WO2016141161A1 - Dual loaded liposomal pharmaceutical formulations - Google Patents

Dual loaded liposomal pharmaceutical formulations Download PDF

Info

Publication number
WO2016141161A1
WO2016141161A1 PCT/US2016/020647 US2016020647W WO2016141161A1 WO 2016141161 A1 WO2016141161 A1 WO 2016141161A1 US 2016020647 W US2016020647 W US 2016020647W WO 2016141161 A1 WO2016141161 A1 WO 2016141161A1
Authority
WO
WIPO (PCT)
Prior art keywords
liposome
cancer
cholesterol
doxorubicin
lipid
Prior art date
Application number
PCT/US2016/020647
Other languages
French (fr)
Inventor
De-Min Zhu
Guoqiang Chen
Original Assignee
Cureport, 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 Cureport, Inc. filed Critical Cureport, Inc.
Priority to EP16759478.7A priority Critical patent/EP3265063A4/en
Priority to CN201680013636.7A priority patent/CN107530291A/en
Priority to JP2017546067A priority patent/JP2018507227A/en
Priority to CA2977397A priority patent/CA2977397A1/en
Publication of WO2016141161A1 publication Critical patent/WO2016141161A1/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
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • 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
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1277Processes for preparing; Proliposomes
    • A61K9/1278Post-loading, e.g. by ion or pH gradient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention generally relates to liposomal pharmaceutical formulations and, in various embodiments, more specifically to liposomal pharmaceutical formulations including an active pharmaceutical ingredient with two components (e.g., a combination of docetaxel and doxorubicin).
  • an active pharmaceutical ingredient e.g., a combination of docetaxel and doxorubicin.
  • Liposome technology has been utilized for drug delivery in clinical therapy and scientific research. To date, a handful of liposomal pharmaceutical formulations have been approved by the US Food and Drug Administration (“FDA”), and a number of new liposomal formulations are in clinical trials. However, the field of liposomal formulation is still evolving and each active pharmaceutical ingredient (“API”) presents unique challenges.
  • FDA US Food and Drug Administration
  • liposomal formulations of doxorubicin are presently available under the trade names Doxil® and Myocet®.
  • Doxil® is a pegylated (polyethylene glycol coated) liposome- encapsulated form of doxorubicin formerly made by Ben Venue Laboratories in the United States for Janssen Products, LP, a subsidiary of Johnson & Johnson.
  • Myocet® is a non- pegylated liposomal doxorubicin made by Enzon Pharmaceuticals for Cephalon in Europe and for Sopherion Therapeutics in the United States and Canada. Myocet® is approved in Europe and Canada for treatment of metastatic breast cancer in combination with
  • a liposome can include (i) an active pharmaceutical ingredient (API) comprising a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin); (ii) a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid; and (iii) an aqueous interior, wherein the first drug (e.g., docetaxel) is in the lipid layer and the second drug (e.g., doxorubicin) is crystallized in the aqueous interior.
  • API active pharmaceutical ingredient
  • a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid
  • an aqueous interior wherein the first drug (e.g., docetaxel) is in the lipid layer and the second drug (e.g., doxorubicin) is crystallized in the aqueous interior.
  • the liposomes can be used to treat a subject, for example, a human subject having cancer.
  • the cancer can be, for example, a lung cancer, preferably non- small cell lung cancer (NSCLC); colon cancer; breast cancer; or liver cancer, preferably hepatocellular carcinoma (HCC).
  • NSCLC non- small cell lung cancer
  • HCC hepatocellular carcinoma
  • the invention can provide for increased efficacy and/or decreased toxicity, for example relative to (i) other pharmaceutical compositions where one or both of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin) are not in a liposomal formulation and/or (ii) other liposomal formulations.
  • the invention can provide for targeted delivery, for example to the liver or avoiding the liver.
  • the invention can mitigate undesired side effects, for example by providing for increased drug loading, thereby reducing the amount of liposomes needed to deliver a quantity of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin).
  • the invention provides a liposome comprising: (i) an active pharmaceutical ingredient (API) comprising docetaxel and doxorubicin; (ii) a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid; and (iii) an aqueous interior, wherein the docetaxel is in the lipid layer and the doxorubicin is crystallized in the aqueous interior.
  • API active pharmaceutical ingredient
  • doxorubicin a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid
  • an aqueous interior wherein the docetaxel is in the lipid layer and the doxorubicin is crystallized in the aqueous interior.
  • the invention also provides a pharmaceutical composition comprising a plurality of liposomes according to any of the aspects or embodiments disclosed herein.
  • the invention also provides a method comprising administering the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
  • the invention also provides a method of treating a subject comprising administering an effective amount of the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
  • the invention also provides a method of making the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, comprising: (i) introducing a lipid solution of an unsaturated phospholipid, cholesterol, a first drug (e.g., docetaxel), and preferably a pegylated phospholipid in ethanol through a first or more inlet port of a manifold into a mixing chamber and an aqueous solution through a second or more inlet port of the manifold into the mixing chamber, the liposomes formed exit the mixing chamber through a third or more outlet port of the manifold, thereby making a plurality of liposomes; and (ii) incubating the plurality of liposomes in a second drug (e.g., doxorubicin) solution.
  • a second drug e.g., doxorubicin
  • the lipid layer consists essentially of the unsaturated phospholipid and cholesterol.
  • the lipid layer consists essentially of the unsaturated phospholipid, cationic lipid, cholesterol, and pegylated phospholipid.
  • the API consists essentially of docetaxel and doxorubicin.
  • the lipid layer comprises: about 20-75%, preferably about 30-60%, (molar) unsaturated phospholipid; about 10-60%, preferably 20-50%, (molar) cholesterol; about 5-75%, preferably about 10-60%, (molar) cationic lipid; and about 0-20%, preferably 1-10%, (molar) pegylated phospholipid.
  • the molar ratio of the lipid layer [0016] In various embodiments, the molar ratio of the lipid layer
  • doxorubicin is about 100: 1 to about 2: 1, preferably about 20: 1 to about 5: 1 ; and the molar ratio of the lipid layer components: docetaxel is about 100: 1 to about 2: 1 , preferably about 20: 1 to about 5: 1.
  • the molar ratio of doxorubicin: docetaxel is about 10: 1 to 1: 10, preferably about 5: 1 to 1:5, and more preferably about 2: 1 to 1:2.
  • the unsaturated phospholipid comprises a polyunsaturated phospholipid or a monounsaturated phospholipid, preferably a
  • phosphatidylcholine and more preferably and soy phosphatidylcholine or 1 ,2-dioleoyl-sn- glycero-3-phosphatidylcholine (DOPC).
  • DOPC 1 ,2-dioleoyl-sn- glycero-3-phosphatidylcholine
  • the cholesterol comprises a cholesterol derivative, preferably a cationic cholesterol derivative, more preferably an amino cholesterol derivative, and still more preferably dimethylaminoethanecarbamoyl-cholesterol (DC-cholesterol).
  • a cholesterol derivative preferably a cationic cholesterol derivative, more preferably an amino cholesterol derivative, and still more preferably dimethylaminoethanecarbamoyl-cholesterol (DC-cholesterol).
  • DC-cholesterol dimethylaminoethanecarbamoyl-cholesterol
  • the pegylated phospholipid comprises a
  • phosphoethanolamine preferably a l,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and wherein the pegylation is a PEG 500 to PEG 3000, preferably PEG 2000.
  • DSPE disistearoyl-sn-glycero-3-phosphoethanolamine
  • the plurality of liposomes are comprised in an intravenous formulation.
  • the Z-average particle size of the liposomes is about 10-200 nm, preferably about 15- 150 nm, and more preferably about 20-120 nm.
  • at least about 10% of the composition is delivered to the liver.
  • the pharmaceutical composition is for use as a medicament.
  • the pharmaceutical composition is for use as a cancer therapeutic.
  • the subject has a cancer.
  • the cancer is a lung cancer, preferably non-small cell lung cancer (NSCLC); colon cancer; breast cancer; or liver cancer, preferably hepatocellular carcinoma (HCC).
  • NSCLC non-small cell lung cancer
  • HCC hepatocellular carcinoma
  • FIG. 1 presents a cryo transmission electron microscopy (TEM) image of liposomal formulation CPT319C.
  • FIG. 2 presents a negative stained TEM image of CPT319C.
  • FIG. 3 illustrates NSCLC tumor growth curves and tumor weight inhibition percentages (TW inh%) after administration of liposomal (CPT319A, CPT319B, or
  • CPT319C CPT319C
  • non-liposomal formulations of docetaxel/doxorubicin compared to a control group.
  • FIG. 4 illustrates NSCLC tumor growth curves and tumor weight inhibition percentages (TW inh%) after administration of liposomal (CPT307A, CPT307B, or
  • CPT307C CPT307C or non-liposomal formulations of docetaxel/doxorubicin, compared to the control group.
  • FIG. 5 illustrates colon cancer tumor growth curves after administration of three different doses of liposomal formulation (CPT319C), compared to the control group.
  • FIG. 6 illustrates colon cancer tumor growth curves after administration of three different doses of liposomal formulation (CPT307C), compared to the control group.
  • FIG. 7 illustrates breast cancer tumor growth curves on Day 31 after administration of liposomal formulations (CPT307C or CPT319C), compared to the control group.
  • FIG. 8 illustrates hepatocellular carcinoma tumor growth curves after administration of liposomal formulation (CPT319C), compared to the control group.
  • FIG. 9 illustrates NSCLC tumor growth curves after administration of liposomal formulations (CPT307A-C or CPT319A-C), compared to the control group.
  • FIG. 10 illustrates colon cancer tumor growth curves and tumor weight inhibition percentages (TW Inh ) after administration of liposomal formulations (CPT307C or CPT219C), compared to the control group.
  • FIG. 11 illustrates plasma concentration curves of doxorubicin after administration of liposomal formulations (CPT319C or CPT307C), compared to non- liposomal formulation of docetaxel/doxorubicin.
  • the invention provides a pharmaceutical composition including a plurality of liposomes comprising a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin).
  • a liposome can include (i) an active pharmaceutical ingredient (API) comprising a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin); (ii) a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid; and (iii) an aqueous interior, wherein the first drug (e.g., docetaxel) is in the lipid layer and the second drug (e.g., doxorubicin) is crystallized in the aqueous interior.
  • the liposomes can be used to treat a subject, for example, a human subject having cancer.
  • the invention can provide for increased efficacy and/or decreased toxicity, for example relative to (i) other pharmaceutical compositions where one or both of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin) are not in a liposomal formulation and/or (ii) other liposomal formulations.
  • the invention can provide for targeted delivery, for example to the liver or avoiding the liver.
  • the invention can mitigate undesired side effects, for example by providing for increased drug loading, thereby reducing the amount of liposomes needed to deliver a quantity of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin).
  • compositions including the liposomes and methods of using and making the liposomes are discussed, in turn, below.
  • the API comprises a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin). While docetaxel and doxorubicin are presented as illustrative examples, other embodiments are possible where the first drug is in the lipid layer of the liposome and the second drug is in (e.g., crystallized in) the aqueous interior of the liposome.
  • the API can comprise two (or more) anticancer agents, an anti-inflammatory agents, an anti-diabetic agents, an anti-fungal agents, and/or antibiotic agents.
  • Docetaxel (as generic or under the trade name Taxotere® or Docecad®) is a clinically well-established anti-mitotic chemotherapy medication that works by interfering with cell division. Docetaxel is approved by the FDA for treatment of locally advanced or metastatic breast cancer, head and neck cancer, gastric cancer, hormone-refractory prostate cancer and non small-cell lung cancer. Docetaxel can be used as a single agent or in combination with other chemotherapeutic drugs as indicated depending on specific cancer type and stage.
  • Docetaxel is a member of the taxane drug class, which also includes the chemotherapeutic medication paclitaxel. Accordingly, in some embodiments, docetaxel can be substituted for another taxane that can be disposed within the lipid layer of the liposome.
  • the invention can increase the efficacy of, and/or decrease undesired side effects from, the docetaxel.
  • Doxorubicin (trade name Adriamycin®; pegylated liposomal form trade name Doxil®; nonpegylated liposomal form trade name Myocet®), also known as
  • hydroxydaunorubicin and hydroxydaunomycin is a drug used in cancer chemotherapy and derived by chemical semisynthesis from a bacterial species. It is an anthracycline antibiotic (note: in this context, this does not mean it is used to treat bacterial infections) closely related to the natural product daunomycin and like all anthracyclines, it is believed to work by intercalating DNA, with the most serious adverse effect being life-threatening heart damage. It is commonly used in the treatment of a wide range of cancers, including hematological malignancies (blood cancers, like leukaemia and lymphoma), many types of carcinoma (solid tumors) and soft tissue sarcomas. It is often used in combination chemotherapy as a component of various chemotherapy regimens. In some embodiments, doxorubicin can be substituted for another anticancer agent that can be disposed within the aqueous interior of the liposome.
  • doxorubicin Common adverse effects of doxorubicin include hair loss (seen in most of those treated with the drug), myelosuppression (a compromised ability of the body's bone marrow to produce new blood cells), nausea and vomiting (which are seen in roughly 30-90% of people treated with the drug), oral mucositis, oesophagitis, diarrhea, skin reactions (including hand-foot syndrome) and localized swelling and redness along the vein in which the drug is delivered. Less common, yet serious reactions include hypersensitivity reactions (including anaphylaxis), radiation recall, heart damage and liver dysfunction.
  • the drug is administered intravenously, as the hydrochloride salt. It is sold under a number of different brand names, including Adriamycin® PFS, Adriamycin® RDF, or Rubex®.
  • Doxorubicin is photosensitive, and containers are often covered by an aluminum bag and/or brown wax paper to prevent light from affecting it.
  • Doxorubicin is also available in liposome-encapsulated forms as Doxil® (pegylated form), Myocet® (nonpegylated form), and Caelyx®, although these forms must also be given by intravenous injection.
  • the invention can increase the efficacy of and/or decrease undesired side effects from, the doxorubicin.
  • the API may be a polynucleotide (including an oligonucleotide) a protein or a small molecule.
  • the API is a polynucleotide.
  • the polynucleotide may be a genomic DNA fragment, cDNA, mRNA, ssRNA, dsRNA, microRNA, siRNA, shRNA, sdRNA, DsiRNA, LNA, and antisense DNA or RNA.
  • the API may be a small molecule drug.
  • the molecule has a molecular weight from about 1500 g/mole to about 50 g/mole.
  • An API can include, for example, two or more of the following: an anticancer agent, an antibiotic agent, an antiviral agent, an anti-fungal agent, or an analgesic.
  • exemplary anticancer agents may include but are not limited acivicin, aclarubicin, acodazole, ametantrone, aminoglutethimide, anthramycin, asparaginase, azacitidine, azetepa, bisantrene, bleomycin, busulfan, cactinomycin, calusterone, caracemide, carboplatin, carfilzomib, carmustine, carubicin, chlorambucil, cisplatin, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, dezaguanine, diaziquone, docetaxel, doxorubicin, epipropidine, erlot
  • triethylenemelamine trimetrexate, uracil mustard, uredepa, vinblastine, vincristine, vindesine, vinepidine, vinrosidine, vinzolidine, zinostatin and zorubicin.
  • Exemplary antibiotic agents may include but are not limited to
  • aminoglycoside aminoglycoside; amikacin; gentamicin; kanamycin; neomycin; netilmicin; steptomycin; tobramycin; ansamycins; geldanamycin; herbimycin; carbacephem; loracarbef;
  • carbacepenem ertapenem; doripenem; imipenem/cilastatin; meropenem; cephalosporin; cefadroxil; cefazolin; cefalotin or cefalothin; cefalexin; cefaclor; cefamandole; cefoxitin; cefprozil; cefuroxime; cefixime; cefdinir; cefditoren; cefoperazone; cefotaxime;
  • cefpodoxime ceftazidime; ceftibuten; ceftizoxime; ceftriaxone; cefepime; ceftobiprole; glycopeptide; teicoplanin; vancomycin; macrolides; azithromycin; clarithromycin;
  • dirithromycin erythromicin; roxithromycin; troleandomycin; telithromycin; spectinomycin; monobactam; aztreonam; penicillins; amoxicillin; ampicillin; azlocillin; carbenicillin;
  • cloxacillin cloxacillin; dicloxacillin; flucloxacillin; mezlocillin; meticillin; nafcillin; oxacillin; penicillin, piperacillin, ticarcillin; bacitracin; colistin; polymyxin B; quinolone; ciprofloxacin; enoxacin; gatifloxacin; levofloxacin; lomefloxacin; moxifloxacin; norfloxacin; ofloxacin; trovafloxacin; sulfonamide; mafenide; prontosil (archaic); sulfacetamide; sulfamethizole; sufanilimide (archaic); sulfasalazine; sulfisoxazole; trimethoprim; trimethoprim-sulfamethoxazole (co- trimoxazole) (TMP-SMX
  • the anti-cancer agent is chosen from daunorubicin, doxorubicin, paclitaxel, docetaxel, cisplatin, carboplatin, cytarabine, floxuridine, fludarabine, fluorouracil, iproplatin, leuprolide acetate, carfilzomib, and methotrexate.
  • Exemplary antiviral agents may include, but are not limited to
  • thiosemicarbazone metisazone; nucleoside and/or nucleotide; acyclovir; idoxuridine;
  • vidarabine ribavirin; ganciclovir; famciclovir; valaciclovir; cidofovir; penciclovir;
  • valganciclovir brivudine
  • ribavirin cyclic amines
  • rimantadine tromantadine
  • phosphonic acid derivative foscamet
  • fosfonet protease inhibitor
  • saquinavir indinavir
  • nelfinavir amprenavir; lopinavir; fosamprenavir; atazanavir; tipranavir; nucleoside and nucleotide reverse transcriptase inhibitor; zidovudine; didanosine; zalcitabine; stavudine; lamivudine; abacavir; tenofovir disoproxil; adefovir dipivoxil; emtricitabine; entecavir; non- nucleoside reverse transcriptase inhibitor; nevirapine; delavirdine; efavirenz; neuraminidase inhibitor; zanamivir; oseltamivir; moroxydine; inosine pranobex; pleconaril; and enfuvirtide.
  • anti-fungal agents may include but are not limited to allylamine; terbinafine; antimetabolite; flucytosine; azole; fluconazole; itraconazole; ketoconazole; ravuconazole; posaconazole; voriconazole; glucan synthesis inhibitor; caspofungin;
  • micafungin anidulafungin; polyenes; amphotericin B; amphotericin B Colloidal Dispersion (ABCD); and griseofulvin.
  • Exemplary analgesics may include, but are not limited to opiate derivative, codeine, meperidine, methadone, and morphine.
  • the API consists essentially of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin).
  • the first drug e.g., docetaxel
  • the second drug e.g., doxorubicin
  • the molar ratio of the lipid layer components: second drug is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1; and the molar ratio of the lipid layer components:first drug (e.g., docetaxel) is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1.
  • the molar ratio of second drug e.g., sodium bicarbonate
  • first drug e.g., docetaxel
  • first drug is about 10: 1 to 1 : 10, preferably about 5: 1 to 1 :5, and more preferably about 3: 1 to 1 :3.
  • the invention utilizes lipid and aqueous solutions, for example in making liposomes in accordance with the invention. Accordingly, the composition lipid and/or aqueous solutions can affect the final composition of the liposomes.
  • the lipid solution may comprise an organic solvent.
  • the organic solvent may be a water miscible solvent.
  • the water miscible solvent is selected from the group consisting of ethanol, methanol, DMSO and isopropanol.
  • the organic solvent is ethanol.
  • cationic lipid refers to a lipid or a cholesterol derivative that carries a net positive charge at about pH 3-pH 9.
  • anionic lipid refers to a lipid or a cholesterol derivative that carries a net negative charge at about pH 3-pH 9.
  • pegylated lipid refers to a lipid that is conjugated with a polyethylene glycol polymer.
  • neutral lipid refers to the lipid that does not carry net charge at about pH 3-pH 9.
  • the lipid solution may include a mixture of lipids.
  • the mixture of lipids preferably includes cholesterol.
  • the mixture of lipids may also include a cationic lipid.
  • the cationic lipid may be, but is not limited to, N,N-dioleyl-N,N-dimethylammonium chloride ("DODAC”); N-(2,3- dioleyloxy)propyl)-N,N,N-trimethylammonium chloride (“DOTMA”); N-(2,3- dioleyloxy)propyl)-N,N-dimethylammonium chloride (“DODMA”); N,N-distearyl-N,N- dimethylammonium bromide (“DDAB”); N-(2,3-dioleoyloxy)propyl)-N,N,N- trimethylammonium chloride (“DOTAP”); N-(2,3-dioleoyloxy)propyl)-N,N- dimethylammonium chloride (“DODAP”); 3-(N-(N',N'- dimethylaminoethane)
  • the mixture of lipids may include an anionic lipid.
  • the anionic lipid may be but is not limited to diacylglycerol phophatidic acid ( 1 ,2-distearoyl-sn- glycero-3-phosphate (DSPA); l,2-dipalmitoyl-sn-glycero-3-phosphate (DPPA); 1,2- dimyristoyl-sn-glycero-3-phosphate (DMPA); 1,2-dilauroyl -sn-glycero-3-phosphate (DLPA); l,2-dioleoyl-sn-glycero-3-phosphate (DOPA)), diacylglycerol phosphoglycerol (l,2-distearoyl-sn-glycero-3-phospho-( -rac-glycerol) (DSPG); 1,2-dipalmitoyl-sn-glycero- 3-phospho-(l'-rac-glycerol)
  • the mixture of lipids may also include a neutral lipid.
  • the neutral lipid may be but is not limited to diacylglycerol phosphocholine (L-a- phosphatidylcholine, hydrogenated (Soy) (HSPC); diacylglycerol phosphocholine (L-a- phosphatidylcholine, (Soy) (Soy PC), l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC); l,2-dipalmitoyl-sn-glycero-3-phosphocholine(DPPC); l,2-dimyristoyl-sn-glycero-3- phosphocholine (DMPC); l,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC); 1,2-dioleoyl- sn-glycero-3-phosphocholine (DOPC), diacylglycerol phosphoethanolamine (1,2- distearoyl- s
  • the mixture of lipids may also include a pegylated lipid.
  • the pegylated lipid may be but is not limited to l,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-2000] (mPEG-2000-DSPE ); 1,2-dioctadecanoyl-sn-glycero- 3-phosphoethanolamine -N-[methoxy(polyethylene glycol)-2000] (mPEG-2000-DOPE ); 1,2- dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (mPEG-2000-DPPE ); 1 ,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-2000] (mPEG-2000-DMPE ); l,2-dilauroyl-s
  • the mixture of lipid may also include a lipid-like molecule or lipidoid.
  • the mixture of lipid may also include a lipid- or cholesterol-conjugated molecule including a protein, or a peptide, or an oligonucleotide.
  • the lipid layer includes one or more of the lipid components disclosed herein.
  • the lipid layer consists essentially of the unsaturated phospholipid and cholesterol.
  • the lipid layer consists essentially of the unsaturated phospholipid, cholesterol, and pegylated phospholipid.
  • the lipid layer comprises: about 20-75%, preferably about 30-60%, (molar) unsaturated phospholipid; about 10-60%, preferably 20-50%, (molar) cholesterol; and about 0-20%, preferably 1-10%, (molar) pegylated phospholipid.
  • doxorubicin is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1; and the molar ratio of the lipid layer components :docetaxel is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1.
  • the molar ratio of doxorubicin :docetaxel is about 10: 1 to 1: 10, preferably about 5: 1 to 1:5, and more preferably about 3: 1 to 1:3.
  • the unsaturated phospholipid comprises a polyunsaturated phospholipid or a monounsaturated phospholipid, preferably a
  • phosphatidylcholine and more preferably and soy phosphatidylcholine or 1 ,2-dioleoyl-sn- glycero-3-phosphatidylcholine (DOPC).
  • DOPC 1 ,2-dioleoyl-sn- glycero-3-phosphatidylcholine
  • the cholesterol comprises a cholesterol derivative, preferably a cationic cholesterol derivative, more preferably an amino cholesterol derivative, and still more preferably dimethylaminoethanecarbamoyl-cholesterol (DC-cholesterol).
  • a cholesterol derivative preferably a cationic cholesterol derivative, more preferably an amino cholesterol derivative, and still more preferably dimethylaminoethanecarbamoyl-cholesterol (DC-cholesterol).
  • DC-cholesterol dimethylaminoethanecarbamoyl-cholesterol
  • the pegylated phospholipid comprises a
  • phosphoethanolamine preferably a l,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and wherein the pegylation is a PEG 500 to PEG 5000, preferably PEG 2000.
  • DSPE disistearoyl-sn-glycero-3-phosphoethanolamine
  • the composition of the lipid layer is tuned to achieve a desired loading of the first drug. Although at least a fraction of the first drug is in the lipid layer, one of ordinary skill will understand that the first drug will have a partition coefficient between the lipid layer and aqueous interior. In some embodiments, essentially all of the first drug will be in the lipid layer.
  • the aqueous solution of the process preferably includes water and a buffer. Buffers may be of but are not limited to phosphate, histidine, HEPES, Tris, acetate, carbonate, and citrate. In various embodiments, the composition of the aqueous solution is tuned to achieve a desired loading (and/or crystallization) of the second drug.
  • the second drug will have a partition coefficient between the lipid layer and aqueous interior. In some embodiments, essentially all of the second drug will be in the aqueous interior.
  • the invention provides a method of making the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, comprising: (i) introducing a lipid solution of an unsaturated phospholipid, cholesterol, a first drug (e.g., docetaxel), and preferably a pegylated phospholipid in ethanol through a first port into a mixing chamber and an aqueous solution through a second port into the mixing chamber, thereby making a plurality of liposomes; and (ii) incubating the plurality of liposomes in a second drug (e.g., doxorubicin) solution.
  • a second drug e.g., doxorubicin
  • the angle between at least one lipid and at one aqueous solution inlet ports is not 180° or a substantially similar angle.
  • at least one stream of lipid solution and at one stream of aqueous solution collide at an angle less than about 180 °.
  • the method does not include a T-connector.
  • the angle between at least one lipid and at one aqueous solution inlet ports is about 120° or less, e.g., 115° or less, 100° or less, 90° or less, 80° or less, 72° or less, 60° or less, 45° or less, 30° or less, 18° or less,
  • the aqueous solution in step ii) is introduced via at least two inlet ports, e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more. In some embodiments, the aqueous solution in step ii) is introduced via at least 3 but no more than 11 inlet ports, e.g., at least 3 but not more than 7, at least 3 but no more than 5, at least 4 but no more than 11 , at least 5 but no more than 11 , at least 6 but no more than 11. [0095] In some embodiments, at least two (e.g., 3, 4, 5, 6, 7, etc.) aqueous inlet ports and at least one (e.g., 2, 3, 4, 5, etc.) lipid solution inlet port are in the same plane.
  • At least one (e.g., 2) outlet port is substantially perpendicular to the plane of inlet ports. In other embodiments, at least one (e.g., 2, 3, 4, 5, etc.) outlet port is substantially not perpendicular to the plane of inlet ports.
  • At least two (e.g., 3, 4, 5, 6, 7, etc.) aqueous solution inlet ports and at least one (e.g., 2, 3, 4, 5, etc.) lipid solution inlet port are not in the same plane.
  • the lipid solution may be made from the stock solutions of individual lipids that are mixed together. Lipids are preferably dissolved in an organic solvent to make a lipid solution.
  • the organic solvent used for making the lipid solution may be miscible with water.
  • the solvent may ethanol, methanol, DMSO, propanol, DMF, THF, acetone, dioxane, ethylene glycol, polyethylene glycol and isopropanol. More preferably, the solvent is polyethylene glycol, isopropanol, and ethanol.
  • the solvent includes less than 10% water.
  • the lipid solution may be made from a mixture of lipids, thereupon dissolving the mixture in an organic solvent.
  • the concentration of the total lipids in the solution may be in the range from about 1 mg/niL to about 200 mg/mL, e.g., from about 1 mg/mL to about 100 mg/mL. More preferably, the concentration of the total lipids in the solution may be in the range from about 5 mg/mL to about 100 mg/mL or form about 10 mg/mL to 100 mg/mL.
  • the organic solvent is ethanol at a
  • concentration of about 70% or more e.g., 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 100%.
  • a water-insoluble API may be dissolved in the lipid solution.
  • concentration of the API in the lipid solution will depend on the efficacy of the agent and may easily be determined by one of ordinary skill in the art.
  • the lipid/ API ratio will determined by the encapsulation power of the liposome to the API.
  • a water-soluble API component may be dissolved in a first aqueous solution (S I).
  • the pH and salinity of the solution may be optimized to accommodate the requirements for the interaction between the API component and the lipids to form liposome. These conditions may be readily determined by one of ordinary skill in the art. Samples are provided in the Examples below.
  • an aqueous solution that lacks an API referred to as (S2), may be similar to a solution having the agent. Alternatively, S I and S2 may be different.
  • the lipid solution and the aqueous solution(s) preferably enter the manifold from different ports, each with a flow rate of from about 1 mL/min to about 6000 mL/min.
  • the flow rates may be from about 5 mL/min to about 1000 mL/min. More preferably, the rates may be from about 20 mL/min to about 600 mL/min.
  • the flow rates are adjusted based on the size of inlet ports to obtain the desired liposome size, morphology, PDI, and manufacturing scales.
  • the lipid solution and/or the aqueous solution is introduced via port size of 0.1-5.0 mm at a flow rate about 1 mL/min to about 2,500 mL/min.
  • the flow velocity of the lipid solution and/or the aqueous solution is from about 0.02m s to about 40 m/s, e.g., from 0.1 m/s to 30 m/s, from 0.2 m/s to 20m/s.
  • the flow velocity is adjusted based on the size of inlet ports to obtain the desired liposome size, morphology, PDI, and manufacturing scale.
  • the lipids are believed to instantaneously assemble into liposome particles.
  • the drug API When carried by the lipid solution or by aqueous solution, it may be encapsulated in the liposome by either lipophilic or electrostatic interaction, or both, between the API and the lipids.
  • the present invention also provides a method of producing liposome that do not contain an API (so-called "empty" liposome).
  • the API is absent from both the lipid solution and the aqueous solution that are mixed in the manifold.
  • the API may be loaded into the liposomes by the process of diffusion or another process.
  • doxorubicin may be loaded into the liposome with a pH gradient. See US Patent application No. 10/ 019,200, PCT Publication No. WO 2001/005373, US Patents Nos.
  • the API is mixed with a liposome solution to upload the API into the liposome by diffusion.
  • the API is dissolved in an aqueous solution, and the solution is mixed with the empty liposome.
  • the API may be readily soluble in the solution of empty liposome, and therefore, the API may be directly mixed with the solution of the empty liposome.
  • the volume ratio of the solution of the API to the empty liposome solution of the API is preferably in the range from about 1:50 to about 1 : 1. A lower volume of the solution is preferred because it avoids a significant dilution to the final liposome solution.
  • the drug encapsulation efficiency is preferably greater than 70%. More preferably the efficiency is greater than 80%. Most preferably, the efficiency is greater than 90%.
  • Tangent flow filtration may be used to concentrate the liposome solution.
  • Residual organic solvent in the liposome solution may be removed by a buffer change.
  • the buffer change is performed by tangent flow filtration.
  • the buffer change may be performed by dialysis.
  • the liposome solutions can be sterilized, for example, by passing the solution through a 0.22 micron sterile filter.
  • the Z-average particle size of the liposomes is about 10-200 nm, preferably about 15- 150 nm, and more preferably about 20-120 nm.
  • more than 70% of API is encapsulated in the liposomes. More preferably, more than 80% of API is encapsulated in the liposomes, most preferably, more than 90% of API is encapsulated in the liposomes.
  • liposomes can be unilamellar.
  • the liposomes can be of multilamellar, or of inverted hexagonal or cubic morphology, or as lipid discs, or hollow liposomes.
  • the mean particle size of the liposomes is from about 10 nm to about 2,000 nm, preferably less than 300 nm, more preferably, the mean particle size may be about 10 to 300 nm or about 20 to about 300 nm. Most preferably, the mean particle size is about 20 to 120 nm.
  • the liposomes have a polydispersity index from about 0.005 to about 0.8, e.g., 0.005 to about 0.5, 0.01 to about 0.5, 0.01 to about 0.4, 0.01 to about 0.2.
  • the pharmaceutical composition is for use as a medicament.
  • the pharmaceutical composition is for use as a cancer therapeutic.
  • the pharmaceutical composition can include one or more antibiotic, antivirus, anti-diabetes, anti-hypertension, anti-fungal, or analgesic.
  • the plurality of liposomes are comprised in an injectable formulation, for example, by subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection.
  • injectable formulations can be aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or
  • the injectable formulation can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the liposomes can be in a dried or powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the invention provides a method comprising administering the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
  • the invention also provides a method of treating a subject comprising administering an effective amount of the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
  • the invention provides methods for treating cancer cells and/or tissue, including cancer cells and/or tissue in a human subject.
  • Cancer can be caused by malignant tumors formed by an abnormal growth of cells and tissue leading to organ failure.
  • Solid tumors can be neoplasms (new growth of cells) or lesions (damage of anatomic structures or disturbance of physiological functions) formed by an abnormal growth of body tissue cells other than blood, bone marrow or lymphatic cells.
  • a solid tumor consists of an abnormal mass of cells which may stem from different tissue types such as liver, colon, breast, or lung, and which initially grows in the organ of its cellular origin. However, such cancers may spread to other organs through metastatic tumor growth in advanced stages of the disease.
  • the subject being treated may have been diagnosed with cancer.
  • the subject may have locally advanced, unresectable, or metastatic cancer and/or may have failed a prior first-line therapy.
  • the cancer is liver cancer (e.g., hepatocellular carcinoma, HCC).
  • the liver cancer e.g., HCC
  • the liver cancer e.g., HCC
  • Liver cancer can include a liver tumor resulting from the metastasis of a non-liver cancer, to the liver.
  • the liver cancer (e.g., HCC) can be resectable or unresectable.
  • the liver cancer can comprise a single tumor, multiple tumors, or a poorly defined tumor with an infiltrative growth pattern (into portal veins or hepatic veins).
  • the liver cancer e.g., HCC
  • the liver cancer can comprise a fibrolamellar, pseudoglandular (adenoid), pleomorphic (giant cell), or clear cell pattern.
  • the liver cancer e.g., HCC
  • the liver cancer (e.g., HCC) can comprise a poorly differentiated form, and malignant epithelial cells are discohesive, pleomorphic, anaplastic, and/or giant.
  • the liver cancer (e.g., HCC) is associated with hepatitis B, hepatitis C, cirrhosis, or type 2 diabetes.
  • the cancer is a lung cancer, preferably non-small cell lung cancer (NSCLC); colon cancer; breast cancer; or liver cancer, preferably hepatocellular carcinoma (HCC).
  • NSCLC non-small cell lung cancer
  • HCC hepatocellular carcinoma
  • the docetaxel can be in a concentration of 10, 20, 30, 40, 50, 75, 80, 100, 125, 150, or 160 mg/mL.
  • a dose can be about 10 mg/m 2 to 150 mg/m 2 (e.g., 10, 20, 25, 30, 40, 50, 60, 70, 75, 80, 90, 100, 110, 120, 125, 130, 140, or 150 mg/m 2 ).
  • a dose can be 75 mg/m 2 .
  • a dose can be administered every 3 weeks for 1, 2, 3, 5, 5, or 6 cycles.
  • dosing guidelines for docetaxel are known in the art, and can be adapted based upon factors including, but not limited to the cancer type, the cancer stage, the dosing regimen, the dose of doxorubicin, and/or the efficacy of the pharmaceutical formulations of the invention.
  • the doxorubicin can be in a concentration of 0.1, 0.5, 1, 1.5, 2, 3, 4, or 5 mg/mL.
  • a dose can be about 1 mg/m 2 to 100 mg/m 2 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 25, 30, 40, 50, 60, 70, 75, 80, 90, or 100 mg/m 2 ).
  • a dose can be 30 mg/m 2 .
  • a dose can be administered every 3 weeks for 1, 2, 3, 5, 5, or 6 cycles.
  • dosing guidelines for docetaxel are known in the art, and can be adapted based upon factors including, but not limited to the cancer type, the cancer stage, the dosing regimen, the dose of doxorubicin, and/or the efficacy of the pharmaceutical formulations of the invention.
  • CPT307 comprises of a nonsaturated lipid l,2-Dioleoyl-sn-glycero-3- Phosphatidylcholine (DOPC), cholesterol, and l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[methoxy9polyethyleneglycol)-2000] (mPEG2000-DSPE). It was found that compared the saturated lipid, the nonsaturated lipid has a greater capacity to encapsulate docetaxel.
  • DOPC nonsaturated lipid l,2-Dioleoyl-sn-glycero-3- Phosphatidylcholine
  • mPEG2000-DSPE l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[methoxy9polyethyleneglycol)-2000
  • Liposomal formulation CPT307B was prepared by first dissolving 2100 mg of DOPC, 280 mg of cholesterol, 700 mg of mPEG2000-DSPE, and 175 mg of docetaxel (DOCE) in 70 mL of anhydrous ethanol.
  • the composition (% molar) of the CPT307B lipid solution is illustrated in Table 1.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Twenty milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing.
  • the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump.
  • the liposome solution exited through an outlet port and was collected in a glass vial.
  • the liposome was concentrated by tangent flow filtration.
  • the buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration.
  • the formulation was then sterilized by filtration through a 0.22 ⁇ filter.
  • the Z-average particle size was 32.9 nm.
  • CPT307C was prepared by loading doxorubicin (DXR) into CPT307B .
  • DXR doxorubicin
  • Fourteen milliliters of CPT307B containing 36 mg/mL of DOCE was mixed with 24 mg of DXR that had been pre-dis solved in the histidine/sucrose buffer, and incubated at 42 °C for 3 hours.
  • the DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter.
  • the composition (% molar) of the CPT307C lipid solution is illustrated in the Table 1, 99.6% of DXR was encapsulated.
  • the molar ratio of DOCE:DXR was 1:1.
  • CPT308C contains a polyunsaturated lipid L-a-phosphatidylcholine (Soy PC) that has a high capacity to encapsulate DOCE.
  • Soy PC polyunsaturated lipid L-a-phosphatidylcholine
  • Two milliliters of lipids/DOCE solution was prepared by dissolving 30 mg of Soy PC, 10 mg of cholesterol, 10 mg of mPEG2000-DSPE, and 6 mg of DOCE in anhydrous ethanol.
  • the composition (% molar) of the liposomal formulation CPT308C lipid solution is illustrated in Table 2.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used.
  • the DOCE loaded liposome was then mixed with DXR that had been pre- dissolved in the histidine/sucrose buffer at a doxorubicin/lipid ratio (w/w) of 1:10, and incubated at 42 °C for 2 hours, 97% of DXR was encapsulated.
  • the DOCE and DXR dual- loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter.
  • the Z-average particle size of the dual-loaded liposome was 38.2 nm for CPT308C.
  • the value represents the molar % of each component vs. total lipids.
  • Example 3 Preparation of Liposomal formulation CPT309C
  • CPT309C contains the polyunsaturated lipid Soy PC at a higher molar ratio than CPT308C in Example 2 and thus showed a greater capacity to encapsulate DEOCE.
  • Two milliliters of lipids DOCE solution was prepared by dissolving 30 mg of L-a- phosphatidylcholine (Soy PC), 4 mg of cholesterol, 10 mg of mPEG2000-DSPE, and 6 mg of DOCE in anhydrous ethanol.
  • the composition (% molar) of the liposomal formulation CPT309C lipid solution is illustrated in Table 3.
  • the DOCE loaded liposome was then mixed with DXR that had been pre- dissolved in the histidine/sucrose buffer at a doxorubicin/lipid ratio (w/w) of 1:10, and incubated at 42 °C for 2 hours, 98.8% of DXR was encapsulated.
  • the DOCE and DXR dual- loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter.
  • the Z-average particle size of the dual-loaded liposome was 38.6 nm for CPT309C.
  • the value represents the molar % of each component vs. total lipids.
  • CPT311C is a cationic liposome as it contains a cationic derivative of cholesterol (DC-cholesterol). It was found that cationic lipids enhances liver- targeting delivery and anti-tumor efficacy of the therapeutic agent in the liposome.
  • DC-cholesterol a cationic derivative of cholesterol
  • lipids/DOCE solution liposomal formulation CPT311B
  • DOPC liposomal formulation CPT311B
  • DC-Cholesterol 3 ⁇ -[ ⁇ -( ⁇ ⁇ '- dimethylaminoethane)-carbamoyl]cholesterol hydrochloride
  • mPEG2000-DSPE 5 mg/mL of mPEG2000-DSPE
  • DOCE 2.0 mg/mL of DOCE.
  • the composition (% molar) of the CPT31 IB lipid solution is illustrated in Table 4.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used.
  • Liposomal formulation CPT311C was prepared by loading doxorubicin (DXR) into CPT31 IB. Two milliliters of CPT31 IB was mixed with 0.5 mg of DXR that had been pre-dissolved in the histidine/sucrose buffer at 10 mg/mL, and incubated at 42 °C for 2 hours, 94.5% of DXR was encapsulated. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter. The composition (% molar) of the CPT311C is illustrated in Table 4. The Z-average particle size of the dual-loaded liposome was 34.9 nm for CPT311C.
  • the value represents the molar % of each component vs. total ipids.
  • the cationic surface charge of liposome promotes liposome delivery to the liver.
  • the alternation of the molar ratio of the cationic lipid, for example, DC- cholesterol in the liposome controls the liver delivery of liposome and the clearance rate from the blood.
  • the following examples comprise of DOCE, cholesterol, DC-cholesterol, and mPEG2000-DSPE with increased molar ratio of DC-cholesterol (from 3.9% increased to 15.4%) and deceased cholesterol molar ratio (from 34.4% reduced to 17.8%) while the molar ratio of DOPC and mPEG2000-DSPE remain unchanged or a minorly changed.
  • lipids/DOCE solution Two and one half (2.5) milliliters of lipids/DOCE solution was prepared by dissolving 37.5 mg DOPC, 11.3 mg cholesterol, 1.4 mg of 3 ⁇ -[ ⁇ -( ⁇ ', ⁇ '- dimethylaminoethane)-carbamoyl]cholesterol hydrochloride (DC-Cholesterol), 12.5 mg mPEG2000-DSPE , and 4 mg DOCE in 2.5 mL anhydrous ethanol. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Two and one half (2.5) milliliter of each of the above four solutions was loaded into a 20 mL syringe.
  • Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass vial. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by dialysis.
  • the DOCE loaded liposome was then mixed with DXR that had been pre- dissolved in the histidine/sucrose buffer at a doxorubicin/lipid ratio (w/w) of 1:16, and incubated at 42 °C for 2 hours, 96.9% of DXR was encapsulated.
  • the DOCE and DXR dual- loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter.
  • the Z-average particle size of the dual-loaded CPT315C was 35.1 nm.
  • lipids/DOCE solution was prepared by dissolving 600 mg of DOPC, 140 mg of cholesterol, 84 mg of DC-Cholesterol, 200 mg of mPEG2000- DSPE, and 50 mg of DOCE in anhydrous ethanol.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Twenty milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump.
  • the buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration.
  • the formulation was then sterilized by filtration through a 0.22 ⁇ filter.
  • Liposomal formulation CPT317B loaded with DOCE was obtained.
  • Liposomal formulation CPT317C was prepared by loading doxorubicin (DXR) into CPT317B. Five milliliters of CPT317B was mixed with 5.2 mg of DXR, and incubated at 42 °C for 3 hours. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter. The composition (% molar) of the CPT317C lipid solution is illustrated in Table 6. 99.8% of DXR was encapsulated.
  • DXR doxorubicin
  • the value represents the molar % of each component vs. total lipids.
  • Example 7 Preparation of Liposomal formulation CPT319C
  • the lipids/DOCE solution was prepared by dissolving 1848 mg of DOPC, 303 mg of cholesterol, 423 mg of DC-Cholesterol, 605 mg of mPEG2000-DSPE, and 154 mg of DOCE in 61.5 mL of anhydrous ethanol.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Twenty milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing.
  • the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump.
  • the liposome solution exited through an outlet port and was collected in a glass bottle and then was concentrated by tangent flow filtration.
  • the buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration.
  • the formulation was then sterilized by filtration through a 0.22 ⁇ filter to obtain liposomal formulation CPT319B loaded with DOCE.
  • Liposomal formulation CPT319C was prepared by loading doxorubicin (DXR) into CPT319B. In a glass bottle 31.4 mg of DXR was dissolved in 30mL of
  • CPT319B The mixture was incubated at 42 °C for 4 hours, 99.8% of doxorubicin was encapsulated. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter.
  • the final composition (% molar) of the CPT317C lipid solution is illustrated in Table 7.
  • the Z-average particle size was 40.7 nm and the molar ratio of DOCE:DXR was 1: 1 for CPT319C.
  • the Cryo-TEM images of CPT319C are shown in FIG. 1. The DXR crystals formed inside the liposome can be seen from these images.
  • the negative stained TEM image of CPT319C is shown in FIG. 2, which indicates particle size and homogeneity.
  • CPT323C was prepared in the absence of pegylated lipid, thus the pegylated lipid is optional to the formulations.
  • the lipids/DOCE solution was prepared by dissolving 300 mg of DOPC, 50 mg of cholesterol, 70 mg of DC-Cholesterol, and 25 mg of DOCE in 10 mL of anhydrous ethanol.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used.
  • Ten milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing.
  • the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump.
  • the liposome solution exited through an outlet port and was collected in a glass bottle and was then concentrated by tangent flow filtration.
  • the buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration.
  • the formulation was then sterilized by filtration through a 0.22 ⁇ filter.
  • CPT324C contains a polyunsaturated lipid- Soy PC and a cationic lipid DOTAP other than DC-cholesterol .
  • the lipids/DOCE solution was prepared by dissolving 60 mg of Soy PC, 40 mg of cholesterol, 60 mg of l,2-dioleoyl-3-trimethylammonium-propane (chloride salt) (DOTAP), 40 mg of mPEG2000-DSPE, and 25 mg of DOCE in 10 mL of anhydrous ethanol.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used.
  • DOCE loaded liposome Three milliliters of the DOCE loaded liposome was then mixed with 0.95 mg of DXR that had been pre-dissolved in the histidine/sucrose buffer at 7 mg/mL, and incubated at 42 °C for 6 hours.
  • the DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 ⁇ filter to obtain liposomal formulation CPT324C.
  • the composition (% molar) of the CPT324C lipid solution is illustrated in Table 9.
  • the Z-average particle size of the dual-loaded liposome was 56 nm for CPT324C.
  • the value represents the molar % of each component vs. total lipids.
  • CPT313C was prepared in the presence of DC-cholesterol and the absence of cholesterol.
  • the lipids/DOCE solution was prepared by dissolving 33 mg of Soy PC, 20.5 mg of DC-Cholesterol, 11 mg of mPEG2000-DSPE, and 4.4 mg of DOCE in 2.2 mL of anhydrous ethanol.
  • three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. 2.2 milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing.
  • the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump.
  • the liposome solution exited through an outlet port and was collected in a glass vial.
  • the buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by dialysis.
  • the formulation was then sterilized by filtration through a 0.22 ⁇ filter.
  • the value represents the molar % of each component vs. total lipids.
  • Example 11 Dual-loaded liposome CPT319C augments efficacy against non-small cell lung cancer (NSCLC)
  • mice Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously on the right flank with NSCLC cell line A549 tumor cells (1 x 10 7 cells/mouse) in 0.1 mL phosphate buffered saline (PBS) buffer for tumor development.
  • PBS phosphate buffered saline
  • FIG. 3 shows that dual-loaded Liposome CPT319C enhances the antitumor activity of the formulation in NSCLC Xenograft mouse model. All of the liposomal formulations were more efficacious than the non-liposomal combination of DOCE/DXR. In addition, the dual-loaded liposome, CPT319C was the most efficacious formulation in this example. Compared to the PBS control group, CPT319C reduced 76% of the tumor weight that was significantly more efficacious than the 51% of DOCE liposome CPT319B, 27% of DXR liposome CPT319A, and 17% of the non-liposomal combination of DOCE/DXR.
  • Example 12 Dual-loaded liposome CPT307C augments efficacy against non-small cell lung cancer (NSCLC)
  • mice Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with NSCLC cell line A549 tumor cells (1 x 10 7 cells/mouse) in 0.1 mL PBS buffer for tumor development.
  • FIG. 4 shows that dual-loaded liposome CPT307C augments efficacy against non-small cell lung cancer (NSCLC). All of the liposomal formulations were more efficacious than the non-liposomal combination of DOCE/DXR. In addition, the dual- loaded liposome, CPT307C was the most efficacious in this example. Compared to the PBS control group, the dual-loaded liposome CPT307C 57% of the tumor weight that is significant more efficacious than the 44% of DOCE liposome CPT307B, 11% of DXR liposome CPT307A, and 17% of the non-liposomal combination of DOCE/DXR. [00175] Example 13: Antitumor activity of dual-loaded liposome CPT319C against human colon cancer in xenograft mouse model
  • mice Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human colon cancer cell line HCT- 116 tumor cells (5 x 10 6 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 9 following tumor cell inoculation (tumor size was approximately 141 mm ), treatments were started with formulations of CPT319C at 3 different doses: 5 mg/kg doxorubicin/7.5 mg kg docetaxel, 2.5 mg/kg doxorubicin/3.75 mg/kg docetaxel, or 1.25 mg kg doxorubicin/1.875 mg/kg docetaxel by intravenous (IV) injection through the tail vein.
  • IV intravenous
  • Example 14 Antitumor activity of dual-loaded liposome CPT307C against human colon cancer in xenograft mouse model
  • mice Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human colon cancer cell line HCT- 116 tumor cells (5 x 10 6 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 9 following tumor cell inoculation (tumor size was approximately 141 mm 3 ), treatments were started with formulations of CPT307C at 3 different doses: 5 mg/kg doxorubicin/7.5 mg kg docetaxel, 2.5 mg/kg doxorubicin/3.75 mg/kg docetaxel, or 1.25 mg/kg doxorubicin/1.875 mg kg docetaxel by intravenous (IV) injection through the tail vein.
  • IV intravenous
  • Example 15 Antitumor activity of dual-loaded liposome CPT319C and CPT307C against human breast cancer in xenograft mouse model
  • mice Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human breast cancer cell line MD A- MB -231 tumor cells (7 x 10 6 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 7 after tumor cell inoculation (tumor size was approximately 174 mm ), treatments were started with formulations of CPT319C or CPT307C at 5 mg/kg doxorubicin/7.5 mg/kg docetaxel by intravenous (IV) injection through the tail vein. Two additional treatments were made on Day 14 and Day 21. The study was terminated on Day 31. The tumor growth curves were shown in FIG. 7.
  • FIG. 7 shows CPT319C and CPT307C antitumor activity in a human breast cancer xenograft model. Compared to the PBS control group, the tumor size was reduced 38% and 32% in the group treated by CPT319C and CPT307C, respectively.
  • Example 16 Antitumor activity of dual-loaded liposome CPT319C against human primary hepatocellular carcinoma in xenograft mouse model
  • mice Female Balb/c nude mice ranging from 6-8 weeks were split up into groups of three. Each mouse was inoculated subcutaneously at the right flank with fragments of human primary hepatocellular carcinoma tumor cells (P3 WP HCC) for tumor development. On Day 32 after tumor inoculation (tumor size was approximately 143 mm 3 ), treatments were started with a formulation of CPT319C at 5 mg/kg doxorubicin/7.5 mg/kg docetaxel by intravenous (IV) injection through the tail vein. Two additional treatments were made on Day 39 and Day 46. The study was terminated on Day 63. The tumor growth curves are shown in FIG. 8.
  • P3 WP HCC human primary hepatocellular carcinoma tumor cells
  • FIG. 8 shows CPT319C antitumor activity in a human primary HCC xenograft model. Tumor growth was almost completely inhibited by CPT319C.
  • CPT319C inhibited 88% of tumor growth when the study was terminated on Day 63 compared to the vehicle control group.
  • Example 17 Cationic lipid DC-Cholesterol enhances the antitumor activity of liposomes against NSCLC
  • mice Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with NSCLC cell line A549 tumor cells (1 x 10 7 cells/mouse) in 0.1 mL PBS buffer for tumor development.
  • FIG. 9 shows that a cationic lipid DC-cholesterol can enhance the antitumor activity of the liposomes against NSCLC.
  • the tumor inhibition rank order (from low to high) was:
  • Example 18 Cationic lipid DC-Cholesterol enhances the antitumor activity of liposomes against colon cancer
  • mice Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human colon cancer cell line HCT- 116 tumor cells (5 x 10 6 cells/mouse) in 0.1 mL PBS buffer for tumor development. On day 9 following tumor cell inoculation (tumor size was approximately 141 mm ), treatments were started with formulations of CPT319C (containing DC -Cholesterol) or CPT307C (without DC- Cholesterol) at 5 mg/kg DXR 7.5 mg/kg DOCE by intravenous (IV) injection through the tail vein. Two additional treatments were made on Day 16 and Day 23. The study was terminated on Day 37. The tumor growth curves and tumor weight inhibition percentages (TW Inh%) on Day 37 of the formulations compared to the vehicle control group are shown in FIG. 10.
  • FIG. 10 shows that a cationic lipid DC-cholesterol can enhance the antitumor activity of the liposomes against colon cancer.
  • CPT319C (with DC- Cholesterol) was more efficacious than CPT307C (without DC-Cholesterol), indicating that the incorporation of the cationic lipid DC-cholesterol enhances the anti-tumor efficiency of the liposomal formulations.
  • Example 19 Liposome improves pharmacokinetics (PK) and cationic lipid DC-Cholesterol increases the half-life (ti /2 ) of DXR
  • mice Male CD-I mice ranging from 20-25 g body weight were split up into groups of three. Each mouse was administered with a single dose of CPT319C or CPT307C at 5 mg/kg DXR/7.5 mg kg DOCE by intravenous (IV) injection through the tail vein. A non- liposomal combination of DXR/DOCE was used as the control. Blood samples were collected at 0.167, 1, 3, 8, 24, and 48h after the injection. DXR plasma concentration was determined by liquid chromatography-tandem mass spectrometry. The plasma concentration curves of DXR are shown in FIG. 11.
  • FIG. 11 shows that a liposome can improve pharmacokinetics and that a cationic lipid DC-cholesterol increases the half-life (ti/ 2 ) of DXR.
  • BLOQ Below Limit of Quantitation.
  • the ti /2 and area under the plasma concentration time curve (AUC) are provided in the table below.
  • the non-liposomal DXR was cleared quickly from the blood and resulted in a very low AUC (688 h x ng/mL), whereas CPT319C and CPT307C increased AUC by 143 and 204 fold, respectively.
  • CPT319C exhibited a 5.9h ti /2 compared to the lh ti/2 of CPT307C, indicating that the cationic lipid DC-Cholesterol in CPT319C improves PK of the formulation by increasing circulation time in the blood.

Abstract

A pharmaceutical composition can include a plurality of liposomes comprising docetaxel and doxorubicin. In various embodiments, a liposome can include (i) an active pharmaceutical ingredient (API) comprising docetaxel and doxorubicin; (ii) a lipid layer comprising an unsaturated phospholipid, a cholesterol, a cationic lipid, and preferably a pegylated phospholipid; and (iii) an aqueous interior, wherein the docetaxel is in the lipid layer and the doxorubicin is crystallized in the aqueous interior. The liposomes can be used to treat a subject, for example, a human subject having cancer. The cancer can be, for example, a lung cancer, preferably non-small cell lung cancer (NSCLC), colon cancer, breast cancer, or liver cancer.

Description

DUAL LOADED LIPOSOMAL PHARMACEUTICAL FORMULATIONS
FIELD OF THE INVENTION
[0001] The present invention generally relates to liposomal pharmaceutical formulations and, in various embodiments, more specifically to liposomal pharmaceutical formulations including an active pharmaceutical ingredient with two components (e.g., a combination of docetaxel and doxorubicin).
BACKGROUND
[0002] Liposome technology has been utilized for drug delivery in clinical therapy and scientific research. To date, a handful of liposomal pharmaceutical formulations have been approved by the US Food and Drug Administration ("FDA"), and a number of new liposomal formulations are in clinical trials. However, the field of liposomal formulation is still evolving and each active pharmaceutical ingredient ("API") presents unique challenges.
[0003] One area where liposomal formulations can be applied is in cancer APIs. For example, liposomal formulations of doxorubicin are presently available under the trade names Doxil® and Myocet®. Doxil® is a pegylated (polyethylene glycol coated) liposome- encapsulated form of doxorubicin formerly made by Ben Venue Laboratories in the United States for Janssen Products, LP, a subsidiary of Johnson & Johnson. Myocet® is a non- pegylated liposomal doxorubicin made by Enzon Pharmaceuticals for Cephalon in Europe and for Sopherion Therapeutics in the United States and Canada. Myocet® is approved in Europe and Canada for treatment of metastatic breast cancer in combination with
cyclophosphamide, but is not yet approved by the FDA for use in the United States.
[0004] Despite the handful of approved liposomal pharmaceutical formulations, the field is still limited by the unique challenges and unpredictability of each different API, as well as the currently available methods of making liposomal formulations, which present difficult problems associated with scalability, low reproducibility, and product heterogeneity. There exists a need for improved liposomal formulations for use in drug delivery.
SUMMARY OF THE INVENTION
[0005] In various aspects and embodiments, the invention provides a pharmaceutical composition including a plurality of liposomes comprising a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin). In various embodiments, a liposome can include (i) an active pharmaceutical ingredient (API) comprising a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin); (ii) a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid; and (iii) an aqueous interior, wherein the first drug (e.g., docetaxel) is in the lipid layer and the second drug (e.g., doxorubicin) is crystallized in the aqueous interior. The liposomes can be used to treat a subject, for example, a human subject having cancer. The cancer can be, for example, a lung cancer, preferably non- small cell lung cancer (NSCLC); colon cancer; breast cancer; or liver cancer, preferably hepatocellular carcinoma (HCC).
[0006] The invention can provide for increased efficacy and/or decreased toxicity, for example relative to (i) other pharmaceutical compositions where one or both of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin) are not in a liposomal formulation and/or (ii) other liposomal formulations. The invention can provide for targeted delivery, for example to the liver or avoiding the liver. The invention can mitigate undesired side effects, for example by providing for increased drug loading, thereby reducing the amount of liposomes needed to deliver a quantity of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin).
[0007] The invention provides a liposome comprising: (i) an active pharmaceutical ingredient (API) comprising docetaxel and doxorubicin; (ii) a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid; and (iii) an aqueous interior, wherein the docetaxel is in the lipid layer and the doxorubicin is crystallized in the aqueous interior.
[0008] The invention also provides a pharmaceutical composition comprising a plurality of liposomes according to any of the aspects or embodiments disclosed herein.
[0009] The invention also provides a method comprising administering the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
[0010] The invention also provides a method of treating a subject comprising administering an effective amount of the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
[0011] The invention also provides a method of making the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, comprising: (i) introducing a lipid solution of an unsaturated phospholipid, cholesterol, a first drug (e.g., docetaxel), and preferably a pegylated phospholipid in ethanol through a first or more inlet port of a manifold into a mixing chamber and an aqueous solution through a second or more inlet port of the manifold into the mixing chamber, the liposomes formed exit the mixing chamber through a third or more outlet port of the manifold, thereby making a plurality of liposomes; and (ii) incubating the plurality of liposomes in a second drug (e.g., doxorubicin) solution.
[0012] In various embodiments, the lipid layer consists essentially of the unsaturated phospholipid and cholesterol.
[0013] In various embodiments, the lipid layer consists essentially of the unsaturated phospholipid, cationic lipid, cholesterol, and pegylated phospholipid.
[0014] In various embodiments, the API consists essentially of docetaxel and doxorubicin.
[0015] In various embodiments, the lipid layer comprises: about 20-75%, preferably about 30-60%, (molar) unsaturated phospholipid; about 10-60%, preferably 20-50%, (molar) cholesterol; about 5-75%, preferably about 10-60%, (molar) cationic lipid; and about 0-20%, preferably 1-10%, (molar) pegylated phospholipid.
[0016] In various embodiments, the molar ratio of the lipid layer
components doxorubicin is about 100: 1 to about 2: 1, preferably about 20: 1 to about 5: 1 ; and the molar ratio of the lipid layer components: docetaxel is about 100: 1 to about 2: 1 , preferably about 20: 1 to about 5: 1.
[0017] In various embodiments, the molar ratio of doxorubicin: docetaxel is about 10: 1 to 1: 10, preferably about 5: 1 to 1:5, and more preferably about 2: 1 to 1:2.
[0018] In various embodiments, the unsaturated phospholipid comprises a polyunsaturated phospholipid or a monounsaturated phospholipid, preferably a
phosphatidylcholine, and more preferably and soy phosphatidylcholine or 1 ,2-dioleoyl-sn- glycero-3-phosphatidylcholine (DOPC).
[0019] In various embodiments, the cholesterol comprises a cholesterol derivative, preferably a cationic cholesterol derivative, more preferably an amino cholesterol derivative, and still more preferably dimethylaminoethanecarbamoyl-cholesterol (DC-cholesterol).
[0020] In various embodiments, the pegylated phospholipid comprises a
phosphoethanolamine, preferably a l,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and wherein the pegylation is a PEG 500 to PEG 3000, preferably PEG 2000.
[0021] In various embodiments, the plurality of liposomes are comprised in an intravenous formulation.
[0022] In various embodiments, the Z-average particle size of the liposomes is about 10-200 nm, preferably about 15- 150 nm, and more preferably about 20-120 nm. [0023] In various embodiments, upon intravenous administration to a subject, at least about 10% of the composition is delivered to the liver.
[0024] In various embodiments, the pharmaceutical composition is for use as a medicament.
[0025] In various embodiments, the pharmaceutical composition is for use as a cancer therapeutic.
[0026] In various embodiments, the subject has a cancer. In various embodiments, the cancer is a lung cancer, preferably non-small cell lung cancer (NSCLC); colon cancer; breast cancer; or liver cancer, preferably hepatocellular carcinoma (HCC).
[0027] These and other advantages of the present technology will be apparent when reference is made to the accompanying drawings and the following description.
BRIEF DESCRIPTION OF THE DRAWINGS
[0028] FIG. 1 presents a cryo transmission electron microscopy (TEM) image of liposomal formulation CPT319C.
[0029] FIG. 2 presents a negative stained TEM image of CPT319C.
[0030] FIG. 3 illustrates NSCLC tumor growth curves and tumor weight inhibition percentages (TW inh%) after administration of liposomal (CPT319A, CPT319B, or
CPT319C) or non-liposomal formulations of docetaxel/doxorubicin, compared to a control group.
[0031] FIG. 4 illustrates NSCLC tumor growth curves and tumor weight inhibition percentages (TW inh%) after administration of liposomal (CPT307A, CPT307B, or
CPT307C) or non-liposomal formulations of docetaxel/doxorubicin, compared to the control group.
[0032] FIG. 5 illustrates colon cancer tumor growth curves after administration of three different doses of liposomal formulation (CPT319C), compared to the control group.
[0033] FIG. 6 illustrates colon cancer tumor growth curves after administration of three different doses of liposomal formulation (CPT307C), compared to the control group.
[0034] FIG. 7 illustrates breast cancer tumor growth curves on Day 31 after administration of liposomal formulations (CPT307C or CPT319C), compared to the control group.
[0035] FIG. 8 illustrates hepatocellular carcinoma tumor growth curves after administration of liposomal formulation (CPT319C), compared to the control group. [0036] FIG. 9 illustrates NSCLC tumor growth curves after administration of liposomal formulations (CPT307A-C or CPT319A-C), compared to the control group.
[0037] FIG. 10 illustrates colon cancer tumor growth curves and tumor weight inhibition percentages (TW Inh ) after administration of liposomal formulations (CPT307C or CPT219C), compared to the control group.
[0038] FIG. 11 illustrates plasma concentration curves of doxorubicin after administration of liposomal formulations (CPT319C or CPT307C), compared to non- liposomal formulation of docetaxel/doxorubicin.
[0039] While the invention comprises embodiments in many different forms, there are shown in the drawings and will herein be described in detail several specific embodiments with the understanding that the present disclosure is to be considered as an exemplification of the principles of the technology and is not intended to limit the invention to the embodiments illustrated.
DETAILED DESCRIPTION
[0040] In various aspects and embodiments, the invention provides a pharmaceutical composition including a plurality of liposomes comprising a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin). In various embodiments, a liposome can include (i) an active pharmaceutical ingredient (API) comprising a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin); (ii) a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid; and (iii) an aqueous interior, wherein the first drug (e.g., docetaxel) is in the lipid layer and the second drug (e.g., doxorubicin) is crystallized in the aqueous interior. The liposomes can be used to treat a subject, for example, a human subject having cancer.
[0041] As described and shown in the examples below, the invention can provide for increased efficacy and/or decreased toxicity, for example relative to (i) other pharmaceutical compositions where one or both of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin) are not in a liposomal formulation and/or (ii) other liposomal formulations. The invention can provide for targeted delivery, for example to the liver or avoiding the liver. The invention can mitigate undesired side effects, for example by providing for increased drug loading, thereby reducing the amount of liposomes needed to deliver a quantity of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin). [0042] The various features of such liposomes, as well as pharmaceutical
compositions including the liposomes and methods of using and making the liposomes are discussed, in turn, below.
[0043] Active Pharmaceutical Ingredient (API)
[0044] In various aspects and embodiments, the API comprises a first drug (e.g., docetaxel) and a second drug (e.g., doxorubicin). While docetaxel and doxorubicin are presented as illustrative examples, other embodiments are possible where the first drug is in the lipid layer of the liposome and the second drug is in (e.g., crystallized in) the aqueous interior of the liposome. In various embodiments, the API can comprise two (or more) anticancer agents, an anti-inflammatory agents, an anti-diabetic agents, an anti-fungal agents, and/or antibiotic agents.
[0045] Docetaxel (as generic or under the trade name Taxotere® or Docecad®) is a clinically well-established anti-mitotic chemotherapy medication that works by interfering with cell division. Docetaxel is approved by the FDA for treatment of locally advanced or metastatic breast cancer, head and neck cancer, gastric cancer, hormone-refractory prostate cancer and non small-cell lung cancer. Docetaxel can be used as a single agent or in combination with other chemotherapeutic drugs as indicated depending on specific cancer type and stage.
[0046] Docetaxel is a member of the taxane drug class, which also includes the chemotherapeutic medication paclitaxel. Accordingly, in some embodiments, docetaxel can be substituted for another taxane that can be disposed within the lipid layer of the liposome.
[0047] The optimal dose scheduling of taxanes remains unconfirmed, but most studies find significant mortality benefit following either a three-week or a one- week administration schedule. While some research suggests weekly administration as an optimal schedule, the official docetaxel package insert recommends administration every three weeks. Important toxicities to note include neutropenia, febrile neutropenia and neurosensory disturbances. Such toxicities have been well documented in Phase II and Phase III clinical trials and can be anticipated and subsequently managed.
[0048] In various embodiments, the invention can increase the efficacy of, and/or decrease undesired side effects from, the docetaxel.
[0049] Doxorubicin (trade name Adriamycin®; pegylated liposomal form trade name Doxil®; nonpegylated liposomal form trade name Myocet®), also known as
hydroxydaunorubicin and hydroxydaunomycin, is a drug used in cancer chemotherapy and derived by chemical semisynthesis from a bacterial species. It is an anthracycline antibiotic (note: in this context, this does not mean it is used to treat bacterial infections) closely related to the natural product daunomycin and like all anthracyclines, it is believed to work by intercalating DNA, with the most serious adverse effect being life-threatening heart damage. It is commonly used in the treatment of a wide range of cancers, including hematological malignancies (blood cancers, like leukaemia and lymphoma), many types of carcinoma (solid tumors) and soft tissue sarcomas. It is often used in combination chemotherapy as a component of various chemotherapy regimens. In some embodiments, doxorubicin can be substituted for another anticancer agent that can be disposed within the aqueous interior of the liposome.
[0050] Common adverse effects of doxorubicin include hair loss (seen in most of those treated with the drug), myelosuppression (a compromised ability of the body's bone marrow to produce new blood cells), nausea and vomiting (which are seen in roughly 30-90% of people treated with the drug), oral mucositis, oesophagitis, diarrhea, skin reactions (including hand-foot syndrome) and localized swelling and redness along the vein in which the drug is delivered. Less common, yet serious reactions include hypersensitivity reactions (including anaphylaxis), radiation recall, heart damage and liver dysfunction.
[0051] The drug is administered intravenously, as the hydrochloride salt. It is sold under a number of different brand names, including Adriamycin® PFS, Adriamycin® RDF, or Rubex®. Doxorubicin is photosensitive, and containers are often covered by an aluminum bag and/or brown wax paper to prevent light from affecting it. Doxorubicin is also available in liposome-encapsulated forms as Doxil® (pegylated form), Myocet® (nonpegylated form), and Caelyx®, although these forms must also be given by intravenous injection.
[0052] In various embodiments, the invention can increase the efficacy of and/or decrease undesired side effects from, the doxorubicin.
[0053] In some embodiments, the API may be a polynucleotide (including an oligonucleotide) a protein or a small molecule.
[0054] In one embodiment the API is a polynucleotide. The polynucleotide may be a genomic DNA fragment, cDNA, mRNA, ssRNA, dsRNA, microRNA, siRNA, shRNA, sdRNA, DsiRNA, LNA, and antisense DNA or RNA.
[0055] Alternatively, the API may be a small molecule drug. Preferably, the molecule has a molecular weight from about 1500 g/mole to about 50 g/mole.
[0056] An API can include, for example, two or more of the following: an anticancer agent, an antibiotic agent, an antiviral agent, an anti-fungal agent, or an analgesic. [0057] Exemplary anticancer agents may include but are not limited acivicin, aclarubicin, acodazole, ametantrone, aminoglutethimide, anthramycin, asparaginase, azacitidine, azetepa, bisantrene, bleomycin, busulfan, cactinomycin, calusterone, caracemide, carboplatin, carfilzomib, carmustine, carubicin, chlorambucil, cisplatin, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, dezaguanine, diaziquone, docetaxel, doxorubicin, epipropidine, erlotinib, etoposide, etoprine, floxuridine, fludarabine, fluorouracil, fluorocitabine, hydroxyurea, iproplatin, leuprolide acetate, lomustine, mechlorethamine, megestrol acetate, melengestrol acetate, mercaptopurine, methotrexate, metoprine, mitocromin, mitogillin, mitomycin, mitosper, mitoxantrone, mycophenolic acid, nocodazole, nogalamycin, oxisuran, paclitaxel, peliomycin, pentamustine, porfiromycin, prednimustine, procarbazine hydrochloride, puromycin, pyrazofurin, riboprine, semustine, sparsomycin, spirogermanium, spiromustine, spiroplatin, streptozocin, talisomycin, tegafur, teniposide, teroxirone, thiamiprine, thioguanine, tiazofurin, triciribine phosphate,
triethylenemelamine, trimetrexate, uracil mustard, uredepa, vinblastine, vincristine, vindesine, vinepidine, vinrosidine, vinzolidine, zinostatin and zorubicin.
[0058] Exemplary antibiotic agents may include but are not limited to
aminoglycoside; amikacin; gentamicin; kanamycin; neomycin; netilmicin; steptomycin; tobramycin; ansamycins; geldanamycin; herbimycin; carbacephem; loracarbef;
carbacepenem; ertapenem; doripenem; imipenem/cilastatin; meropenem; cephalosporin; cefadroxil; cefazolin; cefalotin or cefalothin; cefalexin; cefaclor; cefamandole; cefoxitin; cefprozil; cefuroxime; cefixime; cefdinir; cefditoren; cefoperazone; cefotaxime;
cefpodoxime; ceftazidime; ceftibuten; ceftizoxime; ceftriaxone; cefepime; ceftobiprole; glycopeptide; teicoplanin; vancomycin; macrolides; azithromycin; clarithromycin;
dirithromycin; erythromicin; roxithromycin; troleandomycin; telithromycin; spectinomycin; monobactam; aztreonam; penicillins; amoxicillin; ampicillin; azlocillin; carbenicillin;
cloxacillin; dicloxacillin; flucloxacillin; mezlocillin; meticillin; nafcillin; oxacillin; penicillin, piperacillin, ticarcillin; bacitracin; colistin; polymyxin B; quinolone; ciprofloxacin; enoxacin; gatifloxacin; levofloxacin; lomefloxacin; moxifloxacin; norfloxacin; ofloxacin; trovafloxacin; sulfonamide; mafenide; prontosil (archaic); sulfacetamide; sulfamethizole; sufanilimide (archaic); sulfasalazine; sulfisoxazole; trimethoprim; trimethoprim-sulfamethoxazole (co- trimoxazole) (TMP-SMX); tetracycline; demeclocycline; doxycycline; minocycline;
oxytetracycline; tetracycline; arsphenamine; chloramphenicol; clindamycin; lincomycin; ethambutol; fosfomycin; fusidic acid; furazolidone; isoniazid; linezolid; metronidazole; mupirocin; nitrofuantoin; platensimycin; polymyxin, purazinamide; quinupristin/dalfopristin; rifampin or rifampicin; and timidazole.
[0059] In specific embodiments, the anti-cancer agent is chosen from daunorubicin, doxorubicin, paclitaxel, docetaxel, cisplatin, carboplatin, cytarabine, floxuridine, fludarabine, fluorouracil, iproplatin, leuprolide acetate, carfilzomib, and methotrexate.
[0060] Exemplary antiviral agents may include, but are not limited to
thiosemicarbazone; metisazone; nucleoside and/or nucleotide; acyclovir; idoxuridine;
vidarabine; ribavirin; ganciclovir; famciclovir; valaciclovir; cidofovir; penciclovir;
valganciclovir; brivudine; ribavirin, cyclic amines; rimantadine; tromantadine; phosphonic acid derivative; foscamet; fosfonet; protease inhibitor; saquinavir; indinavir; ritonavir;
nelfinavir; amprenavir; lopinavir; fosamprenavir; atazanavir; tipranavir; nucleoside and nucleotide reverse transcriptase inhibitor; zidovudine; didanosine; zalcitabine; stavudine; lamivudine; abacavir; tenofovir disoproxil; adefovir dipivoxil; emtricitabine; entecavir; non- nucleoside reverse transcriptase inhibitor; nevirapine; delavirdine; efavirenz; neuraminidase inhibitor; zanamivir; oseltamivir; moroxydine; inosine pranobex; pleconaril; and enfuvirtide.
[0061] Exemplary anti-fungal agents may include but are not limited to allylamine; terbinafine; antimetabolite; flucytosine; azole; fluconazole; itraconazole; ketoconazole; ravuconazole; posaconazole; voriconazole; glucan synthesis inhibitor; caspofungin;
micafungin; anidulafungin; polyenes; amphotericin B; amphotericin B Colloidal Dispersion (ABCD); and griseofulvin.
[0062] Exemplary analgesics may include, but are not limited to opiate derivative, codeine, meperidine, methadone, and morphine.
[0063] In various embodiments, the API consists essentially of the first drug (e.g., docetaxel) and the second drug (e.g., doxorubicin).
[0064] In various embodiments, the molar ratio of the lipid layer components: second drug (e.g., doxorubicin) is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1; and the molar ratio of the lipid layer components:first drug (e.g., docetaxel) is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1.
[0065] In various embodiments, the molar ratio of second drug (e.g.,
doxorubicin): first drug (e.g., docetaxel) is about 10: 1 to 1 : 10, preferably about 5: 1 to 1 :5, and more preferably about 3: 1 to 1 :3. [0066] The Lipid Layer and Aqueous Solutions
[0067] The invention utilizes lipid and aqueous solutions, for example in making liposomes in accordance with the invention. Accordingly, the composition lipid and/or aqueous solutions can affect the final composition of the liposomes.
[0068] In various embodiments, the lipid solution may comprise an organic solvent. The organic solvent may be a water miscible solvent. Preferably, the water miscible solvent is selected from the group consisting of ethanol, methanol, DMSO and isopropanol. Most preferably, the organic solvent is ethanol.
[0069] As used herein the term of "cationic lipid" refers to a lipid or a cholesterol derivative that carries a net positive charge at about pH 3-pH 9.
[0070] As used herein the term of "anionic lipid" refers to a lipid or a cholesterol derivative that carries a net negative charge at about pH 3-pH 9.
[0071] As used herein the term "pegylated lipid" refers to a lipid that is conjugated with a polyethylene glycol polymer.
[0072] As used herein the term "neutral lipid" refers to the lipid that does not carry net charge at about pH 3-pH 9.
[0073] The lipid solution may include a mixture of lipids. The mixture of lipids preferably includes cholesterol.
[0074] The mixture of lipids may also include a cationic lipid. The cationic lipid may be, but is not limited to, N,N-dioleyl-N,N-dimethylammonium chloride ("DODAC"); N-(2,3- dioleyloxy)propyl)-N,N,N-trimethylammonium chloride ("DOTMA"); N-(2,3- dioleyloxy)propyl)-N,N-dimethylammonium chloride ("DODMA"); N,N-distearyl-N,N- dimethylammonium bromide ("DDAB"); N-(2,3-dioleoyloxy)propyl)-N,N,N- trimethylammonium chloride ("DOTAP"); N-(2,3-dioleoyloxy)propyl)-N,N- dimethylammonium chloride ("DODAP"); 3-(N-(N',N'- dimethylaminoethane)carbamoyl)cholesterol ("DC-Choi"); N-( 1 ,2-dimyristyloxyprop-3-yl)- N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE"); 1.2-dilinoleyloxy-N,N- dimethyl-3-aminopropane (DLinDMA); l,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA); 1.2-dilinolenyloxy-N,N-dimethyl-3-aminopropane (DLenDMA); 2-{4-[(3b)- cholest-5-en-3-yloxy]butoxy}-N,N-dimethyl-3-[(9Z,12Z)-octadeca-9,12-dien-l- yloxy]propan-amine (CLinDMA).
[0075] In some embodiments the mixture of lipids may include an anionic lipid. The anionic lipid may be but is not limited to diacylglycerol phophatidic acid ( 1 ,2-distearoyl-sn- glycero-3-phosphate (DSPA); l,2-dipalmitoyl-sn-glycero-3-phosphate (DPPA); 1,2- dimyristoyl-sn-glycero-3-phosphate (DMPA); 1,2-dilauroyl -sn-glycero-3-phosphate (DLPA); l,2-dioleoyl-sn-glycero-3-phosphate (DOPA)), diacylglycerol phosphoglycerol (l,2-distearoyl-sn-glycero-3-phospho-( -rac-glycerol) (DSPG); 1,2-dipalmitoyl-sn-glycero- 3-phospho-(l'-rac-glycerol) (DPPG); l,2-dimyristoyl-sn-glycero-3-phospho-(l'-rac-glycerol) (DMPG); 1,2-dilauroyl -sn-glycero-3-phospho-(l'-rac-glycerol) (DLPG); 1,2-dioleoyl-sn- glycero-3-phospho-(l'-rac-glycerol) (DOPG)), phosphatidylglycerol, cardiolipin, diacylphosphatidylserine, N-succinyl phosphatidylethanolamines, N- glutarylphosphatidylethanolamines, lysylphosphatidylglycerols, and other anionic modifying groups joined to neutral lipids. The mixture of lipids may also include a neutral lipid. The neutral lipid may be but is not limited to diacylglycerol phosphocholine (L-a- phosphatidylcholine, hydrogenated (Soy) (HSPC); diacylglycerol phosphocholine (L-a- phosphatidylcholine, (Soy) (Soy PC), l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC); l,2-dipalmitoyl-sn-glycero-3-phosphocholine(DPPC); l,2-dimyristoyl-sn-glycero-3- phosphocholine (DMPC); l,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC); 1,2-dioleoyl- sn-glycero-3-phosphocholine (DOPC), diacylglycerol phosphoethanolamine (1,2- distearoyl- sn-glycero-3-phosphoethanolamine (DSPE); 1 ,2-dipalmitoyl-sn-glycero-3- phosphoethanolamine (DPPE); l,2-dimyristoyl-sn-glycero-3 -phosphoethanolamine (DMPE); 1 ,2-dilauroyl-sn-glycero-3-phosphoethanolamine (DLPE); 1 ,2-dioleoyl-sn-glycero-3- phosphoethanolamine (DOPE), and phosphatidylserine.
[0076] The mixture of lipids may also include a pegylated lipid. The pegylated lipid may be but is not limited to l,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-2000] (mPEG-2000-DSPE ); 1,2-dioctadecanoyl-sn-glycero- 3-phosphoethanolamine -N-[methoxy(polyethylene glycol)-2000] (mPEG-2000-DOPE ); 1,2- dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (mPEG-2000-DPPE ); 1 ,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-2000] (mPEG-2000-DMPE ); l,2-dilauroyl-sn-glycero-3- phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (mPEG-2000-DLPE ); 1,2- distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-5000] (mPEG-5000-DSPE); l,2-dioctadecanoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-5000] (mPEG-5000-DOPE ); l,2-dipalmitoyl-sn-glycero-3- phosphoethanolamine-N-[methoxy(polyethylene glycol)-5000] (mPEG-5000-DPPE); 1,2- dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-5000] (mPEG-5000-DMPE ) ; 1 ,2-dilauroyl- sn-glycero-3 -phosphoethanolamine-N- [methoxy(polyethylene glycol)-5000] (mPEG-5000-DLPE). [0077] The mixture of lipid may also include a lipid-like molecule or lipidoid. The mixture of lipid may also include a lipid- or cholesterol-conjugated molecule including a protein, or a peptide, or an oligonucleotide.
[0078] In various embodiments, the lipid layer includes one or more of the lipid components disclosed herein.
[0079] In various embodiments, the lipid layer consists essentially of the unsaturated phospholipid and cholesterol.
[0080] In various embodiments, the lipid layer consists essentially of the unsaturated phospholipid, cholesterol, and pegylated phospholipid.
[0081] In various embodiments, the lipid layer comprises: about 20-75%, preferably about 30-60%, (molar) unsaturated phospholipid; about 10-60%, preferably 20-50%, (molar) cholesterol; and about 0-20%, preferably 1-10%, (molar) pegylated phospholipid.
[0082] In various embodiments, the molar ratio of the lipid layer
components doxorubicin is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1; and the molar ratio of the lipid layer components :docetaxel is about 100: 1 to about 5: 1, preferably about 20: 1 to about 10: 1.
[0083] In various embodiments, the molar ratio of doxorubicin :docetaxel is about 10: 1 to 1: 10, preferably about 5: 1 to 1:5, and more preferably about 3: 1 to 1:3.
[0084] In various embodiments, the unsaturated phospholipid comprises a polyunsaturated phospholipid or a monounsaturated phospholipid, preferably a
phosphatidylcholine, and more preferably and soy phosphatidylcholine or 1 ,2-dioleoyl-sn- glycero-3-phosphatidylcholine (DOPC).
[0085] In various embodiments, the cholesterol comprises a cholesterol derivative, preferably a cationic cholesterol derivative, more preferably an amino cholesterol derivative, and still more preferably dimethylaminoethanecarbamoyl-cholesterol (DC-cholesterol).
[0086] In various embodiments, the pegylated phospholipid comprises a
phosphoethanolamine, preferably a l,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and wherein the pegylation is a PEG 500 to PEG 5000, preferably PEG 2000.
[0087] In various embodiments, the composition of the lipid layer is tuned to achieve a desired loading of the first drug. Although at least a fraction of the first drug is in the lipid layer, one of ordinary skill will understand that the first drug will have a partition coefficient between the lipid layer and aqueous interior. In some embodiments, essentially all of the first drug will be in the lipid layer. [0088] The aqueous solution of the process preferably includes water and a buffer. Buffers may be of but are not limited to phosphate, histidine, HEPES, Tris, acetate, carbonate, and citrate. In various embodiments, the composition of the aqueous solution is tuned to achieve a desired loading (and/or crystallization) of the second drug. Although at least a fraction of the second drug is in the aqueous interior of the liposome, one of ordinary skill will understand that the second drug will have a partition coefficient between the lipid layer and aqueous interior. In some embodiments, essentially all of the second drug will be in the aqueous interior.
[0089] Methods for Making Liposomes
[0090] Examples of apparatuses and methods that can be adapted for making the liposomes of the invention can be found, for example, in US Patent Application Serial Number 14/209,187 (and published as US20140348900), which is herein incorporated by reference in its entirety. A description of a number of different methods of making liposomes in accordance with the invention are presented in the Examples below.
[0091] The invention provides a method of making the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, comprising: (i) introducing a lipid solution of an unsaturated phospholipid, cholesterol, a first drug (e.g., docetaxel), and preferably a pegylated phospholipid in ethanol through a first port into a mixing chamber and an aqueous solution through a second port into the mixing chamber, thereby making a plurality of liposomes; and (ii) incubating the plurality of liposomes in a second drug (e.g., doxorubicin) solution.
[0092] In various embodiments, the angle between at least one lipid and at one aqueous solution inlet ports is not 180° or a substantially similar angle. In some aspects, at least one stream of lipid solution and at one stream of aqueous solution collide at an angle less than about 180 °. Thus, in some aspects, the method does not include a T-connector.
[0093] In some embodiments, the angle between at least one lipid and at one aqueous solution inlet ports is about 120° or less, e.g., 115° or less, 100° or less, 90° or less, 80° or less, 72° or less, 60° or less, 45° or less, 30° or less, 18° or less,
[0094] In some embodiments, the aqueous solution in step ii) is introduced via at least two inlet ports, e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more. In some embodiments, the aqueous solution in step ii) is introduced via at least 3 but no more than 11 inlet ports, e.g., at least 3 but not more than 7, at least 3 but no more than 5, at least 4 but no more than 11 , at least 5 but no more than 11 , at least 6 but no more than 11. [0095] In some embodiments, at least two (e.g., 3, 4, 5, 6, 7, etc.) aqueous inlet ports and at least one (e.g., 2, 3, 4, 5, etc.) lipid solution inlet port are in the same plane.
[0096] In some embodiments, at least one (e.g., 2) outlet port is substantially perpendicular to the plane of inlet ports. In other embodiments, at least one (e.g., 2, 3, 4, 5, etc.) outlet port is substantially not perpendicular to the plane of inlet ports.
[0097] In some embodiments, at least two (e.g., 3, 4, 5, 6, 7, etc.) aqueous solution inlet ports and at least one (e.g., 2, 3, 4, 5, etc.) lipid solution inlet port are not in the same plane.
[0098] Preparing Lipid Solutions
[0099] The lipid solution may be made from the stock solutions of individual lipids that are mixed together. Lipids are preferably dissolved in an organic solvent to make a lipid solution. The organic solvent used for making the lipid solution may be miscible with water. Preferably the solvent may ethanol, methanol, DMSO, propanol, DMF, THF, acetone, dioxane, ethylene glycol, polyethylene glycol and isopropanol. More preferably, the solvent is polyethylene glycol, isopropanol, and ethanol. Preferably, the solvent includes less than 10% water. In some cases, the lipid solution may be made from a mixture of lipids, thereupon dissolving the mixture in an organic solvent. The concentration of the total lipids in the solution may be in the range from about 1 mg/niL to about 200 mg/mL, e.g., from about 1 mg/mL to about 100 mg/mL. More preferably, the concentration of the total lipids in the solution may be in the range from about 5 mg/mL to about 100 mg/mL or form about 10 mg/mL to 100 mg/mL. In some embodiments, the organic solvent is ethanol at a
concentration of about 70% or more (e.g., 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 100%).
[00100] The mixture of lipids will be optimized as required for optimal delivery of the API and is readily optimized by routine experimentation by one of ordinary skill in the art.
[00101] In certain embodiments, a water-insoluble API may be dissolved in the lipid solution. The concentration of the API in the lipid solution will depend on the efficacy of the agent and may easily be determined by one of ordinary skill in the art. The lipid/ API ratio will determined by the encapsulation power of the liposome to the API.
[00102] Preparing Aqueous Solutions
[00103] A water-soluble API component may be dissolved in a first aqueous solution (S I). The pH and salinity of the solution may be optimized to accommodate the requirements for the interaction between the API component and the lipids to form liposome. These conditions may be readily determined by one of ordinary skill in the art. Samples are provided in the Examples below. As will be readily apparent to those of skill in the art, an aqueous solution that lacks an API, referred to as (S2), may be similar to a solution having the agent. Alternatively, S I and S2 may be different.
[00104] Liposome Preparation, Mixing the Solutions
[00105] The lipid solution and the aqueous solution(s) preferably enter the manifold from different ports, each with a flow rate of from about 1 mL/min to about 6000 mL/min. Preferably, the flow rates may be from about 5 mL/min to about 1000 mL/min. More preferably, the rates may be from about 20 mL/min to about 600 mL/min. In some embodiments, the flow rates are adjusted based on the size of inlet ports to obtain the desired liposome size, morphology, PDI, and manufacturing scales.
[00106] In some embodiments, the lipid solution and/or the aqueous solution is introduced via port size of 0.1-5.0 mm at a flow rate about 1 mL/min to about 2,500 mL/min.
[00107] In some embodiments, the flow velocity of the lipid solution and/or the aqueous solution is from about 0.02m s to about 40 m/s, e.g., from 0.1 m/s to 30 m/s, from 0.2 m/s to 20m/s. The flow velocity is adjusted based on the size of inlet ports to obtain the desired liposome size, morphology, PDI, and manufacturing scale.
[00108] Loading of the API Into liposome
[00109] In the mixing chamber the lipids are believed to instantaneously assemble into liposome particles. When the drug API is carried by the lipid solution or by aqueous solution, it may be encapsulated in the liposome by either lipophilic or electrostatic interaction, or both, between the API and the lipids.
[00110] The present invention also provides a method of producing liposome that do not contain an API (so-called "empty" liposome). In such embodiments, the API is absent from both the lipid solution and the aqueous solution that are mixed in the manifold. The API may be loaded into the liposomes by the process of diffusion or another process. For example, doxorubicin may be loaded into the liposome with a pH gradient. See US Patent application No. 10/ 019,200, PCT Publication No. WO 2001/005373, US Patents Nos.
5,785,987, 5,380,531, 5,316,771, and 5,192,549, all of which are incorporated herein by reference.
[00111] Preferably, the API is mixed with a liposome solution to upload the API into the liposome by diffusion. In one aspect, the API is dissolved in an aqueous solution, and the solution is mixed with the empty liposome. In another aspect, the API may be readily soluble in the solution of empty liposome, and therefore, the API may be directly mixed with the solution of the empty liposome. [00112] The volume ratio of the solution of the API to the empty liposome solution of the API is preferably in the range from about 1:50 to about 1 : 1. A lower volume of the solution is preferred because it avoids a significant dilution to the final liposome solution.
[00113] The drug encapsulation efficiency is preferably greater than 70%. More preferably the efficiency is greater than 80%. Most preferably, the efficiency is greater than 90%.
[00114] Liposome Concentration Adjustment
[00115] Tangent flow filtration may be used to concentrate the liposome solution.
[00116] Buffer Change
[00117] Residual organic solvent in the liposome solution may be removed by a buffer change. Preferably, the buffer change is performed by tangent flow filtration. In another embodiment, the buffer change may be performed by dialysis.
[00118] Sterile Filtration
[00119] The liposome solutions can be sterilized, for example, by passing the solution through a 0.22 micron sterile filter.
[00120] Liposomes
[00121] In various embodiments, the Z-average particle size of the liposomes is about 10-200 nm, preferably about 15- 150 nm, and more preferably about 20-120 nm.
[00122] Preferably, more than 70% of API is encapsulated in the liposomes. More preferably, more than 80% of API is encapsulated in the liposomes, most preferably, more than 90% of API is encapsulated in the liposomes.
[00123] Optionally, liposomes can be unilamellar. Alternatively, the liposomes can be of multilamellar, or of inverted hexagonal or cubic morphology, or as lipid discs, or hollow liposomes.
[00124] In some embodiments, the mean particle size of the liposomes is from about 10 nm to about 2,000 nm, preferably less than 300 nm, more preferably, the mean particle size may be about 10 to 300 nm or about 20 to about 300 nm. Most preferably, the mean particle size is about 20 to 120 nm In some embodiments, the liposomes have a polydispersity index from about 0.005 to about 0.8, e.g., 0.005 to about 0.5, 0.01 to about 0.5, 0.01 to about 0.4, 0.01 to about 0.2.
[00125] Pharmaceutical Compositions
[00126] In various embodiments, the pharmaceutical composition is for use as a medicament. In various embodiments, the pharmaceutical composition is for use as a cancer therapeutic. In various embodiments, the pharmaceutical composition can include one or more antibiotic, antivirus, anti-diabetes, anti-hypertension, anti-fungal, or analgesic.
[00127] In various embodiments, the plurality of liposomes are comprised in an injectable formulation, for example, by subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection. Injectable formulations can be aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or
physiological saline buffer. The injectable formulation can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the liposomes can be in a dried or powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[00128] Treatment and Administration
[00129] The invention provides a method comprising administering the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
[00130] The invention also provides a method of treating a subject comprising administering an effective amount of the liposome according to any of the aspects or embodiments disclosed herein, or the pharmaceutical composition according to any of the aspects or embodiments disclosed herein, to a subject.
[00131] Accordingly, the invention provides methods for treating cancer cells and/or tissue, including cancer cells and/or tissue in a human subject. Cancer can be caused by malignant tumors formed by an abnormal growth of cells and tissue leading to organ failure.
[00132] Solid tumors can be neoplasms (new growth of cells) or lesions (damage of anatomic structures or disturbance of physiological functions) formed by an abnormal growth of body tissue cells other than blood, bone marrow or lymphatic cells. A solid tumor consists of an abnormal mass of cells which may stem from different tissue types such as liver, colon, breast, or lung, and which initially grows in the organ of its cellular origin. However, such cancers may spread to other organs through metastatic tumor growth in advanced stages of the disease.
[00133] The subject being treated may have been diagnosed with cancer. The subject may have locally advanced, unresectable, or metastatic cancer and/or may have failed a prior first-line therapy. In various embodiments, the cancer is liver cancer (e.g., hepatocellular carcinoma, HCC). In various embodiments, the liver cancer (e.g., HCC) can be intermediate, advanced, or terminal stage. The liver cancer (e.g., HCC) can be metastatic or non-metastatic. Liver cancer can include a liver tumor resulting from the metastasis of a non-liver cancer, to the liver. The liver cancer (e.g., HCC) can be resectable or unresectable. The liver cancer (e.g., HCC) can comprise a single tumor, multiple tumors, or a poorly defined tumor with an infiltrative growth pattern (into portal veins or hepatic veins). The liver cancer (e.g., HCC) can comprise a fibrolamellar, pseudoglandular (adenoid), pleomorphic (giant cell), or clear cell pattern. The liver cancer (e.g., HCC) can comprise a well differentiated form, and tumor cells resemble hepatocytes, form trabeculae, cords, and nests, and/or contain bile pigment in cytoplasm. The liver cancer (e.g., HCC) can comprise a poorly differentiated form, and malignant epithelial cells are discohesive, pleomorphic, anaplastic, and/or giant. In some embodiments, the liver cancer (e.g., HCC) is associated with hepatitis B, hepatitis C, cirrhosis, or type 2 diabetes.
[00134] In various embodiments, the cancer is a lung cancer, preferably non-small cell lung cancer (NSCLC); colon cancer; breast cancer; or liver cancer, preferably hepatocellular carcinoma (HCC).
[00135] In various embodiments, the docetaxel can be in a concentration of 10, 20, 30, 40, 50, 75, 80, 100, 125, 150, or 160 mg/mL. A dose can be about 10 mg/m2 to 150 mg/m2 (e.g., 10, 20, 25, 30, 40, 50, 60, 70, 75, 80, 90, 100, 110, 120, 125, 130, 140, or 150 mg/m2). For example, a dose can be 75 mg/m2. A dose can be administered every 3 weeks for 1, 2, 3, 5, 5, or 6 cycles. One skilled in the art will appreciate that dosing guidelines for docetaxel are known in the art, and can be adapted based upon factors including, but not limited to the cancer type, the cancer stage, the dosing regimen, the dose of doxorubicin, and/or the efficacy of the pharmaceutical formulations of the invention.
[00136] In various embodiments, the doxorubicin can be in a concentration of 0.1, 0.5, 1, 1.5, 2, 3, 4, or 5 mg/mL. A dose can be about 1 mg/m2 to 100 mg/m2 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 25, 30, 40, 50, 60, 70, 75, 80, 90, or 100 mg/m2). For example, a dose can be 30 mg/m2. A dose can be administered every 3 weeks for 1, 2, 3, 5, 5, or 6 cycles. One skilled in the art will appreciate that dosing guidelines for docetaxel are known in the art, and can be adapted based upon factors including, but not limited to the cancer type, the cancer stage, the dosing regimen, the dose of doxorubicin, and/or the efficacy of the pharmaceutical formulations of the invention.
[00137] The following examples are illustrative and not restrictive. Many variations of the technology will become apparent to those of skill in the art upon review of this disclosure. The scope of the technology should, therefore, be determined not with reference to the examples, but instead should be determined with reference to the appended claims along with their full scope of equivalents. EXAMPLES
[00138] Example 1: Preparation of Liposomal formulation CPT307C
[00139] CPT307 comprises of a nonsaturated lipid l,2-Dioleoyl-sn-glycero-3- Phosphatidylcholine (DOPC), cholesterol, and l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[methoxy9polyethyleneglycol)-2000] (mPEG2000-DSPE). It was found that compared the saturated lipid, the nonsaturated lipid has a greater capacity to encapsulate docetaxel. Liposomal formulation CPT307B was prepared by first dissolving 2100 mg of DOPC, 280 mg of cholesterol, 700 mg of mPEG2000-DSPE, and 175 mg of docetaxel (DOCE) in 70 mL of anhydrous ethanol. The composition (% molar) of the CPT307B lipid solution is illustrated in Table 1. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Twenty milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass vial. The liposome was concentrated by tangent flow filtration. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration. The formulation was then sterilized by filtration through a 0.22 μιη filter. The Z-average particle size was 32.9 nm.
[00140] CPT307C was prepared by loading doxorubicin (DXR) into CPT307B . Fourteen milliliters of CPT307B containing 36 mg/mL of DOCE was mixed with 24 mg of DXR that had been pre-dis solved in the histidine/sucrose buffer, and incubated at 42 °C for 3 hours. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 μηι filter. The composition (% molar) of the CPT307C lipid solution is illustrated in the Table 1, 99.6% of DXR was encapsulated. The molar ratio of DOCE:DXR was 1:1.
Table 1. Lipid Compositions of Example 1.
Figure imgf000020_0001
*The value represents the molar % of each component vs. total lipids.
[00141] Example 2: Preparation of Liposomal formulation CPT308C
[00142] Different from CPT307C in Example 1, CPT308C contains a polyunsaturated lipid L-a-phosphatidylcholine (Soy PC) that has a high capacity to encapsulate DOCE. Two milliliters of lipids/DOCE solution was prepared by dissolving 30 mg of Soy PC, 10 mg of cholesterol, 10 mg of mPEG2000-DSPE, and 6 mg of DOCE in anhydrous ethanol. The composition (% molar) of the liposomal formulation CPT308C lipid solution is illustrated in Table 2. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Two milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass vial. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by dialysis. The formulation was then sterilized by filtration through a 0.22 μηι filter.
[00143] The DOCE loaded liposome was then mixed with DXR that had been pre- dissolved in the histidine/sucrose buffer at a doxorubicin/lipid ratio (w/w) of 1:10, and incubated at 42 °C for 2 hours, 97% of DXR was encapsulated. The DOCE and DXR dual- loaded liposome was then sterilized by filtration through a 0.22 μιη filter. The Z-average particle size of the dual-loaded liposome was 38.2 nm for CPT308C.
Table 2. Lipid Composition of Example 2.
Figure imgf000021_0001
The value represents the molar % of each component vs. total lipids.
[00144] Example 3: Preparation of Liposomal formulation CPT309C [00145] CPT309C contains the polyunsaturated lipid Soy PC at a higher molar ratio than CPT308C in Example 2 and thus showed a greater capacity to encapsulate DEOCE. Two milliliters of lipids DOCE solution was prepared by dissolving 30 mg of L-a- phosphatidylcholine (Soy PC), 4 mg of cholesterol, 10 mg of mPEG2000-DSPE, and 6 mg of DOCE in anhydrous ethanol. The composition (% molar) of the liposomal formulation CPT309C lipid solution is illustrated in Table 3. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Two milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass vial. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by dialysis. The formulation was then sterilized by filtration through a 0.22 μιη filter.
[00146] The DOCE loaded liposome was then mixed with DXR that had been pre- dissolved in the histidine/sucrose buffer at a doxorubicin/lipid ratio (w/w) of 1:10, and incubated at 42 °C for 2 hours, 98.8% of DXR was encapsulated. The DOCE and DXR dual- loaded liposome was then sterilized by filtration through a 0.22 μιη filter. The Z-average particle size of the dual-loaded liposome was 38.6 nm for CPT309C.
Table 3. Lipid Composition of Example 3.
Figure imgf000022_0001
The value represents the molar % of each component vs. total lipids.
[00147] Example 4: Preparation of Liposomal formulation CPT311C
[00148] CPT311C is a cationic liposome as it contains a cationic derivative of cholesterol (DC-cholesterol). It was found that cationic lipids enhances liver- targeting delivery and anti-tumor efficacy of the therapeutic agent in the liposome. Two milliliters of lipids/DOCE solution (liposomal formulation CPT311B) was prepared in anhydrous ethanol to give concentrations of 15 mg/mL DOPC, 9.3 mg/mL of 3β-[Ν-(Ν\ Ν'- dimethylaminoethane)-carbamoyl]cholesterol hydrochloride (DC-Cholesterol), 5 mg/mL of mPEG2000-DSPE, and 2.0 mg/mL of DOCE. The composition (% molar) of the CPT31 IB lipid solution is illustrated in Table 4. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Two milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass vial. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by dialysis. The formulation was then sterilized by filtration through a 0.22 μηι filter. The Z-average particle size was 34.5 nm.
[00149] Liposomal formulation CPT311C was prepared by loading doxorubicin (DXR) into CPT31 IB. Two milliliters of CPT31 IB was mixed with 0.5 mg of DXR that had been pre-dissolved in the histidine/sucrose buffer at 10 mg/mL, and incubated at 42 °C for 2 hours, 94.5% of DXR was encapsulated. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 μιη filter. The composition (% molar) of the CPT311C is illustrated in Table 4. The Z-average particle size of the dual-loaded liposome was 34.9 nm for CPT311C.
Table 4. Lipid Compositions of Example 4.
Figure imgf000023_0001
The value represents the molar % of each component vs. total ipids.
[00150] It was found that the cationic surface charge of liposome promotes liposome delivery to the liver. The alternation of the molar ratio of the cationic lipid, for example, DC- cholesterol in the liposome, controls the liver delivery of liposome and the clearance rate from the blood. The following examples (from Examples 5 to Example 7) comprise of DOCE, cholesterol, DC-cholesterol, and mPEG2000-DSPE with increased molar ratio of DC-cholesterol (from 3.9% increased to 15.4%) and deceased cholesterol molar ratio (from 34.4% reduced to 17.8%) while the molar ratio of DOPC and mPEG2000-DSPE remain unchanged or a minorly changed.
[00151] Example 5: Preparation of Liposomal formulation CPT315C
[00152] Two and one half (2.5) milliliters of lipids/DOCE solution was prepared by dissolving 37.5 mg DOPC, 11.3 mg cholesterol, 1.4 mg of 3β-[Ν-(Ν', Ν'- dimethylaminoethane)-carbamoyl]cholesterol hydrochloride (DC-Cholesterol), 12.5 mg mPEG2000-DSPE , and 4 mg DOCE in 2.5 mL anhydrous ethanol. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Two and one half (2.5) milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass vial. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by dialysis.
[00153] The DOCE loaded liposome was then mixed with DXR that had been pre- dissolved in the histidine/sucrose buffer at a doxorubicin/lipid ratio (w/w) of 1:16, and incubated at 42 °C for 2 hours, 96.9% of DXR was encapsulated. The DOCE and DXR dual- loaded liposome was then sterilized by filtration through a 0.22 μιη filter. The Z-average particle size of the dual-loaded CPT315C was 35.1 nm.
Table 5. Lipid Composition of Example 5.
Figure imgf000024_0001
*The value represents the molar % of each component vs. total lipids.
[00154] Example 6: Preparation of Liposomal formulation CPT317C
[00155] Twenty milliliters of lipids/DOCE solution was prepared by dissolving 600 mg of DOPC, 140 mg of cholesterol, 84 mg of DC-Cholesterol, 200 mg of mPEG2000- DSPE, and 50 mg of DOCE in anhydrous ethanol. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Twenty milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass bottle and then was concentrated by tangent flow filtration. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration. The formulation was then sterilized by filtration through a 0.22 μιη filter. Liposomal formulation CPT317B loaded with DOCE was obtained. The Z-average particle size of CPT317B was 37.5 nm.
[00156] Liposomal formulation CPT317C was prepared by loading doxorubicin (DXR) into CPT317B. Five milliliters of CPT317B was mixed with 5.2 mg of DXR, and incubated at 42 °C for 3 hours. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 μιη filter. The composition (% molar) of the CPT317C lipid solution is illustrated in Table 6. 99.8% of DXR was encapsulated.
Table 6. Lipid Composition of Example 6.
Figure imgf000025_0001
The value represents the molar % of each component vs. total lipids.
[00157] Example 7: Preparation of Liposomal formulation CPT319C [00158] The lipids/DOCE solution was prepared by dissolving 1848 mg of DOPC, 303 mg of cholesterol, 423 mg of DC-Cholesterol, 605 mg of mPEG2000-DSPE, and 154 mg of DOCE in 61.5 mL of anhydrous ethanol. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Twenty milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass bottle and then was concentrated by tangent flow filtration. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration. The formulation was then sterilized by filtration through a 0.22 μιη filter to obtain liposomal formulation CPT319B loaded with DOCE.
[00159] Liposomal formulation CPT319C was prepared by loading doxorubicin (DXR) into CPT319B. In a glass bottle 31.4 mg of DXR was dissolved in 30mL of
CPT319B. The mixture was incubated at 42 °C for 4 hours, 99.8% of doxorubicin was encapsulated. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 μιη filter. The final composition (% molar) of the CPT317C lipid solution is illustrated in Table 7. The Z-average particle size was 40.7 nm and the molar ratio of DOCE:DXR was 1: 1 for CPT319C. The Cryo-TEM images of CPT319C are shown in FIG. 1. The DXR crystals formed inside the liposome can be seen from these images. The negative stained TEM image of CPT319C is shown in FIG. 2, which indicates particle size and homogeneity.
Table 7. Lipid Composition of Example 7.
Figure imgf000026_0001
The value represents the molar % of each component vs. total lipids. [00160] Example 8: Preparation of Liposomal formulation CPT323C
[00161] CPT323C was prepared in the absence of pegylated lipid, thus the pegylated lipid is optional to the formulations. The lipids/DOCE solution was prepared by dissolving 300 mg of DOPC, 50 mg of cholesterol, 70 mg of DC-Cholesterol, and 25 mg of DOCE in 10 mL of anhydrous ethanol. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Ten milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing.
Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass bottle and was then concentrated by tangent flow filtration. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration. The formulation was then sterilized by filtration through a 0.22 μηι filter.
[00162] Three milliliters of the DOCE loaded liposome was then mixed with 3 mg of DXR that had been pre-dis solved in the histidine/sucrose buffer at 7 mg/mL, incubated at 42 °C for 6 hours, 96.3% of DXR was encapsulated The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 μιη filter to obtain CPT323C. The composition (% molar) of the liposomal formulation CPT323C lipid solution is illustrated in Table 8. The Z-average particle size of the dual-loaded liposome was 49.0 nm for CPT323C.
Table 8. Lipid Composition of Example 8.
Figure imgf000027_0001
*The value represents the molar % of each component vs. total lipids.
[00163] Example 9: Preparation of Liposomal formulation CPT324C
[00164] Different from other exemplary formulations, CPT324C contains a polyunsaturated lipid- Soy PC and a cationic lipid DOTAP other than DC-cholesterol . The lipids/DOCE solution was prepared by dissolving 60 mg of Soy PC, 40 mg of cholesterol, 60 mg of l,2-dioleoyl-3-trimethylammonium-propane (chloride salt) (DOTAP), 40 mg of mPEG2000-DSPE, and 25 mg of DOCE in 10 mL of anhydrous ethanol. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. Ten milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing. Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass bottle and was then concentrated by tangent flow filtration. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by tangent flow filtration. The formulation was then sterilized by filtration through a 0.22 μιη filter.
[00165] Three milliliters of the DOCE loaded liposome was then mixed with 0.95 mg of DXR that had been pre-dissolved in the histidine/sucrose buffer at 7 mg/mL, and incubated at 42 °C for 6 hours. The DOCE and DXR dual-loaded liposome was then sterilized by filtration through a 0.22 μιη filter to obtain liposomal formulation CPT324C. The composition (% molar) of the CPT324C lipid solution is illustrated in Table 9. The Z-average particle size of the dual-loaded liposome was 56 nm for CPT324C.
Table 9. Lipid Composition of Example 9.
Figure imgf000028_0001
The value represents the molar % of each component vs. total lipids.
[00166] Example 10: Preparation of Liposomal formulation CPT313C
[00167] CPT313C was prepared in the presence of DC-cholesterol and the absence of cholesterol. The lipids/DOCE solution was prepared by dissolving 33 mg of Soy PC, 20.5 mg of DC-Cholesterol, 11 mg of mPEG2000-DSPE, and 4.4 mg of DOCE in 2.2 mL of anhydrous ethanol. In addition, three aqueous solutions of 250 mM ammonium sulfate, pH 6.5 were used. 2.2 milliliter of each of the above four solutions was loaded into a 20 mL syringe. Each syringe was connected to an inlet port of a five-port manifold by tubing.
Through the tubing, the solutions in the syringes were pumped into the mixing chamber of the manifold by a syringe pump. The liposome solution exited through an outlet port and was collected in a glass vial. The buffer was changed into a histidine/sucrose buffer (10 mM histidine, 9.2% sucrose, pH 6.5) by dialysis. The formulation was then sterilized by filtration through a 0.22 μιη filter.
[00168] Two milliliters of the DOCE loaded liposome was then mixed with 0.5 mg of DXR that had been pre-dissolved in the histidine/sucrose buffer at 10 mg/mL, and incubated at 42 °C for 2 hours, 90.6% of DXR was encapsulated. The encapsulated liposome was then sterilized by filtration through a 0.22 μιη filter to obtain liposomal formulation CPT313C. The composition (% molar) of the CPT313C lipid solution is illustrated in Table 10. The Z- average particle size of the dual-loaded liposome was 38.7 nm for CPT313C.
Table 10. Lipid Composition of Example 10.
Figure imgf000029_0001
The value represents the molar % of each component vs. total lipids.
[00169] Example 11: Dual-loaded liposome CPT319C augments efficacy against non-small cell lung cancer (NSCLC)
[00170] Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously on the right flank with NSCLC cell line A549 tumor cells (1 x 107 cells/mouse) in 0.1 mL phosphate buffered saline (PBS) buffer for tumor development. On Day 16 following tumor cell inoculation (tumor size was approximately 117 mm3), treatments were started with formulations of CPT319A at 5 mg/kg doxorubicin, CPT319B at 7.5 mg kg docetaxel, CPT319C at 5 mg kg doxorubicin/7.5 mg/kg docetaxel, or the non- liposomal combination formulation of 5 mg/kg doxorubicin/7.5 mg/kg docetaxel by intravenous (IV) injection through the tail vein. Three additional treatments were administered on Day 20, Day 27, and Day 34. The study was terminated on Day 45. The tumor growth curves and tumor weight inhibition percentages (TW inh%) on Day 45 of the formulations compared to the PBS control group are shown in FIG. 3.
[00171] To summarize, FIG. 3 shows that dual-loaded Liposome CPT319C enhances the antitumor activity of the formulation in NSCLC Xenograft mouse model. All of the liposomal formulations were more efficacious than the non-liposomal combination of DOCE/DXR. In addition, the dual-loaded liposome, CPT319C was the most efficacious formulation in this example. Compared to the PBS control group, CPT319C reduced 76% of the tumor weight that was significantly more efficacious than the 51% of DOCE liposome CPT319B, 27% of DXR liposome CPT319A, and 17% of the non-liposomal combination of DOCE/DXR.
[00172] Example 12: Dual-loaded liposome CPT307C augments efficacy against non-small cell lung cancer (NSCLC)
[00173] Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with NSCLC cell line A549 tumor cells (1 x 107 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 16 following tumor cell inoculation (tumor size was approximately 117 mm ), treatments were started with formulations of CPT307A at 5 mg/kg doxorubicin, CPT307B at 7.5 mg kg docetaxel, CPT307C at 5 mg/kg doxorubicin/7.5 mg kg docetaxel, or the non-liposomal combination formulation of 5 mg/kg doxorubicin/7.5 mg kg docetaxel by intravenous (IV) injection through the tail vein. Three additional treatments were administered on Day 20, Day 27, and Day 34. The study was terminated on Day 45. The tumor growth curves and tumor weight inhibition percentages (TW inh%) on Day 45 of the formulations compared to the PBS control group are shown in FIG. 4.
[00174] To summarize, FIG. 4 shows that dual-loaded liposome CPT307C augments efficacy against non-small cell lung cancer (NSCLC). All of the liposomal formulations were more efficacious than the non-liposomal combination of DOCE/DXR. In addition, the dual- loaded liposome, CPT307C was the most efficacious in this example. Compared to the PBS control group, the dual-loaded liposome CPT307C 57% of the tumor weight that is significant more efficacious than the 44% of DOCE liposome CPT307B, 11% of DXR liposome CPT307A, and 17% of the non-liposomal combination of DOCE/DXR. [00175] Example 13: Antitumor activity of dual-loaded liposome CPT319C against human colon cancer in xenograft mouse model
[00176] Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human colon cancer cell line HCT- 116 tumor cells (5 x 106 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 9 following tumor cell inoculation (tumor size was approximately 141 mm ), treatments were started with formulations of CPT319C at 3 different doses: 5 mg/kg doxorubicin/7.5 mg kg docetaxel, 2.5 mg/kg doxorubicin/3.75 mg/kg docetaxel, or 1.25 mg kg doxorubicin/1.875 mg/kg docetaxel by intravenous (IV) injection through the tail vein. Two additional treatments were administered on Day 16 and Day 23. The study was terminated on Day 37. The tumor growth curves shown in FIG. 5, which illustrates dose responses of the liposomal formulations in a HCT-116 human colon cancer xenograft model. Compared to the PBS control group, the dual-loaded CPT319C reduced 77% of the HCT- 116 tumor size on Day 37 in the 5 mg/kg doxorubicin/7.5 mg kg docetaxel group, 49% in the group treated with 2.5 mg/kg doxorubicin/3.75 mg/kg docetaxel, and 29% in the group treated with 1.25 mg/kg doxorubicin/1.875 mg/kg docetaxel.
[00177] Example 14: Antitumor activity of dual-loaded liposome CPT307C against human colon cancer in xenograft mouse model
[00178] Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human colon cancer cell line HCT- 116 tumor cells (5 x 106 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 9 following tumor cell inoculation (tumor size was approximately 141 mm3), treatments were started with formulations of CPT307C at 3 different doses: 5 mg/kg doxorubicin/7.5 mg kg docetaxel, 2.5 mg/kg doxorubicin/3.75 mg/kg docetaxel, or 1.25 mg/kg doxorubicin/1.875 mg kg docetaxel by intravenous (IV) injection through the tail vein. Two additional treatments were administered on Day 16 and Day 23. The study was terminated on Day 37. The tumor growth curves shown in FIG. 6, which illustrates dose responses of the liposomal formulations in a HCT-116 human colon cancer xenograft model. Compared to the PBS control group, the dual-loaded CPT307C reduced 67% of the HCT- 116 tumor size on Day 37 in the 5 mg/kg doxorubicin/7.5 mg kg docetaxel group, 61% in the group treated with 2.5 mg/kg doxorubicin/3.75 mg/kg docetaxel, and 45% in the group treated with 1.25 mg/kg doxorubicin/1.875 mg/kg docetaxel. [00179] Example 15: Antitumor activity of dual-loaded liposome CPT319C and CPT307C against human breast cancer in xenograft mouse model
[00180] Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human breast cancer cell line MD A- MB -231 tumor cells (7 x 106 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 7 after tumor cell inoculation (tumor size was approximately 174 mm ), treatments were started with formulations of CPT319C or CPT307C at 5 mg/kg doxorubicin/7.5 mg/kg docetaxel by intravenous (IV) injection through the tail vein. Two additional treatments were made on Day 14 and Day 21. The study was terminated on Day 31. The tumor growth curves were shown in FIG. 7.
[00181] To summarize, FIG. 7 shows CPT319C and CPT307C antitumor activity in a human breast cancer xenograft model. Compared to the PBS control group, the tumor size was reduced 38% and 32% in the group treated by CPT319C and CPT307C, respectively.
[00182] Example 16: Antitumor activity of dual-loaded liposome CPT319C against human primary hepatocellular carcinoma in xenograft mouse model
[00183] Female Balb/c nude mice ranging from 6-8 weeks were split up into groups of three. Each mouse was inoculated subcutaneously at the right flank with fragments of human primary hepatocellular carcinoma tumor cells (P3 WP HCC) for tumor development. On Day 32 after tumor inoculation (tumor size was approximately 143 mm3), treatments were started with a formulation of CPT319C at 5 mg/kg doxorubicin/7.5 mg/kg docetaxel by intravenous (IV) injection through the tail vein. Two additional treatments were made on Day 39 and Day 46. The study was terminated on Day 63. The tumor growth curves are shown in FIG. 8.
[00184] To summarize, FIG. 8 shows CPT319C antitumor activity in a human primary HCC xenograft model. Tumor growth was almost completely inhibited by CPT319C.
Specifically, CPT319C inhibited 88% of tumor growth when the study was terminated on Day 63 compared to the vehicle control group.
[00185] Example 17: Cationic lipid DC-Cholesterol enhances the antitumor activity of liposomes against NSCLC
[00186] Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with NSCLC cell line A549 tumor cells (1 x 107 cells/mouse) in 0.1 mL PBS buffer for tumor development. On Day 16 following tumor cell inoculation (tumor size was approximately 117 mm ), treatments were started with formulations of CPT307A or CPT319A at 5 mg kg doxorubicin, CPT307B or CPT319B at 7.5 mg kg docetaxel, CPT307C or CPT319C at 5 mg kg doxorubicin/7.5 mg kg docetaxel, or the non-liposomal combination formulation of 5 mg/kg doxorubicin/7.5 mg kg docetaxel by intravenous (IV) injection through the tail vein. Three additional treatments were made on Day 20, Day 27, and Day 34. The study was terminated on Day 45. The lipid compositions of the CPT307 (without DC-Cholesterol) and CPT319 (with DC-Cholesterol) formulations are shown in Table 11. The tumor growth curves are shown in FIG. 9.
[00187] To summarize, FIG. 9 shows that a cationic lipid DC-cholesterol can enhance the antitumor activity of the liposomes against NSCLC. The tumor inhibition rank order (from low to high) was:
PBS<CPT307A<CPT319A<CPT307B<CPT319B<CPT307C<CPT319C. For each instance, CPT319 (with DC-Cholesterol) was no exceptionally more efficacious than CPT307 (without DC-Cholesterol) indicating that the incorporation of the cationic lipid DC-cholesterol enhances the anti-tumor efficiency of the liposomal formulations.
Table 11. Lipid Compositions of Example 17.
Figure imgf000033_0001
[00188] Example 18: Cationic lipid DC-Cholesterol enhances the antitumor activity of liposomes against colon cancer
[00189] Female Balb/c nude mice ranging from 6-8 weeks of age were inoculated subcutaneously at the right flank with human colon cancer cell line HCT- 116 tumor cells (5 x 106 cells/mouse) in 0.1 mL PBS buffer for tumor development. On day 9 following tumor cell inoculation (tumor size was approximately 141 mm ), treatments were started with formulations of CPT319C (containing DC -Cholesterol) or CPT307C (without DC- Cholesterol) at 5 mg/kg DXR 7.5 mg/kg DOCE by intravenous (IV) injection through the tail vein. Two additional treatments were made on Day 16 and Day 23. The study was terminated on Day 37. The tumor growth curves and tumor weight inhibition percentages (TW Inh%) on Day 37 of the formulations compared to the vehicle control group are shown in FIG. 10.
[00190] To summarize, FIG. 10 shows that a cationic lipid DC-cholesterol can enhance the antitumor activity of the liposomes against colon cancer. CPT319C (with DC- Cholesterol) was more efficacious than CPT307C (without DC-Cholesterol), indicating that the incorporation of the cationic lipid DC-cholesterol enhances the anti-tumor efficiency of the liposomal formulations.
[00191] Example 19: Liposome improves pharmacokinetics (PK) and cationic lipid DC-Cholesterol increases the half-life (ti/2) of DXR
[00192] Male CD-I mice ranging from 20-25 g body weight were split up into groups of three. Each mouse was administered with a single dose of CPT319C or CPT307C at 5 mg/kg DXR/7.5 mg kg DOCE by intravenous (IV) injection through the tail vein. A non- liposomal combination of DXR/DOCE was used as the control. Blood samples were collected at 0.167, 1, 3, 8, 24, and 48h after the injection. DXR plasma concentration was determined by liquid chromatography-tandem mass spectrometry. The plasma concentration curves of DXR are shown in FIG. 11.
[00193] To summarize, FIG. 11 shows that a liposome can improve pharmacokinetics and that a cationic lipid DC-cholesterol increases the half-life (ti/2) of DXR. BLOQ = Below Limit of Quantitation. The ti/2 and area under the plasma concentration time curve (AUC) are provided in the table below. The non-liposomal DXR was cleared quickly from the blood and resulted in a very low AUC (688 h x ng/mL), whereas CPT319C and CPT307C increased AUC by 143 and 204 fold, respectively. Moreover, CPT319C exhibited a 5.9h ti/2 compared to the lh ti/2 of CPT307C, indicating that the cationic lipid DC-Cholesterol in CPT319C improves PK of the formulation by increasing circulation time in the blood.
Table 12. t and AUC of DXR in CD-I mice
Figure imgf000034_0001

Claims

1. A liposome comprising:
an active pharmaceutical ingredient (API) comprising docetaxel and doxorubicin; a lipid layer comprising an unsaturated phospholipid, a cholesterol, and preferably a pegylated phospholipid; and
an aqueous interior,
wherein the docetaxel is in the lipid layer and the doxorubicin is crystallized in the aqueous interior.
2. The liposome of claim 1, wherein the lipid layer consists essentially of the unsaturated phospholipid and cholesterol.
3. The liposome of claim 1, wherein the lipid layer consists essentially of the unsaturated phospholipid, cholesterol, and pegylated phospholipid.
4. The liposome of any of claims 1-3, wherein the API consists essentially of docetaxel and doxorubicin.
5. The liposome of any of claims 1-4, wherein the lipid layer comprises:
about 20-75%, preferably about 30-60%, (molar) unsaturated phospholipid;
about 10-60%, preferably 20-50%, (molar) cholesterol; and
about 0-20%, preferably 1-10%, (molar) pegylated phospholipid.
6. The liposome of any of claims 1-5, wherein:
the molar ratio of the lipid layer components doxorubicin is about 100: 1 to about 2:1, preferably about 20: 1 to about 5:1; and
the molar ratio of the lipid layer components:docetaxel is about 100:1 to about 2: 1, preferably about 20: 1 to about 5:1.
7. The liposome of any of claims 1-6, wherein the molar ratio of doxorubicin: docetaxel is about 10: 1 to 1:10, preferably about 5:1 to 1:5, and more preferably about 3:1 to 1:3.
8. The liposome of any of claims 1-7, wherein the unsaturated phospholipid comprises a polyunsaturated phospholipid or a monounsaturated phospholipid, preferably a
phosphatidylcholine, and more preferably and soy phosphatidylcholine or 1,2-dioleoyl-sn- glycero-3-phosphatidylcholine (DOPC).
9. The liposome of any of claims 1-8, wherein the cholesterol comprises a cholesterol derivative, preferably a cationic cholesterol derivative, more preferably an amino cholesterol derivative, and still more preferably dimethylaminoethanecarbamoyl-cholesterol (DC- cholesterol), a cationic lipid, or a cationic polymer and/or wherein the composition is adapted to target one or more organs in a subject.
10. The liposome of any of claims 1-9, wherein the pegylated phospholipid comprises a phosphoethanolamine, preferably a l,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and wherein the pegylation is a PEG 500 to PEG 3000, preferably PEG 2000.
11. A pharmaceutical composition comprising a plurality of liposomes according to any of claims 1-10.
12. The pharmaceutical composition of claim 11, wherein the plurality of liposomes are comprised in an intravenous formulation.
13. The pharmaceutical composition of any of claims 11-12, wherein the Z-average particle size of the liposomes is about 10-200 nm, preferably about 15-150 nm, and more preferably about 20-120 nm.
14. The pharmaceutical composition of any of claims 11-13, wherein, upon intravenous administration to a subject, at least about 10% of the composition is delivered to the liver.
15. The pharmaceutical composition of any of claims 11-14, for use as a medicament.
16. The pharmaceutical composition of any of claims 11-14, for use as a cancer therapeutic.
17. A method comprising administering the liposome of any of claims 1-10 or the pharmaceutical composition of any of claims 11-16 to a subject.
18. A method of treating a subject comprising administering an effective amount the liposome of any of claims 1-10 or the pharmaceutical composition of any of claims 11-16 to the subject, wherein the subject has a cancer.
19. The pharmaceutical composition of claim 16 or method of claim 16-18, wherein the cancer is a lung cancer, preferably non-small cell lung cancer (NSCLC); colon cancer; breast cancer; stomach cancer, esophagus cancer , prostate cancer, leukemia, head and neck cancer, pancreatic cancer, multiple myeloma, or liver cancer.
20. A method of making the liposome of any of claims 1-10 or the pharmaceutical composition of any of claims 11-16, comprising:
concurrently introducing a lipid solution of an unsaturated phospholipid, cholesterol, docetaxel, and preferably a pegylated phospholipid in ethanol through a first or plural inlet port of a manifold into a mixing chamber of the manifold and an aqueous solution through a second or plural inlet port of the manifold into the mixing chamber of the manifold into the mixing chamber, and the liposomes formed exit the mixing chamber through one or plural outlet chambers of the manifold, thereby making a plurality of liposomes; and
incubating the plurality of liposomes in a doxorubicin solution.
PCT/US2016/020647 2015-03-03 2016-03-03 Dual loaded liposomal pharmaceutical formulations WO2016141161A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP16759478.7A EP3265063A4 (en) 2015-03-03 2016-03-03 Dual loaded liposomal pharmaceutical formulations
CN201680013636.7A CN107530291A (en) 2015-03-03 2016-03-03 Double heavy duty liposomal pharmaceutical preparations
JP2017546067A JP2018507227A (en) 2015-03-03 2016-03-03 Liposome drug formulation with two drugs
CA2977397A CA2977397A1 (en) 2015-03-03 2016-03-03 Dual loaded liposomal pharmaceutical formulations

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562127479P 2015-03-03 2015-03-03
US62/127,479 2015-03-03

Publications (1)

Publication Number Publication Date
WO2016141161A1 true WO2016141161A1 (en) 2016-09-09

Family

ID=56848644

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/020647 WO2016141161A1 (en) 2015-03-03 2016-03-03 Dual loaded liposomal pharmaceutical formulations

Country Status (6)

Country Link
US (1) US10736845B2 (en)
EP (1) EP3265063A4 (en)
JP (1) JP2018507227A (en)
CN (1) CN107530291A (en)
CA (1) CA2977397A1 (en)
WO (1) WO2016141161A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2017061562A1 (en) * 2015-10-07 2018-07-05 塩水港精糖株式会社 Liposomes encapsulating taxane compounds
EP3915544A1 (en) * 2020-05-25 2021-12-01 Leon-Nanodrugs GmbH Method for producing a liposome dispersion

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016141167A1 (en) 2015-03-03 2016-09-09 Cureport, Inc. Combination liposomal pharmaceutical formulations
IL264842B (en) * 2016-08-18 2022-08-01 Troy Bremer Delivery of urea to cells of the macula and retina using liposome constructs
KR102068295B1 (en) * 2017-12-26 2020-01-20 한국화학연구원 Cancer microenvironment-sensitive combinatorial drug delivery system containing a new lipid-peptide-polymer conjugate as a substrate of cancer-overexpressing enzyme
KR102081810B1 (en) * 2017-12-26 2020-05-28 한국화학연구원 Liposome encapsulated with double anti-cancer drug and method for producing the same
CN111773377B (en) * 2019-04-04 2023-03-14 中南大学 Application of anidulafungin in preparation of antitumor drugs and antitumor drugs
US20230172856A1 (en) * 2020-05-06 2023-06-08 Nanotech Pharma Inc. Liposome formulations for treatment of cancers and drug resistance of cancers
CN112618559A (en) * 2020-12-28 2021-04-09 福建省立医院 Anti-tumor preparation and preparation method thereof
EP4333808A1 (en) * 2021-05-05 2024-03-13 leon-nanodrugs GmbH Methods for producing nanoparticle dispersions
CN114259468A (en) * 2021-12-30 2022-04-01 广西大学 Preparation method of long-circulating liposome carrying docetaxel and adriamycin together

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090098212A1 (en) * 2007-03-30 2009-04-16 Epitarget As Acoustically sensitive drug delivery particle
US20110070294A1 (en) * 2009-09-23 2011-03-24 Javeri Indu Methods for the Administration of Drugs Using Liposomes

Family Cites Families (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4898735A (en) * 1985-12-06 1990-02-06 Yissum Research And Development Company Of The Hebrew University Of Jerusalem Liposome/doxorubicin composition and method
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5264618A (en) * 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
WO2000064946A2 (en) 1999-04-28 2000-11-02 Board Of Regents, The University Of Texas System Compositions and methods for cancer treatment by selectively inhibiting vegf
GB9921960D0 (en) 1999-09-16 1999-11-17 Pharmacia & Upjohn Spa Formulations for parenteral use of estramustine phosphate and amino acids
ES2186484B1 (en) 2000-10-10 2004-07-01 Lipotec, S.A. LIPOSOMES ENCAPSULATING ANTI-CANCER DRUGS AND USE OF THEM IN THE TREATMENT OF EVIL TUMORS.
GB0106041D0 (en) * 2001-03-12 2001-05-02 Cancer Res Ventures Ltd Lipids and liposomes
CN1512890A (en) 2001-04-25 2004-07-14 开米卡公司 HCG formulation
US7850990B2 (en) 2001-10-03 2010-12-14 Celator Pharmaceuticals, Inc. Compositions for delivery of drug combinations
US20080075762A1 (en) 2001-10-03 2008-03-27 Paul Tardi Compositions for delivery of drug combinations
CA2491216C (en) 2002-06-26 2012-01-03 Medigene Oncology Gmbh Method of producing a cationic liposomal preparation comprising a lipophilic compound
US7678386B2 (en) 2002-07-15 2010-03-16 Board Of Regents The University Of Texas Liposomes coated with selected antibodies that bind to aminophospholipids
JP2006523681A (en) 2003-03-24 2006-10-19 ルイトポルド・ファーマシューティカルズ・インコーポレーテッド Xanthone, thioxanthone and acridinone as DNA-PK inhibitors
CA2521414A1 (en) 2003-04-02 2004-10-21 Celator Pharmaceuticals, Inc. Methods to individualize combination therapy
CA2526278A1 (en) 2003-05-20 2004-12-02 Aronex Pharmaceuticals, Inc. Combination chemotherapy comprising a liposomal platinum complex
US20060165744A1 (en) 2003-05-22 2006-07-27 Neopharm, Inc Combination liposomal formulations
EP1537858A1 (en) 2003-12-04 2005-06-08 Vectron Therapeutics AG Drug delivery vehicles and uses thereof
WO2005070126A2 (en) 2004-01-08 2005-08-04 The Regents Of The University Of Colorado Methods and compositions for treating human diseases and wounds with ucp and fas inhibitors
WO2006004935A2 (en) 2004-06-29 2006-01-12 Neopharm, Inc. Pegylated cardiolipin analogs, methods of synthesis, and uses thereof
US7511016B2 (en) 2004-07-07 2009-03-31 Mosamedix B.V. Annexins, derivatives thereof, and annexin-cys variants, as well as therapeutic and diagnostic uses thereof
EP1827487A2 (en) 2004-11-17 2007-09-05 Board of Regents, The University of Texas System Cancer immunotherapy incorporating p53
US8765116B2 (en) 2005-03-24 2014-07-01 Medifocus, Inc. Apparatus and method for pre-conditioning/fixation and treatment of disease with heat activation/release with thermoactivated drugs and gene products
US8148392B2 (en) 2005-05-25 2012-04-03 Lorus Therapeutics Inc. 2-indolyl imidazo [4,5-d] phenanthroline derivatives and their use in the treatment of cancer
US7662405B2 (en) * 2005-08-09 2010-02-16 The Research Foundation Of State University Of New York Compositions and methods of preparation of liposomal microparticulate IL-12
WO2007028154A2 (en) 2005-09-02 2007-03-08 Northwestern University Encapsulated arsenic drugs
JP2009512682A (en) 2005-10-21 2009-03-26 パナセア バイオテック リミテッド Pharmaceutical composition comprising at least one anticancer drug and at least one polymer
WO2007072221A2 (en) 2005-11-10 2007-06-28 Aurelium Biopharma Inc. Surface marker-directed cancer therapeutics
GR20060100144A (en) 2006-03-03 2007-10-17 Cancer treatment using oxaliplatin encapsulated into liposomes and co-encapsulation into the liposome particle of more than one pharmaceutical preparations, or genes
WO2007149433A2 (en) 2006-06-19 2007-12-27 The Johns Hopkins University Tumor-specific delivery of therapeutic agents via liposomase
EP2046293A2 (en) 2006-07-10 2009-04-15 Medigene AG Use of a cationic colloidal preparation for the diagnosis and treatment of ocular diseases
CA2664919A1 (en) 2006-08-08 2008-02-21 Board Of Regents Of The University Of Texas Multistage delivery of active agents
EP2057179A4 (en) 2006-08-24 2010-11-10 British Columbia Cancer Agency Compositions and methods for treating myelosuppression
EP2079380B1 (en) 2006-09-27 2018-09-05 Medifocus, Inc. Method of treating cancer comprising introduction of heat and delivery of liposome containing an active agent or thermo-activated drug, gene or virus to tissue
AU2007337897A1 (en) 2006-12-22 2008-07-03 Imuthes Limited Lipids and their use as non-viral delivery vehicle
WO2008114274A1 (en) * 2007-03-19 2008-09-25 Fresenius Kabi Onclology Ltd. Proliposomal and liposomal compositions
WO2009038779A2 (en) 2007-09-19 2009-03-26 University Of Tennessee Research Foundation Methods and compositions for inhibiting undesirable cellular proliferation by targeted liposome delivery of active agents
WO2009051712A1 (en) 2007-10-15 2009-04-23 Pronai Therapeutics, Inc. Dnai amphoteric liposome formulation
WO2009059450A1 (en) 2007-11-05 2009-05-14 Shanghai Jiaotong University Peptide ligand directed drug delivery
CN101909581B (en) 2007-11-14 2012-11-14 加利福尼亚大学董事会 Sterol-modified amphiphilic lipids
US20100104629A1 (en) 2008-04-16 2010-04-29 Abbott Laboratories Cationic lipids and uses thereof
WO2010009186A1 (en) * 2008-07-15 2010-01-21 The Board Of Trustees Of The University Of Illinois Liposome formulation having hydrophilic and hydrophobic pharmaceutical compounds co-encapsulated therein
WO2010065329A2 (en) 2008-11-25 2010-06-10 The Board Of Regents Of The University Of Texas System Nanoparticles for cancer treatment
AU2010227549B2 (en) 2009-03-25 2014-02-27 Novartis Ag Pharmaceutical composition containing a drug and siRNA
PE20120923A1 (en) 2009-03-30 2012-08-27 Eisai Randd Man Co Ltd PHARMACEUTICAL COMPOSITIONS INCLUDING LIPOSOMES CONTAINING ERIBULIN OR A SALT OF IT
EP2415464B1 (en) 2009-03-30 2017-05-10 Eisai R&D Management Co., Ltd. Method for producing liposome composition
WO2010140869A2 (en) 2009-06-05 2010-12-09 Iljin Copper Foil Co., Ltd. Complex, multilayer using the same, and device coated with the multilayer
US20100331819A1 (en) 2009-06-24 2010-12-30 Abbott Cardiovascular Systems Inc. Drug Delivery System and Method of Treatment of Vascular Diseases Using Photodynamic Therapy
US8591942B2 (en) 2009-09-23 2013-11-26 Indu JAVERI Methods for the preparation of liposomes comprising docetaxel
US8802138B2 (en) 2009-10-12 2014-08-12 Jessie L.-S. Au Methods and compositions for improved deliver, expression or activity of RNA interference agents
US20110313017A1 (en) 2010-01-13 2011-12-22 Protiva Biotherapeutics, Inc. Snalp formulations containing polyoxazoline-dialkyloxypropyl conjugates
BR112012023501A2 (en) 2010-03-19 2016-05-31 Massachusetts Inst Technology multilamellar lipid vesicles, pharmaceutical composition and method
WO2011120023A1 (en) 2010-03-26 2011-09-29 Marina Biotech, Inc. Nucleic acid compounds for inhibiting survivin gene expression uses thereof
WO2011119995A2 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Formulations and methods of use
US20110250259A1 (en) 2010-04-12 2011-10-13 Kevin Buckman Method of treating and preventing breast diseases and breast cancer with medicated formula
EP2384743A1 (en) 2010-04-27 2011-11-09 Zoser B. Salama Siosomal formulation for intracellular delivery and targeting of therapeutic agents
CA2800497C (en) 2010-05-10 2019-03-12 The Regents Of The University Of California Methods for preparing ratiometric combinatorial nanoparticules
WO2012015901A1 (en) 2010-07-28 2012-02-02 Genzyme Corporation Methods for treating gastric and pancreatic malignancies
WO2012021383A2 (en) 2010-08-10 2012-02-16 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF MITOGEN-ACTIVATED PROTEIN KINASE 1 (MAPK1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
CN103370054A (en) 2010-11-09 2013-10-23 阿尔尼拉姆医药品有限公司 Lipid formulated compositions and methods for inhibiting expression of EG5 and VEGF genes
NZ611427A (en) 2010-12-06 2015-08-28 Penn State Res Found Compositions and methods relating to proliferative diseases
EP2649182A4 (en) 2010-12-10 2015-05-06 Alnylam Pharmaceuticals Inc Compositions and methods for increasing erythropoietin (epo) production
CN102091036A (en) * 2011-01-10 2011-06-15 中国药科大学 Compound liposome containing anti-tumor drugs and preparation method and application thereof
TWI592411B (en) 2011-02-23 2017-07-21 英特爾立秦有限責任公司 Combination of kinase inhibitors and uses thereof
KR101085203B1 (en) * 2011-04-01 2011-11-21 서울대학교산학협력단 Phospholipid nanoparticles for delivery of drugs
WO2012154942A2 (en) 2011-05-10 2012-11-15 The Penn State Research Foundation Ceramide anionic liposome compositions
CN107115314B (en) 2011-06-02 2022-04-29 加利福尼亚大学董事会 Film-encapsulated nanoparticles and methods of use
AU2012268619B2 (en) 2011-06-06 2017-08-17 Sirna Therapeutics, Inc. RNA interference mediated inhibition of isocitrate dehydrogenase (IDH1) gene expression
CN102225054B (en) 2011-06-20 2013-01-16 河北科技大学 Preparation carried with particles of antitumor drug and preparation method thereof
EP2734191A4 (en) 2011-07-19 2015-04-29 Stc Unm Intraperitoneally-administered nanocarriers that release their therapeutic load based on the inflammatory environment of cancers
JP6158180B2 (en) 2011-07-29 2017-07-05 メディベイション プロステイト セラピューティクス, インコーポレイテッド Breast cancer treatment
US9775803B2 (en) 2011-10-19 2017-10-03 Samsung Electronics Co., Ltd. Liposome comprising elastin-like polypeptide and tumor cell targeting material and use thereof
KR20130042905A (en) 2011-10-19 2013-04-29 삼성전자주식회사 Liposome comprising elastin-like polypeptide and use thereof
CA2853729A1 (en) 2011-10-28 2013-05-02 Board Of Regents, The University Of Texas System Novel compositions and methods for treating cancer
EP2773426B1 (en) 2011-10-31 2018-08-22 Mallinckrodt LLC Combinational liposome compositions for cancer therapy
KR102145835B1 (en) 2012-04-12 2020-08-20 조지타운 유니버시티 Compositions for treating ewings sarcoma family of tumors
WO2013169479A1 (en) 2012-05-11 2013-11-14 Massachusetts Institute Of Technology Compositions and methods of treatment of drug resistant cancers
JP6427097B2 (en) 2012-06-15 2018-11-21 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッドThe Brigham and Women’s Hospital, Inc. Compositions for treating cancer and methods for producing said compositions
MX2015000813A (en) 2012-07-18 2015-09-07 Onyx Therapeutics Inc Liposomal compositions of epoxyketone-based proteasome inhibitors.
ITRM20120480A1 (en) 2012-10-09 2014-04-10 Uni Degli Studi Camerino MULTICOMPONENT LIPID NANOPARTICLES AND PROCEDURES FOR THEIR PREPARATION.
CN102935068B (en) 2012-10-19 2014-12-17 浙江海正药业股份有限公司 Preparation method of liposome entrapping water-soluble medicines
JP2016509572A (en) 2012-11-05 2016-03-31 プロナイ セラピューティクス インコーポレイテッド Methods of using biomarkers for the treatment of cancer by modulating BCL2 expression
CN103087124B (en) * 2012-11-21 2016-01-13 浙江海正药业股份有限公司 A kind of method preparing Zorubicin
US10172795B2 (en) 2012-12-12 2019-01-08 University of Pittsburgh—of the Commonwealth System of Higher Education Formulations and carrier systems including compound interactive domains
KR20150100706A (en) 2012-12-28 2015-09-02 블렌드 세라퓨틱스, 인코포레이티드 Targeted conjugates encapsulated in particles and formulations thereof
MX2015012199A (en) * 2013-03-13 2015-11-30 Mallinckrodt Llc Liposomal cisplatin compositions for cancer therapy.
US9693958B2 (en) * 2013-03-15 2017-07-04 Cureport, Inc. Methods and devices for preparation of lipid nanoparticles
CN103622912B (en) * 2013-12-05 2016-02-24 常州金远药业制造有限公司 doxorubicin hydrochloride-docetaxel or paclitaxel liposome preparation and preparation method thereof
WO2016141167A1 (en) 2015-03-03 2016-09-09 Cureport, Inc. Combination liposomal pharmaceutical formulations

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090098212A1 (en) * 2007-03-30 2009-04-16 Epitarget As Acoustically sensitive drug delivery particle
US20110070294A1 (en) * 2009-09-23 2011-03-24 Javeri Indu Methods for the Administration of Drugs Using Liposomes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3265063A4 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2017061562A1 (en) * 2015-10-07 2018-07-05 塩水港精糖株式会社 Liposomes encapsulating taxane compounds
EP3915544A1 (en) * 2020-05-25 2021-12-01 Leon-Nanodrugs GmbH Method for producing a liposome dispersion
WO2021239709A1 (en) * 2020-05-25 2021-12-02 Leon-Nanodrugs Gmbh Method for producing a liposome dispersion

Also Published As

Publication number Publication date
JP2018507227A (en) 2018-03-15
CN107530291A (en) 2018-01-02
US10736845B2 (en) 2020-08-11
EP3265063A4 (en) 2018-11-07
US20160256387A1 (en) 2016-09-08
EP3265063A1 (en) 2018-01-10
CA2977397A1 (en) 2016-09-09

Similar Documents

Publication Publication Date Title
US10736845B2 (en) Dual loaded liposomal pharmaceutical formulations
US10561611B2 (en) Combination liposomal pharmaceutical formulations
US11497715B2 (en) Methods and devices for preparation of lipid nanoparticles
US10639276B2 (en) Liposomes with ginsenoside as membrane material and preparations and use thereof
EP2410988A1 (en) Pharmaceutical composition containing a drug and sirna
CN109528654B (en) Irinotecan hydrochloride and adriamycin hydrochloride co-carried liposome and preparation method thereof
EP3265060B1 (en) Cochleates and methods of using the same to enhance tissue penetration of pharmacologically active agent
US10617672B2 (en) Liposome composition co-encapsulating doxorubicin and a prodrug of mitomycin C
EP4353223A1 (en) Application of pharmaceutical composition having specific drug-to-lipid ratio in antitumor
CN110548006A (en) Corosolic acid liposome and preparation method and application thereof
CN112957329B (en) Doxorubicin hydrochloride-forskolin co-loaded nano liposome as well as preparation method and application thereof
WO2018214944A1 (en) Long-circulating liposome modified with c(rgd-acp-k)
CN116407548A (en) Anthracycline antitumor drug and platinum antitumor drug composition and preparation method thereof

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: 16759478

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2977397

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017546067

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2016759478

Country of ref document: EP