evaporating the acetonitrile; and d) filtering the stirred and evaporated second mixture, thereby manufacturing the polymeric nanoparticles.
46. The method of claim 45, wherein the salinomycin and at least one anthracycline chemotherapeutic compound are dissolved in DMSO when used in step a).
47. The method of claim 45 or 46, wherein the PEG-PPG-PEG triblock copolymer comprises poloxamer 407.
48. The method of any one of claims 45-47, further comprising adding triethylamine during step b).
49. The method of any one of claims 45-48, wherein the anthracycline chemotherapeutic compound is selected from the group consisting of: doxorubicin, pirarubicin, daunorubicin, epirubicin, idarubicin, nemorubicin and a combination thereof.
50. The method of claim 49, wherein the anthracycline chemotherapeutic compound is doxorubicin or pirarubicin.
51. The method of claim 49, wherein the anthracycline chemotherapeutic compound is doxorubicin.
52. The method of claim 49, wherein the anthracycline chemotherapeutic compound is pirarubicin.
53. The method of any one of claims 45-52, wherein the total mass of anthracycline chemotherapeutic compound is greater than or equal to the total mass of the salinomycin.
54. The method of claim 53, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1.5:1.
55. The method of claim 53 or 54, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 2:1.
56. The method of any one of claims 53-55, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 2.5:1 to 3.5:1.
57. The method of any one of claims 45-52, wherein the total mass of anthracycline chemotherapeutic compound is less than or equal to the total mass of the salinomycin.
58. The method of claim 57, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1:1.5.
59. The method of claim 57 or 58, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of at least 1 :2.
60. The method of any one of claims 57-59, wherein the anthracycline chemotherapeutic compound and the salinomycin are present in a mass ratio of 1 :2.5 to 1 :3.5.
61. The method of any one of claims 45-52, wherein the total mass of anthracycline chemotherapeutic compound is about equal to the total mass of the salinomycin.
62. The method of any one of claims 45-61, wherein less PLA-PEG-PPG-PEG-PLA penta-block copolymer is present, by mass, than PLA-PEG di-block copolymer.
63. The method of claim 62, wherein a mass ratio of PLA-PEG-PPG-PEG-PLA penta- block copolymer to PLA-PEG di-block copolymer is from 1 :20 to 1:1.
64. The method of claim 62 or 63, wherein a mass ratio of PLA-PEG-PPG-PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1:15 to 1:2.
65. The method of any one of claims 62-64, wherein a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di -block copolymer is from 1 :8 to 1:10.
66. The method of any one of claims 62-64, wherein a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1:3 to 1:5.
67. The method of any one of claims 62-64, wherein a mass ratio of PLA-PEG-PPG- PEG-PLA penta-block copolymer to PLA-PEG di-block copolymer is from 1 :2 to 3 :8.
68. The method of any one of claims 45-67, wherein the average diameter of the polymeric nanoparticles is between 50 and 170 nm.
69. The method of any one of claims 45-67, wherein the average diameter of the polymeric nanoparticles is between 60 and 130 nm.
70. The method of any one of claims 45-67, wherein the average diameter of the polymeric nanoparticles is between 60 and 100 nm.
71. The method of any one of claims 45-67, wherein the average diameter of the polymeric nanoparticles is between 80 and 110 nm.
72. The method of any one of claims 45-67, wherein the average diameter of the polymeric nanoparticles is between 100 and 170 nm.
73. The method of any one of claims 45-72, wherein a polydispersity index (PDI) of the polymeric nanoparticles is not more than 0.5.
74. The method of any one of claims 45-73, wherein a PDI of the polymeric nanoparticles is not more than 0.3.
75. The method of any one of claims 45-74, wherein a zeta potential of the polymeric nanoparticles is from -5 mV to -40 mV.
76. The pharmaceutical composition of claim 31 for use in reducing proliferation, survival, migration, or colony formation ability of a rapidly proliferating cell in a subject in need thereof.
77. The pharmaceutical composition for use according to claim 76, wherein the cell is a cancer cell.
78. The pharmaceutical composition for use according to claim 76 or 77, wherein the cell is a cancer stem cell.
79. The pharmaceutical composition of claim 31 for use in treating cancer.
80. The pharmaceutical composition for use according to claim 79, wherein the cancer comprises a solid tumor cancer or a cancer of the blood.
81. The pharmaceutical composition for use according to any one of claims 79 or 80, wherein the cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, leukemia, lymphoma, osteosarcoma, gastric cancer, prostate cancer, colon cancer, non-small cell lung cancer and small cell lung cancer, liver cancer, kidney cancer, head and neck cancer, cervical cancer, acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, and combinations thereof.
82. The pharmaceutical composition for use according to any one of claims 79-81, wherein the cancer or breast cancer comprises TNBC.
83. The pharmaceutical composition for use according to any one of claims 79-82, wherein the cancer is metastatic.
84. The pharmaceutical composition for use according to any one of claims 79-83, further comprising administering an additional anti-cancer therapy to the subject.
85. The pharmaceutical composition for use according to claim 84, wherein the additional anti-cancer therapy comprises surgery, chemotherapy, radiation, hormone therapy, immunotherapy, or a combination thereof.
86. The pharmaceutical composition for use according to any one of claims 76-85, wherein the cancer is resistant or refractory to a chemotherapeutic agent.
87. The pharmaceutical composition for use according to any one of claims 76-86, wherein the subject is a human.
88. The pharmaceutical composition for use according to any one of claims 76-87, wherein the pharmaceutical composition is administered intravenously, intratumorally, or subcutaneously.
PCT/US2022/072563 2021-05-25 2022-05-25 Polymeric nanoparticles comprising chemotherapeutic compounds and related methods WO2022251844A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163192828P 2021-05-25 2021-05-25
US63/192,828 2021-05-25

Publications (1)

Publication Number Publication Date
WO2022251844A1 true WO2022251844A1 (en) 2022-12-01

Family

ID=82361337

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/072563 WO2022251844A1 (en) 2021-05-25 2022-05-25 Polymeric nanoparticles comprising chemotherapeutic compounds and related methods

Country Status (1)

Country Link
WO (1) WO2022251844A1 (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060165987A1 (en) 2002-04-05 2006-07-27 Patrice Hildgen Stealthy polymeric biodegradable nanospheres and uses thereof
US20080081075A1 (en) 2006-10-02 2008-04-03 National Tsing Hua University Multifunctional mixed micelle of graft and block copolymers and preparation thereof
US20100004398A1 (en) 2006-07-28 2010-01-07 Xiaorong Wang Polymeric core-shell nanoparticles with interphase region
US8883842B2 (en) 2005-08-03 2014-11-11 Novartis Ag Use of HDAC inhibitors for the treatment of myeloma
US9149426B2 (en) 2012-02-15 2015-10-06 University Of Tennessee Research Foundation Nanoparticle composition and methods to make and use the same
WO2018049155A1 (en) * 2016-09-08 2018-03-15 Dana-Farber Cancer Institute, Inc. Compositions comprising polymeric nanoparticles and mcl-1 antagonists
US20190091280A1 (en) * 2012-04-23 2019-03-28 Nanoproteagen Ltd. Polymeric nanoparticles and a process of preparation thereof
US20190321305A1 (en) * 2016-11-02 2019-10-24 Nanoproteagen Polymeric nanoparticles
US20200046648A1 (en) * 2018-07-18 2020-02-13 Surender Kharbanda Polymeric nanoparticles comprising salinomycin

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060165987A1 (en) 2002-04-05 2006-07-27 Patrice Hildgen Stealthy polymeric biodegradable nanospheres and uses thereof
US8883842B2 (en) 2005-08-03 2014-11-11 Novartis Ag Use of HDAC inhibitors for the treatment of myeloma
US20100004398A1 (en) 2006-07-28 2010-01-07 Xiaorong Wang Polymeric core-shell nanoparticles with interphase region
US20080081075A1 (en) 2006-10-02 2008-04-03 National Tsing Hua University Multifunctional mixed micelle of graft and block copolymers and preparation thereof
US9149426B2 (en) 2012-02-15 2015-10-06 University Of Tennessee Research Foundation Nanoparticle composition and methods to make and use the same
US20190091280A1 (en) * 2012-04-23 2019-03-28 Nanoproteagen Ltd. Polymeric nanoparticles and a process of preparation thereof
WO2018049155A1 (en) * 2016-09-08 2018-03-15 Dana-Farber Cancer Institute, Inc. Compositions comprising polymeric nanoparticles and mcl-1 antagonists
US20190321305A1 (en) * 2016-11-02 2019-10-24 Nanoproteagen Polymeric nanoparticles
US20200046648A1 (en) * 2018-07-18 2020-02-13 Surender Kharbanda Polymeric nanoparticles comprising salinomycin

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BORCHARD ET AL., PHARM. RES., vol. 7, 1996, pages 1055 - 1058
CHO ET AL.: "Therapeutic Nanoparticles for Drug Delivery in Cancer", CLIN. CANCER RES., vol. 14, 2008, pages 1310 - 1316, XP055067421, DOI: 10.1158/1078-0432.CCR-07-1441
DONG WEI ET AL: "Precise engineering of dual drug-loaded polymeric nanoparticles system to improve the treatment of glioma-specific targeting therapy", PROCESS BIOCHEMISTRY, ELSEVIER LTD, GB, vol. 102, 23 January 2021 (2021-01-23), pages 341 - 348, XP086497152, ISSN: 1359-5113, [retrieved on 20210123], DOI: 10.1016/J.PROCBIO.2021.01.018 *
FAROKHZAD OC ET AL.: "Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo", PROC. NATL. ACAD. SCI. USA, vol. 103, no. 16, 2006, pages 6315 - 20, XP002495677, DOI: 10.1073/pnas.0601755103
FONSECA C ET AL.: "Paclitaxel-loaded PLGA nanoparticles: preparation, physicochemical characterization and in vitro anti-tumoral activity", J. CONTROLLED RELEASE, vol. 83, no. 2, 2002, pages 273 - 86, XP004383637, DOI: 10.1016/S0168-3659(02)00212-2
HOOD ET AL., NANOMEDICINE, vol. 6, no. 7, 2011, pages 1257 - 1272
SOSNIK ET AL.: "Polymeric Nanocarriers: New Endeavors for the Optimization of the Technological Aspects of Drugs", RECENT PATENTS ON BIOMEDICAL ENGINEERING, vol. 1, 2008, pages 43 - 59
SPADA ET AL.: "Protein delivery of polymeric nanoparticles", WORLD ACADEMY OF SCIENCE, ENGINEERING AND TECHNOLOGY, vol. 76, 2011
SUNDAR ET AL., SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS, 2010

Similar Documents

Publication Publication Date Title
KR100849911B1 (en) Tumor-targeting drug-loaded particles
US20150099001A1 (en) Nanocell Drug Delivery System
CN101605535A (en) Treat breast carcinoma with the nano-particle that comprises taxane based on hormone receptor status
US9220773B2 (en) Pharmaceutical composition
AU2017343553A1 (en) Nanoparticle formulations and methods of making and using thereof
Oliveira et al. Strategies to target tumors using nanodelivery systems based on biodegradable polymers, aspects of intellectual property, and market
Moradpour et al. Novel approaches for efficient delivery of tyrosine kinase inhibitors
WO2018085407A1 (en) Polymeric nanoparticles
WO2018049155A1 (en) Compositions comprising polymeric nanoparticles and mcl-1 antagonists
WO2020210805A1 (en) Nanoparticles for drug delivery to brain
US20200046648A1 (en) Polymeric nanoparticles comprising salinomycin
US10709795B2 (en) Method for delivering pharmaceutical nanoparticles to cancer cells
WO2022251844A1 (en) Polymeric nanoparticles comprising chemotherapeutic compounds and related methods
EP3714257A1 (en) Polymeric nanoparticles comprising bortezomib
JP6824535B2 (en) Compositions and Methods for Improving Nanoparticle Distribution in the Brain Interstitium
CN114632069A (en) Polymeric nanoparticles
EP3077007A1 (en) Reversible pegylation of nanocarriers
Pancholi et al. Nanocarrier-based Targeted Delivery in Cancer
Qamar et al. Combination drug loaded lipid-based nanocarriers as treatment entity for battling glioblastoma multiforme
RU2675810C1 (en) Polymeric complex for molecular targeting therapy and method for its obtaining
WO2023034605A1 (en) Polymeric nanoparticles comprising a histone deacetylase 6 / phosphoinositide 3-kinase-8 dual inhibitor and related methods
Ahuja et al. NANOPARTICLES IN TARGETED DRUG DELIVERY SYSTEM
BINGFENG Trastuzumab-decorated nanoparticles of biodegradable polymers for targeted small molecule chemotherapy
TW201720447A (en) Nanoparticles loaded with active ingredients, their process of preparation and their uses

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: 22736458

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE