CN111936147A - Gamma polyglutamated antifolate agent and its use - Google Patents

Gamma polyglutamated antifolate agent and its use Download PDF

Info

Publication number
CN111936147A
CN111936147A CN201980024669.5A CN201980024669A CN111936147A CN 111936147 A CN111936147 A CN 111936147A CN 201980024669 A CN201980024669 A CN 201980024669A CN 111936147 A CN111936147 A CN 111936147A
Authority
CN
China
Prior art keywords
composition
gamma
cancer
antifolate
pantol
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN201980024669.5A
Other languages
Chinese (zh)
Inventor
C·尼基萨
V·M·莫约
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
LEAF Holdings Group LLC
Original Assignee
LEAF Holdings Group LLC
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 LEAF Holdings Group LLC filed Critical LEAF Holdings Group LLC
Publication of CN111936147A publication Critical patent/CN111936147A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • A61K47/6913Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome the liposome being modified on its surface by an antibody
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • C07D475/06Heterocyclic compounds containing pteridine ring systems with a nitrogen atom directly attached in position 4
    • C07D475/08Heterocyclic compounds containing pteridine ring systems with a nitrogen atom directly attached in position 4 with a nitrogen atom directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The present disclosure relates generally to gamma polyglutamated antifolates, formulations containing liposomes filled with gamma polyglutamated antifolates, methods of making formulations containing the gamma polyglutamated antifolates and liposomes, and methods of treating hyperproliferative disorders (e.g., cancer) and immune system disorders (e.g., inflammation and autoimmune diseases, such as rheumatoid arthritis) using formulations containing gamma polyglutamated antifolates and liposomes.

Description

Gamma polyglutamated antifolate agent and its use
Background
The present disclosure generally relates to gamma polyglutamated antifolate compositions comprising a delivery vehicle, such as liposomes containing said gamma polyglutamated antifolate compositions; as well as methods of making and using the compositions to treat diseases including hyperproliferative diseases (such as cancer), immune system disorders (including inflammatory and autoimmune diseases, such as rheumatoid arthritis), and infectious diseases (such as HIV, malaria, and schistosomiasis).
Folate is an essential cofactor that mediates the transfer of one-carbon units involved in nucleotide biosynthesis and DNA repair, the re-methylation of homocysteine (Hcy), and the methylation of DNA, proteins, and lipids. The only circulating form of folate in the blood is monoglutamate, and folate monoglutamate is the only form of folate transported across the cell membrane-likewise, the monoglutamate form of polyglutamated antifolates is transported across cell membranes. Once taken up into the cell, intracellular folate is converted to polyglutamate by the enzyme folyl poly-gamma-glutamate synthetase (FPGS).
Antifolates are transported into cells via a Reducing Folate Carrier (RFC) system and Folate Receptors (FR) α and β and proton-coupled folate transporters (PCFTs), which are typically most active in lower pH environments. RFC is a major transporter for antifolates at physiological pH and is ubiquitously expressed in both normal and diseased cells. Thus, antifolate treatment often suffers from dose-limiting toxicity, which is a major obstacle to cancer chemotherapy. Once inside the cell, the antifolate is polyglutamated by FPGS, which can add up to 6 glutamyl groups in the L- γ carboxy linkage of the antifolate. FPGS have at least two main therapeutic objectives for L-gamma polyglutamation of antifolates: (1) it greatly enhances the affinity and inhibitory activity of antifolates on DHFR; and (2) it promotes the accumulation of polyglutamated antifolates, which, unlike antifolates (monoglutamates), are not readily transported out of the cell by the extracellular pumps.
While targeting folate metabolism and nucleotide biosynthesis are well established cancer treatment strategies, clinical efficacy is limited for antifolates due to lack of tumor selectivity and the presence of primary (de novo) and acquired resistance. Antifolates generally play a role in DNA and RNA synthesis and therefore have a more toxic role in rapidly dividing cells (e.g., malignant and myeloid cells). Myelosuppression is generally a dose-limiting toxicity of antifolate therapy and limits clinical application of antifolates.
Resistance to antifolate therapy is typically associated with one or more of the following: (a) increased extracellular pump activity, (b) decreased transport of the antifolate into the cell, (c) increased DHFR activity, (d) decreased activity of folyl poly-gamma-glutamate synthetase (FPGS), and (e) increased activity of gamma-glutamyl hydrolase (GGH) which cleaves the gamma polyglutamic acid chain linked to folic acid and the antifolate.
The challenge of long-term (> 30 years) observations, i.e., higher levels of polyglutamate of various antifolates having greater potency than lower levels of glutamate, has been that the scientific community relies on intracellular FPGS-mediated mechanisms to convert lower levels of glutamate into their higher level forms. The present invention provides a means to deliver higher levels of antifolates in the form of polyglutamates directly into cells without having to rely on cellular mechanisms to accomplish this.
The provided gamma polyglutamated antifolate compositions provide strategies for overcoming the pharmacological challenges associated with dose limiting toxicity and therapeutic resistance associated with antifolate therapy. In some embodiments, the provided methods deliver a gamma polyglutamated form of an antifolate to cancer cells while (1) minimizing/reducing exposure to normal tissue cells, (2) optimizing/improving the cytotoxic effects of antifolate-based agents on cancer cells, and (3) minimizing/reducing the impact of efflux pumps and other resistance mechanisms that limit the therapeutic efficacy of antifolates.
Disclosure of Invention
The present disclosure relates generally to gamma polyglutamated antifolate (gamma pantol) compositions and methods of making and using the compositions to treat diseases, including hyperproliferative diseases (such as cancer), immune system disorders (such as inflammation and rheumatoid arthritis), cardiovascular diseases (such as coronary artery disease), and infectious diseases (such as HIV, malaria, and schistosomiasis).
In some embodiments, the present disclosure provides:
[1] a composition comprising a gamma polyglutamated antifolate agent;
[2] the composition of [1], wherein the antifolate is selected from the group consisting of: pirtrexin, pralatrexate, AG2034, GW1843, and LY309887, or stereoisomers thereof;
[3] the composition of [1], wherein the antifolate is selected from the group consisting of: PMX, MTX, RTX and LTX, or stereoisomers thereof;
[4] the composition according to any one of [1] to [3], wherein the antifolate is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof;
[5] The composition of [1], wherein the antifolate is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89;
[6] the composition of any one of [1] to [5], wherein the gamma polyglutamated antifolate agent contains 4, 5, 2-10, 4-6, or more than 5 glutamyl groups;
[7] the composition of any one of [1] - [6], wherein the gamma polyglutamated antifolate agent:
(a) is a gamma-tetraglutaminic antifolate;
(b) is a gamma-pentaglutaminated antifolate; or
(c) Is a gamma-hexaglutaminated antifolate;
[8] the composition of any one of [1] to [7], wherein the gamma polyglutamated antifolate agent comprises 1-10 glutamyl groups having gamma carboxyl linkages;
[9] the composition according to any one of [1] to [8], wherein:
(a) at least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form,
(b) each glutamyl group of the gamma polyglutamated antifolate is in the L form,
(c) At least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form,
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate, or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form;
[10] the composition of any one of [1] to [9], wherein the polyglutamate is linear;
[11] the composition of any one of [1] to [9], wherein the polyglutamate is branched;
[12] a liposomal composition comprising a gamma polyglutamated antifolate (Lp-gamma pantol) according to any one of [1] to [11 ];
[13] the Lp- γ PANTIFOL composition of [12], wherein the polyglutamated antifolate agent is selected from the group consisting of:
(a) AG2034, pirtroxine, pralatrexate, GW1843, antifolate and LY 309887; or
(b) PMX, MTX, RTX and LTX, or stereoisomers thereof;
[14] the Lp- γ PANTIFOL composition of [12] or [13], wherein the polyglutamated antifolate agent is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and AG377, 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof;
[15] The Lp- γ PANTIFOL composition of any one of [12] to [14], wherein the antifolate is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89;
[16] the Lp- γ PANTIFOL composition of any one of [12] to [15], wherein the liposome comprises a gamma polyglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 gamma glutamyl groups;
[17] the Lp- γ PANTIFOL composition of any one of [12] to [16], wherein the liposome comprises a γ tetraglutamated antifolate;
[18] the Lp- γ PANTIFOL composition of any one of [12] to [16], wherein the liposome comprises a γ -pentaglutamated antifolate;
[19] the Lp- γ PANTIFOL composition of any one of [12] to [16], wherein the liposome comprises a γ hexaglutaminated antifolate;
[20] the Lp- γ PANTIFOL composition of any one of [12] - [19], wherein the gamma polyglutamated antifolate agent comprises 1-10 glutamyl groups having a gamma carboxyl linkage;
[21] The Lp- γ PANTIFOL composition according to any one of [12] to [20], wherein:
(a) at least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form;
(b) each glutamyl group of the gamma polyglutamated antifolate agent is in the L form;
(c) at least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form;
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate; or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form;
[22] the Lp-gamma PANTIFOL composition as claimed in any one of [12] to [21], wherein
(a) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form;
(b) each glutamyl group of the gamma polyglutamated antifolate agent is in the L form;
(c) at least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form;
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate; or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form;
[23] The Lp- γ PANTIFLOL composition of any one of [12] - [22], wherein the liposome is pegylated (PLp- γ PANTIFLOL);
[24] the Lp- γ PANTIFOL composition of any one of [12] to [22], wherein the liposome is non-pegylated;
[25] the Lp- γ PANTIFOL composition of any one of [12] to [24], wherein the liposome has a diameter in the range of 20nm to 200 nm;
[26] the Lp- γ PANTIFOL composition of any one of [12] to [25], wherein the liposome has a diameter in the range of 80nm to 120 nm;
[27] the Lp- γ PANTIFOL composition of any one of [12] to [26], wherein the liposome is formed from liposome components;
[28] the Lp- γ PANTIFOL composition of [27], wherein the liposomal composition comprises at least one of anionic lipids and neutral lipids;
[29] the Lp- γ PANTIFOL composition of [27] or [28], wherein the liposome component comprises at least one selected from the group consisting of: DSPE; DSPE-PEG; DSPE-PEG-maleimide; HSPC; HSPC-PEG; cholesterol; cholesterol-PEG; and cholesterol-maleimide.
[30] The Lp- γ PANTIFOL composition of any one of [27] - [29], wherein the liposome component comprises at least one selected from the group consisting of: DSPE; DSPE-PEG; DSPE-PEG-FITC; DSPE-PEG-maleimide; cholesterol; and HSPC;
[31] The Lp- γ PANTIFOL composition of any one of [27] to [30], wherein the one or more liposomal components further comprises a steric stabilizer;
[32] the Lp- γ PANTIFLOL composition of [31], wherein the steric stabilizer is at least one selected from the group consisting of: polyethylene glycol (PEG); poly-L-lysine (PLL); monosialoganglioside (GM 1); poly (vinyl pyrrolidone) (PVP); poly (acrylamide) (PAA); poly (2-methyl-2-oxazoline); poly (2-ethyl-2-oxazoline); a phosphatidylpolyglycerol; poly [ N- (2-hydroxypropyl) methacrylamide ]; amphiphilic poly-N-vinylpyrrolidone; an L-amino acid-based polymer; oligomerization of glycerol; copolymers comprising polyethylene glycol and polypropylene oxide; poloxamer 188; and polyvinyl alcohol;
[33] the Lp- γ PANTIFLOL composition of [32], wherein the steric stabilizer is PEG, and the PEG has a number average molecular weight (Mn) of 200 to 5000 daltons;
[34] the Lp- γ PANTIFOL composition of any one of [12] to [33], wherein the liposome is anionic or neutral;
[35] the Lp- γ PANTIFOL composition of any one of [12] to [33], wherein the liposome has a zeta potential less than or equal to zero;
[36] The Lp- γ PANTIFOL composition of any one of [12] - [33], wherein the liposome has a zeta potential between 0 to-150 mV;
[37] the Lp- γ PANTIFOL composition of any one of [12] - [33], wherein the liposome has a zeta potential between-30 to-50 mV;
[38] the Lp- γ PANTIFOL composition of any one of [12] to [33], wherein the liposome is cationic;
[39] the Lp- γ PANTIFOL composition of any one of [12] - [38], wherein the liposome has an interior space comprising the γ polyglutamated antifolate agent and a pharmaceutically acceptable aqueous carrier;
[40] the Lp- γ PANTIFOL composition of [39], wherein the pharmaceutically acceptable carrier comprises a tonicity agent such as dextrose, mannitol, glycerol, potassium chloride, sodium chloride at a concentration of greater than 1%;
[41] the Lp- γ PANTIFOL composition of [39], wherein the pharmaceutically acceptable aqueous carrier is trehalose;
[42] the Lp- γ PANTIFOL composition of [41], wherein the pharmaceutically acceptable carrier comprises 1% to 50% trehalose;
[43] the Lp- γ PANTIFOL composition of any one of [39] - [42], wherein the pharmaceutically acceptable carrier comprises a dextrose solution of 1% to 50%;
[44] The Lp- γ PANTIFOL composition of any one of [39] - [43], wherein the interior space of the liposome comprises 5% dextrose suspended in HEPES buffer solution;
[45] the Lp- γ PANTIFOL composition of any one of [39] - [44], wherein the pharmaceutically acceptable carrier comprises a buffer, such as HEPES Buffered Saline (HBS) or the like, at a concentration of between 1 and 200mM and a pH of between 2 and 8;
[46] the Lp- γ PANTIFOL composition of any one of [39] - [45], wherein the pharmaceutically acceptable carrier comprises sodium acetate and calcium acetate in a total concentration between 50mM to 500 mM;
[47] the Lp- γ PANTIFOL composition of any one of [12] to [46], wherein the interior space of the liposome has a pH of 5-8 or a pH of 6-7, or any range therebetween;
[48] the Lp- γ PANTIFOL composition of any one of [12] - [47], wherein the liposome comprises less than 500,000 or less than 200,000 of the γ polyglutamated antifolate molecules;
[49] the Lp- γ PANTIFOL composition of any one of [12] - [48], wherein the liposomes comprise between 10 to 100,000 of the γ polyglutamated antifolate molecules, or any range therebetween;
[50] The Lp- γ PANTIFLOL composition of any one of [12] to [49], further comprising a targeting moiety, and wherein the targeting moiety has specific affinity for a surface antigen on a target cell of interest;
[51] the Lp- γ PANTIFLOL composition of [50], wherein the targeting moiety is attached to one or both of PEG and the exterior of the liposome, optionally wherein targeting moiety is attached to one or both of the PEG and the exterior of the liposome by a covalent bond;
[52] the Lp- γ PANTIFLOL composition of [50] or [51], wherein the targeting moiety is a polypeptide;
[53] the Lp- γ PANTIFLOL composition of any one of [50] - [52], wherein the targeting moiety is an antibody or an antigen-binding fragment of an antibody;
[54]according to [50]-[53]The Lp-gamma PANTIFOL composition of any one of the above, wherein the use is as follows
Figure BDA0002715408560000061
Analyzing the assay, the targeting moiety to bind to the target at 0; 5x 10-10 to 10x 10-6An equilibrium dissociation constant (Kd) in a range binds to the surface antigen;
[55] the Lp- γ PANTIFOL composition of any one of [50] - [54], wherein the targeting moiety specifically binds to one or more folate receptors selected from the group consisting of: folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and folate receptor (FR-);
[56] The Lp- γ PANTIFLOL composition of any one of [50] - [55], wherein the targeting moiety comprises one or more selected from the group consisting of: antibodies, humanized antibodies, antigen-binding fragments of antibodies, single chain antibodies, single domain antibodies, bispecific antibodies, synthetic antibodies, pegylated antibodies, and multimeric antibodies;
[57] the Lp- γ PANTIFOL composition of any one of [50] - [56], wherein each pegylated liposome comprises 1 to 1000 or 30 to 200 targeting moieties;
[58] the Lp- γ PANTIFLOL composition of any one of [39] - [57], further comprising one or more of an immunostimulatory agent, a detectable label, and a maleimide, wherein the immunostimulatory agent, the detectable label, or the maleimide is attached to the PEG or the exterior of the liposome;
[59] the Lp- γ PANTIFLOL composition of [58], wherein the immunostimulatory agent is at least one selected from the group consisting of: a protein immunostimulant; a nucleic acid immunostimulant; a chemical immunostimulant; a hapten; and an adjuvant;
[60] the Lp- γ PANTIFLOL composition of [58] or [59], wherein the immunostimulant is at least one selected from the group consisting of: fluorescein; fluorescein Isothiocyanate (FITC); DNP; beta glucan; beta-1, 3-glucan; beta-1, 6-glucan; resolvin (resolvin) (e.g., resolvin D such as Dn-6DPA or Dn-3DPA, resolvin E, or T series resolvin); and Toll-like receptor (TLR) modulators, such as oxidized low density lipoproteins (e.g., OXPAC, PGPC) and eritoran lipids (e.g., E5564);
[61] The Lp- γ PANTIFOL composition of any one of [58] to [60], wherein the immunostimulatory agent and the detectable label are the same;
[62] the Lp- γ PANTIFLOL composition of any one of [58] to [61], further comprising a hapten;
[63] the Lp- γ PANTIFLOL composition of [62], wherein the hapten comprises one or more of fluorescein or β 1, 6-glucan;
[64] the Lp- γ PANTIFLOL composition of any one of [12] - [63], further comprising at least one cryoprotectant selected from the group consisting of: mannitol; trehalose; sorbitol; and sucrose;
[65] a targeting composition comprising the composition according to any one of [1] to [64 ];
[66] a targeting composition comprising the composition according to any one of [1] to [49 ];
[67] the Lp- γ PANTIFOL composition of any one of [12] - [66], further comprising carboplatin and/or pembrolizumab (pembrolizumab);
[68] a pharmaceutical composition comprising a liposomal gamma polyglutamated antifolate composition according to any one of [12] to [67 ];
[69] A pharmaceutical composition comprising a gamma polyglutamated antifolate composition according to any one of [1] to [7 ];
[70] a composition as described in any one of [1] to [69], for use in the treatment of a disease;
[71] use of a composition as defined in any one of [1] to [70] in the manufacture of a medicament for the treatment of a disease;
[72] a method for treating or preventing a disease in a subject in need of such treatment or prevention, the method comprising administering to the subject a composition as described in any one of [1] to [70 ];
[73] a method for treating or preventing a disease in a subject in need of such treatment or prevention, the method comprising administering to the subject a liposomal gamma polyglutamated antifolate composition of any one of [12] to [69 ];
[74] a method of killing a hyperproliferative cell, said method comprising contacting a hyperproliferative cell with a composition as described in any of [1] to [69 ];
[75] a method of killing hyperproliferative cells, said method comprising contacting hyperproliferative cells with a liposomal gamma polyglutamated antifolate composition of any one of [12] to [69 ];
[76] The method of [74] or [75], wherein the hyperproliferative cell is a cancer cell, a mammalian cell, and/or a human cell;
[77] a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of the composition of any one of [1] to [69 ];
[78] a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [12] to [68 ];
[79] the method of [77] or [78], wherein the cancer is selected from: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dyscrasias;
[80] the method of [77] or [78], wherein the cancer is selected from: lung, breast, colon, pancreatic, gastric, bladder, head and neck, ovarian and cervical cancer;
[81] The method of [77] or [78], wherein the cancer is selected from: colorectal, lung, breast, head and neck, and pancreatic cancers;
[82] the method of [77] or [78], wherein the cancer is selected from: colorectal cancer, breast cancer, ovarian cancer, lung cancer, head and neck cancer, pancreatic cancer, gastric cancer, and mesothelioma;
[83] a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer cells expressing on their surface folate receptors bound by a targeting moiety an effective amount of an Lp- γ pantol composition according to any one of [50] - [66 ];
[84] a maintenance therapy for a subject undergoing or having undergone cancer therapy, the maintenance therapy comprising administering to the subject undergoing or having undergone cancer therapy an effective amount of the composition of any one of [1] - [69 ];
[85] a maintenance therapy for a subject undergoing or having undergone cancer therapy, said maintenance therapy comprising administering to a subject undergoing or having undergone cancer therapy an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [12] - [69 ];
[86] A method for treating an immune system disorder, the method comprising administering to a subject having or at risk of having an immune system disorder an effective amount of the composition of any one of [1] - [69], optionally wherein the immune system disorder is selected from: inflammation (e.g., acute and chronic inflammation), systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, systemic lupus erythematosus and takayasu's arteritis, and psoriasis;
[87] a method for treating an immune system disorder, the method comprising administering to a subject having or at risk of having an immune system disorder an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [8] - [69], optionally wherein the immune system disorder is selected from: inflammation (e.g., acute and chronic inflammation), systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, systemic lupus erythematosus and takayasu's arteritis, and psoriasis;
[88] a method for treating the following diseases:
(a) an infectious disease, the method comprising administering to a subject having or at risk of having an infectious disease an effective amount of the composition according to any one of [1] to [69 ];
(b) An infectious disease, a cardiovascular disease, a metabolic disease, or another disease, the method comprising administering to a subject having or at risk of having an infectious disease, a cardiovascular disease, or another disease an effective amount of a composition according to any one of [1] - [69], wherein the disease is a member selected from: atherosclerosis, cardiovascular disease (CVD), coronary artery disease, myocardial infarction, stroke, metabolic syndrome, gestational trophoblastic disease, and ectopic pregnancy;
(c) an autoimmune disease, the method comprising administering to a subject having or at risk of having an autoimmune disease an effective amount of a composition according to any one of [1] to [69 ];
(d) rheumatoid arthritis, the method comprising administering to a subject having or at risk of having rheumatoid arthritis an effective amount of the composition according to any one of [1] to [69 ];
(e) an inflammatory disorder, the method comprising administering to a subject having or at risk of having inflammation an effective amount of a composition according to any one of [1] - [69], optionally wherein the inflammation is acute, chronic and/or systemic inflammation; or
(f) A skin condition, the method comprising administering to a subject having or at risk of having a skin condition an effective amount of the composition according to any one of [1] - [69], optionally wherein the skin condition is psoriasis;
[89] a method for treating an infectious disease, the method comprising administering to a subject having or at risk of having an infectious disease an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [12] to [69 ];
[90] a method of delivering a gamma polyglutamated antifolate to a tumor expressing a folate receptor on the surface, the method comprising: administering to a subject having the tumor an amount of an Lp- γ PANTIFOL composition of any one of [1] - [69] to deliver a therapeutically effective dose of a γ polyglutamated antifolate to the tumor;
[91] a method of making a gamma polyglutamated antifolate composition comprising a liposomal gamma polyglutamated antifolate composition according to any one of [12] to [69], the method comprising: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; and processing the mixture to form liposomes containing a gamma polyglutamated antifolate agent;
[92] A method of making the composition of any one of [12] to [69], the method comprising the steps of: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; processing the mixture to form liposomes encapsulating and/or encapsulating the gamma polyglutamated antifolate agent; and providing a targeting moiety on the surface of the liposome, the targeting moiety having specific affinity for at least one of folate receptor alpha (FR-a), folate receptor beta (FR- β), and folate receptor (FR-);
[93] the method of [92], wherein the processing step comprises one or more of: film hydration, extrusion, online mixing, an ethanol injection technology, a freeze thawing technology, reversed phase evaporation, dynamic high-pressure micro-jet, micro-jet mixing, a multiple emulsion method, a freeze drying multiple emulsion method, 3D printing, a membrane contactor method and stirring; and/or
[94] The method of [92], wherein the processing step comprises one or more steps of altering the size of the liposomes by one or more of extrusion, high pressure microfluidization, and/or sonication steps.
In some embodiments, the present disclosure provides a gamma polyglutamated antifolate (gamma pantol) composition, wherein at least 2 glutamyl residues of the gamma polyglutamated antifolate have a gamma carboxyl linkage. In some embodiments, the gamma pantol contains 2-20, 2-15, 2-10, 2-5, or more than 5 glutamyl groups (including glutamyl groups of antifolates). In some embodiments, the gamma polyglutamated antifolate agent is selected from the group consisting of: (a) AG2034, pirtroxine, pralatrexate, GW1843, antifolate and LY 309887; or (b) PMX, MTX, RTX and LTX, or stereoisomers thereof. In some embodiments, the gamma polyglutamated antifolate agent is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and AG377, 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof. In some embodiments, the gamma polyglutamated antifolate agent is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89. In some embodiments, the γ pantol comprises two or more glutamyl groups in the form of L. In other embodiments, the γ pantol comprises a glutamyl group in D form. In other embodiments, the γ pantol comprises a glutamyl group in D form and two or more glutamyl groups in L form.
In one embodiment, the gamma pantol composition contains a chain of 3 glutamyl groups linked to a glutamyl group in the antifolate (i.e., gamma tetraglutamated antifolate). In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the tetraglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, the tetraglutamated antifolate comprises a glutamyl group in the D form. In some embodiments, the tetraglutamated antifolate comprises two or more glutamyl groups in the D form. In other embodiments, the tetraglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form. In some embodiments, the tetraglutamated antifolate comprises one, two or three glutamyl groups in the D form and three, two or one glutamyl groups in the L form, respectively.
In one embodiment, the gamma pantol composition contains a chain of 4 gamma-glutamyl groups linked to a glutamyl group in the antifolate (i.e., gamma pentaglutamated antifolate). In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the pentaglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, the pentaglutamated antifolate comprises a glutamyl group in the D form. In some embodiments, the pentaglutamated antifolate comprises two or more glutamyl groups in D form. In other embodiments, the pentaglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form. In some embodiments, the pentaglutamated antifolate comprises one, two, three or four glutamyl groups in D form and four, three, two or one glutamyl groups in L form, respectively.
In one embodiment, the gamma pantol composition contains a chain of 5 gamma-glutamyl groups linked to a glutamyl group in the antifolate (i.e., a gamma hexaglutamylated antifolate). In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the hexaglutaminated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, the hexaglutamated antifolate comprises a glutamyl group in the D form. In some embodiments, the hexaglutaminated antifolate comprises two or more glutamyl groups in the D form. In other embodiments, the hexaglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form. In some embodiments, the pentaglutamated antifolate comprises one, two, three, four or five glutamyl groups in D form and five, four, three, two or one glutamyl group in L form, respectively.
In further embodiments, the present disclosure provides compositions containing a delivery vehicle such as liposomes filled with (i.e., encapsulating) and/or otherwise associated with a gamma polyglutamated antifolate agent; as well as methods of making and methods of using the gamma PANTIFLO-populated/associated delivery vehicle compositions (DV-gamma PANTIFLO) to deliver gamma-polyglutamated antifolates to diseased (e.g., cancerous) and/or targeted cells. These compositions have uses including, but not limited to, the treatment of diseases including, for example, hyperproliferative diseases (such as cancer), immune system disorders (such as inflammation and rheumatoid arthritis), and infectious diseases (such as HIV, malaria, and schistosomiasis). The γ pantol bulking/associating delivery vehicle composition provides an improvement in efficacy and safety of delivery of antifolates to cancer cells by providing a more cytotoxic payload (e.g., polyglutamated antifolates) compared to the cytotoxicity of Antifolates (ANTIFOLs) administered in their monoglutamate state. In some embodiments, the gamma polyglutamated antifolate agent in said DV-gamma pantol contains 2-20, 2-15, 2-10, 2-5, more than 5, or more than 20 glutamyl groups (including glutamyl groups of antifolate agents). In some embodiments, the delivery vehicle contains a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention section. In some embodiments, the delivery vehicle contains a polyglutamated antifolate agent described in the summary section. In some embodiments, the delivery vehicle is a liposome according to any one of [12] to [67] of the summary of the invention section.
In a further embodiment, the present disclosure provides a composition comprising liposomes (Lp- γ pantol) encapsulating (filled with) a gamma polyglutamated antifolate agent. In some embodiments, the gamma polyglutamated antifolate agent in Lp-gamma pantol contains 2-20, 2-15, 2-10, 2-5, or more than 20 glutamyl groups (including glutamyl groups in antifolate agents). In some embodiments, the gamma polyglutamated antifolate agent encapsulated by the liposomes is selected from the group consisting of: (a) AG2034, pirtroxine, pralatrexate, GW1843, antifolate and LY 309887; or (b) PMX, MTX, RTX and LTX, or stereoisomers thereof. In some embodiments, the gamma polyglutamated antifolate agent encapsulated by the liposomes is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and AG377, 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof. In some embodiments, the gamma polyglutamated antifolate agent encapsulated by the liposomes is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC945(ONX-0801), CB300638 and BW1843U 89. In some embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises two or more glutamyl groups in the L form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in D form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In one embodiment, the Lp- γ pantol composition comprises a gamma-polyglutamated antifolate containing a chain of 3 glutamyl groups linked to a glutamyl group in the antifolate (i.e., a tetraglutamated antifolate). In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the tetraglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, the tetraglutamated antifolate comprises a glutamyl group in the D form. In other embodiments, the tetraglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In one embodiment, the Lp- γ pantol composition comprises a γ polyglutamated antifolate comprising a chain of 4 γ -glutamyl groups linked to glutamyl groups in the antifolate (i.e., a γ -pentaglutamated antifolate). In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the gamma pentaglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, the pentaglutamated antifolate comprises a glutamyl group in the D form. In other embodiments, the pentaglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In one embodiment, the Lp- γ pantol composition comprises a γ polyglutamated antifolate comprising a chain of 5 γ -glutamyl groups linked to glutamyl groups in the antifolate (i.e., a γ -hexaglutamated antifolate). In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the gamma hexaglutaminated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, the gamma hexaglutaminated antifolate comprises a glutamyl group in the D form. In other embodiments, the gamma hexaglutaminated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the Lp- γ pantol composition is cationic. In some embodiments, the Lp- γ pantol liposomes are cationic and have a diameter in the range of 20nm to 200nm, 30nm to 175nm, or 50nm to 150nm, or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are cationic and have a diameter in the range of 30nm to 175nm or 50nm to 150nm or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are cationic and the composition has a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the cationic Lp- γ pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-entrapped gamma tetraglutamylated antifolate agent. In some embodiments, the cationic Lp- γ pantol composition comprises at least 11%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-entrapped gamma-pentaglutaminated antifolate agent. In other embodiments, the Lp- γ pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma hexaglutamylated antifolate. In further embodiments, the gamma polyglutamated antifolate encapsulated by the liposome is located in HEPES buffer within the liposome.
In other embodiments, the Lp- γ pantol composition is anionic or neutral. In some embodiments, the Lp- γ pantol composition is cationic. In some embodiments, the Lp- γ pantol liposome is anionic or neutral and has a diameter in the range of 20nm to 200nm, 30nm to 175nm, or 50nm to 150nm, or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are anionic or neutral and have a diameter in the range of 30nm to 175nm or 50nm to 150nm or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are anionic or neutral and the composition has a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the Lp- γ pantol liposome is anionic and has a diameter in the range of 20nm to 200nm, 30nm to 175nm, or 50nm to 150nm, or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are anionic and have a diameter in the range of 30nm to 175nm or 50nm to 150nm or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are anionic and the composition has a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the Lp- γ pantol liposomes are neutral and have a diameter in the range of 20nm to 200nm, 30nm to 175nm, or 50nm to 150nm, or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are neutral and have a diameter in the range of 30nm to 175nm or 50nm to 150nm or any range therebetween. In other embodiments, the Lp- γ pantol liposomes are neutral and the composition has a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the anionic or neutral Lp- γ pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded γ -tetraglutamylated antifolate agent. In some embodiments, the anionic or neutral Lp- γ pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-entrapped gamma-pentaglutamated antifolate agent. In other embodiments, the Lp- γ pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma hexaglutamylated antifolate. In further embodiments, the gamma polyglutamated antifolate encapsulated by the liposome is located in HEPES buffer within the liposome.
In a further embodiment, the liposomal gamma polyglutamated antifolate composition is pegylated (PLp-gamma pantol).
In some embodiments, the liposomal gamma polyglutamated antifolate composition is non-targeted (NTLp-gamma pantol). That is, the NTLp- γ pantol composition has no specific affinity for an epitope expressed on the surface of the target cell of interest (e.g., an epitope on a surface antigen). In some embodiments, the NTLp- γ pantol composition does not comprise a targeting moiety. In other embodiments, the non-targeted liposomal gamma polyglutamated antifolate composition is pegylated (NTPLp-gamma pantol).
In other embodiments, the liposomal gamma polyglutamated antifolate composition is targeted (TLp-gamma PANTIFOL). That is, the TLp- γ PANTIFLOL composition contains a targeting moiety with specific affinity for an epitope (surface antigen) on a target cell of interest. In some embodiments, the TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL targeting moiety is not covalently attached to the liposome. In other embodiments, the targeting moiety of TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL is attached to one or both of PEG and the outside of the liposome. In some embodiments, the targeting moiety of TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL is attached to the liposome by a covalent bond. The functions of the targeting moiety of the TLp- γ PANTIFLOL and/or TPLp- γ PANTIFLOL compositions include, but are not limited to, targeting liposomes to target cells of interest in vivo or in vitro; or a surface antigen for which the targeting moiety has specific affinity, and delivers a liposomal payload (γ pantol) into the cell. Suitable targeting moieties are known in the art and include, but are not limited to, antibodies, antigen-binding antibody fragments, scaffold proteins, polypeptides and peptides. In some embodiments, the targeting moiety is a polypeptide. In other embodiments, the targeting moiety is a polypeptide comprising at least 3, 5, 10, 15, 20, 30, 40, 50, or 100 amino acid residues.
Targeted liposomal gamma-polyglutamated antifolate compositions (TLp-gamma-pantol and TPLp-gamma-pantol) provide further improvements in the efficacy and safety profile of antifolates by specifically delivering gamma-polyglutamated (e.g., gamma-pentaglutamated and/or gamma-hexaglutamated) antifolates to target cells such as cancer cells. In other embodiments, the targeted liposomal gamma polyglutamated antifolate composition is pegylated (TPLp-gamma pantol). In some embodiments, the targeting moiety of TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL is attached to one or both of PEG and the outside of the liposome. In some embodiments, the targeting moiety of TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL is attached to the liposome by a covalent bond. γ pantol). The functions of the targeting moiety of the TLp- γ PANTIFLOL and/or TPLp- γ PANTIFLOL compositions include, but are not limited to, targeting liposomes to target cells of interest in vivo or in vitro; or a surface antigen for which the targeting moiety has specific affinity, and delivers a liposomal payload (γ pantol) into the cell. Suitable targeting moieties are known in the art and include, but are not limited to, antibodies, antigen-binding antibody fragments, scaffold proteins, polypeptides and peptides. In some embodiments, the targeting moiety is a polypeptide. In other embodiments, the targeting moiety is a polypeptide comprising at least 3, 5, 10, 15, 20, 30, 40, 50, or 100 amino acid residues.
In some embodiments, the targeting moiety of TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL is an antibody or antigen-binding antibody fragment. In other embodiments, the targeting moiety comprises one or more of an antibody, a humanized antibody, an antigen-binding fragment of an antibody, a single chain antibody, a single domain antibody, a bispecific antibody, a synthetic antibody, a pegylated antibody, and a multimeric antibody. In some embodiments, the TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL targeting moiety has specific affinity for an epitope preferentially expressed on a target cell, such as a tumor cell, as compared to a normal or non-tumor cell. In some embodiments, the targeting moiety has specific affinity for an epitope on a tumor cell surface antigen that is present on a tumor cell but absent or inaccessible on a non-tumor cell. In some embodiments, e.g., using
Figure BDA0002715408560000131
The targeting moiety is measured by analysis to be at 0.5x 10-10To 10x 10-6Equilibrium dissociation constants (Kd) within the range bind to the target epitope.
In particular embodiments, the TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL targeting moiety comprises a polypeptide that specifically binds to a folate receptor. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the folate receptor bound by the targeting moiety is one or more folate receptors selected from the group consisting of: folate receptor alpha (FR-alpha, FOLR1), folate receptor beta (FR-beta, FOLR2) and folate receptor (FR-, FOLR 4). In some embodiments, the folate receptor bound by the targeting moiety is folate receptor alpha (FR-alpha). In some embodiments, the folate receptor bound by the targeting moiety is folate receptor beta (FR- β). In some embodiments, the targeting moiety specifically binds FR- α and FR- β.
In further embodiments, the Lp- γ pantol composition comprises one or more of an immunostimulatory agent, a detectable label, and a maleimide disposed on at least one of the PEG and the exterior of the liposome. In some embodiments, the liposomal gamma pantol composition (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol) is cationic. In other embodiments, the liposomal gamma pantol composition (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol) is anionic or neutral. In further embodiments, the liposomes of the liposomal gamma pantol composition (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol) have a diameter in the range of 20nm to 200nm or any range therebetween. In other embodiments, the liposomes of the liposomal gamma pantol composition have a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the liposomal gamma pantol composition is pegylated (e.g., PLp-gamma pantol, NTPLp-gamma pantol, or TPLp-gamma pantol). In some embodiments, the liposomal gamma pantol composition comprises a targeting moiety (e.g., TLp-gamma pantol or TPLp-gamma pantol). In other embodiments, the liposomal gamma pantol composition is pegylated and targeted (e.g., TPLp-gamma pantol). In some embodiments, the liposomal gamma pantol composition comprises a gamma polyglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomal γ pantol composition comprises a γ -tetraglutamated antifolate. In some embodiments, the liposomal γ pantol composition comprises a γ -pentaglutaminated antifolate agent. In other embodiments, the liposomal γ pantol composition comprises a γ -hexaglutaminated antifolate agent. In some embodiments, the liposome composition comprises a gamma polyglutamated antifolate agent as in any one of [1] to [11] of the summary of the invention. In some embodiments, the liposome comprises a liposome composition according to any one of [11] to [69] of the summary of the invention section. In some embodiments, the compositions comprise a gamma polyglutamated antifolate agent described in the summary of the invention section.
In additional embodiments, the liposomal gamma panto-fol composition (i.e., Lp-gamma pantol, such as PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol) comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma polyglutamated antifolate agent. In some embodiments, the liposomal gamma pantol composition comprises 1% to 98.5% liposome-embedded gamma polyglutamated antifolate. In further embodiments, the liposomal gamma pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma polyglutamated antifolate containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomal gamma pantol composition comprises 1% to 98.5% liposome-entrapped gamma polyglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomal gamma pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma tetraglutamylated antifolate. In some embodiments, the liposomal gamma pantol composition comprises 1% to 98.5% liposome-embedded gamma tetraglutamated antifolate. In some embodiments, the liposomal gamma pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma-pentaglutaminated antifolate agent. In some embodiments, the liposomal gamma pantol composition comprises 1% to 98.5% liposome-embedded gamma pentaglutamated antifolate. In some embodiments, the liposomal gamma pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma hexaglutamylated antifolate. In some embodiments, the liposomal gamma pantol composition comprises 1% to 98.5% liposome-embedded gamma pentaglutamated antifolate. In some embodiments, the liposome composition comprises a gamma polyglutamated antifolate agent as in any one of [1] to [11] of the summary of the invention. In some embodiments, the liposome comprises a liposome composition according to any one of [11] to [69] of the summary of the invention section. In some embodiments, the compositions comprise a gamma polyglutamated antifolate agent as described in the summary of the invention section or figures herein.
Also provided are liposome compositions comprising liposomes encapsulating gamma PANTIFOL. In some embodiments, the liposome composition comprises a pegylated gamma pantol composition. In some embodiments, the liposome composition comprises a γ pantol composition linked to or otherwise associated with a targeting moiety. In other embodiments, the liposome composition comprises a γ pantol composition that is pegylated and linked or otherwise associated with a targeting moiety. In some embodiments, the liposome composition comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposome composition comprises a gamma-tetraglutamated antifolate. In some embodiments, the liposome composition comprises a gamma-pentaglutaminated antifolate. In other embodiments, the liposome composition comprises a gamma-hexaglutaminated antifolate. In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section.
In some embodiments, the liposome composition comprises liposomal gamma pantol (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, and TPLp-gamma pantol). In some embodiments, the liposomal gamma pantol is pegylated (e.g., NTPLp-gamma pantol and TPLp-gamma pantol). In some embodiments, the pharmaceutical composition comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the pharmaceutical composition comprises a gamma-tetraglutamated antifolate. In some embodiments, the pharmaceutical composition comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the pharmaceutical composition comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposomal gamma pantol comprises a targeting moiety (e.g., TLp-gamma pantol or TPLp-gamma pantol) having specific affinity for an epitope of an antigen on the surface of a target cell of interest, such as a cancer cell. In other embodiments, the liposome composition comprises pegylated liposomal gamma pantol, and further comprises a targeting moiety (e.g., TPLp-gamma pantol) having specific affinity for an epitope of an antigen on the surface of a target cell of interest, such as a cancer cell. In some embodiments, the liposome composition comprises a cationic liposomal γ pantol. In other embodiments, the liposome composition comprises an anionic or neutral liposomal γ pantol. In further embodiments, the liposome composition comprises a liposomal γ pantol having a diameter in the range of 20nm to 200nm or any range therebetween. In other embodiments, the liposomal γ pantol has a diameter in the range of 80nm to 120nm or any range therebetween.
Also provided are pharmaceutical compositions comprising a gamma polyglutamated antifolate agent (gamma pantol) comprising a delivery vehicle, such as liposomal gamma pantol. In some embodiments, the pharmaceutical composition comprises a pegylated gamma pantol composition. In some embodiments, the pharmaceutical composition comprises a gamma pantol composition linked to or otherwise associated with a targeting moiety. In other embodiments, the pharmaceutical composition comprises a γ pantol composition that is pegylated and linked to or otherwise associated with a targeting moiety. In some embodiments, the pharmaceutical composition comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the pharmaceutical composition comprises a gamma-tetraglutamated antifolate. In some embodiments, the pharmaceutical composition comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the pharmaceutical composition comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the gamma polyglutamated antifolate agent is a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the gamma polyglutamated antifolate agent is a polyglutamated antifolate agent described in the summary of the invention section.
In some embodiments, the pharmaceutical composition comprises liposomal gamma pantol (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, and TPLp-gamma pantol). In some embodiments, the liposomal gamma pantol composition is pegylated (e.g., NTPLp-gamma pantol and TPLp-gamma pantol). In some embodiments, the liposomal gamma pantol comprises a targeting moiety (e.g., TLp-gamma pantol or TPLp-gamma pantol) having specific affinity for an epitope of an antigen on the surface of a target cell of interest, such as a cancer cell. In other embodiments, the pharmaceutical composition comprises a pegylated liposomal γ pantol composition, and further comprises a targeting moiety (e.g., TPLp- γ pantol) having specific affinity for an epitope of an antigen on the surface of a target cell of interest, such as a cancer cell. In some embodiments, the pharmaceutical composition comprises a cationic liposomal γ pantol. In other embodiments, the pharmaceutical composition comprises an anionic or neutral liposomal γ pantol. In a further embodiment, the pharmaceutical composition comprises a liposomal γ pantol having a diameter in the range of 20nm to 200nm or any range therebetween. In other embodiments, the liposomal gamma pantol composition has a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the pharmaceutical composition comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the pharmaceutical composition comprises a gamma-tetraglutamated antifolate. In some embodiments, the pharmaceutical composition comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the pharmaceutical composition comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the compositions comprise a gamma polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the pharmaceutical composition comprises a liposome composition according to any one of [11] to [69] of the summary of the invention section. In some embodiments, the compositions comprise a gamma polyglutamated antifolate agent described in the summary of the invention section.
In additional embodiments, the present disclosure provides a method of modulating activation, chemokine production, or metabolic activity of a cell, comprising contacting the cell with a composition comprising a gamma polyglutamated antifolate (gamma pantol) composition. In some embodiments, the contacted cell is a mammalian cell. In other embodiments, the contacted cell is a human cell. In some embodiments, the contacted cell is a hyperproliferative cell. In other embodiments, the cell is an immune cell. In some embodiments, the method is performed in vivo. In other embodiments, the method is performed in vitro. In some embodiments, the γ pantol contains 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the gamma pantol composition comprises a gamma tetraglutamated antifolate. In some embodiments, the gamma pantol composition comprises a gamma pentaglutaminated antifolate agent. In other embodiments, the gamma pantol composition comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the compositions comprise a gamma polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the pharmaceutical composition comprises a liposome composition according to any one of [11] to [69] of the summary of the invention section. In some embodiments, the compositions comprise a gamma polyglutamated antifolate agent as described in the summary of the invention section or figures herein.
In additional embodiments, the present disclosure provides a method of modulating activation, chemokine production, or metabolic activity of a cell, comprising contacting the cell with a liposome comprising a gamma polyglutamated antifolate (gamma pantol) composition. In some embodiments, the contacted cell is a mammalian cell. In other embodiments, the contacted cell is a human cell. In some embodiments, the contacted cell is a hyperproliferative cell. In other embodiments, the cell is an immune cell. In some embodiments, the method is performed in vivo. In other embodiments, the method is performed in vitro. In some embodiments, the γ pantol contains 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the gamma pantol composition comprises a gamma tetraglutamated antifolate. In some embodiments, the gamma pantol composition comprises a gamma pentaglutaminated antifolate agent. In other embodiments, the gamma pantol composition comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the polyglutamated antifolate is an antifolate listed in [2] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [3] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [4] of the summary of the invention. In some embodiments, the polyglutamated antifolate is an antifolate listed in [5] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section.
In additional embodiments, the present disclosure provides a method of killing a cell, the method comprising contacting the cell with a composition comprising a gamma polyglutamated antifolate (gamma pantol) composition. In some embodiments, the contacted cell is a mammalian cell. In other embodiments, the contacted cell is a human cell. In some embodiments, the contacted cell is a hyperproliferative cell. In other embodiments, the hyperproliferative cell is a cancer cell. In other embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a cancer selected from: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dysplasias or dyscrasias. In some embodiments, the cancer is selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from colorectal cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from ovarian cancer. In some embodiments, the contacted cancer cells are primary cells or cells from a cell line obtained/derived from endometrial cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from pancreatic cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from liver cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a head and neck cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from osteosarcoma. In some embodiments, the method is performed in vivo. In other embodiments, the method is performed in vitro. In some embodiments, the gamma pantol contains 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the gamma pantol composition comprises a gamma tetraglutamated antifolate. In some embodiments, the gamma pantol composition comprises a gamma pentaglutaminated antifolate agent. In other embodiments, the gamma pantol composition comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the gamma polyglutamated antifolate agent is a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the gamma polyglutamated antifolate agent is a polyglutamated antifolate agent described in the summary of the invention section.
In additional embodiments, the present disclosure provides a method of killing a cell comprising contacting the cell with a liposome containing a gamma-polyglutamated antifolate (e.g., Lp-gamma pantol, such as PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol). In some embodiments, the contacted cell is a mammalian cell. In other embodiments, the contacted cell is a human cell. In some embodiments, the contacted cell is a hyperproliferative cell. In other embodiments, the hyperproliferative cell contacted is a cancer cell. In other embodiments, the cancer cell is a primary cell or a cell from a cell line obtained/derived from a cancer selected from: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dysplasias or dyscrasias. In some embodiments, the cell is a primary cell or a cell from a cell line obtained/derived from a cancer selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from colorectal cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from ovarian cancer. In some embodiments, the contacted cancer cells are primary cells or cells from a cell line obtained/derived from endometrial cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from pancreatic cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from liver cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a head and neck cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from osteosarcoma. In some embodiments, the method is performed in vivo. In other embodiments, the method is performed in vitro. In some embodiments, the liposome contains gamma pantol, which contains 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomes contain a gamma-tetraglutamated antifolate. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In some embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the gamma polyglutamated antifolate agent is a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome is a liposome according to any one of [12] to [67] of the summary of the invention section.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a delivery vehicle (e.g., an antibody immunoconjugate or liposome) comprising a gamma polyglutamated antifolate. In some embodiments, the delivery vehicle is an antibody-containing immunoconjugate (including, e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the administered delivery vehicle comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of the cancer cell. In further embodiments, the delivery vehicle comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the delivery vehicle comprises a targeting moiety that specifically binds to one or more cell surface antigens (such as neoantigens) derived from or determined to be expressed on a cancer (tumor) in a particular subject. In some embodiments, the targeting moiety specifically binds to one or more cell surface antigens (such as neoantigens) derived from or determined to be expressed on a tumor in a particular subject. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the cancer is selected from: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcoma, brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dysplasias or dyscrasias. In some embodiments, the cancer is selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention section. In some embodiments, the delivery vehicle comprises a polyglutamated antifolate agent described in the summary section. In some embodiments, the delivery vehicle administered is a liposome composition comprising a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention section. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of liposomes comprising a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, such as PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol or TPLp-gamma pantol). In some embodiments, the liposome is pegylated. In some embodiments, the liposome is non-pegylated. In additional embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of a cancer cell. In further embodiments, the liposome comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. Also included are uses of cancer stem cell targeting moieties, such as those targeting CD34, CD133 and CD44, CD138, and CD 15. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the liposome comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposome comprises a gamma-tetraglutamated antifolate. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the polyglutamated antifolate agent is an antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section. In some embodiments, the liposome comprises a L γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises a D γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered liposomes comprise 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the cancer is selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, renal cancer, biliary cancer, gall bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, and hematologic malignancies (e.g., leukemia or lymphoma). In some embodiments, the cancer is selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposome composition comprising liposomes comprising a gamma polyglutamated antifolate and a targeting moiety having specific affinity for an epitope of an antigen on the surface of the cancer. In some embodiments, the liposome comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the liposome comprises a targeting moiety that specifically binds to one or more cell surface antigens (such as neoantigens) derived from or determined to be expressed on a tumor in a particular subject. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the liposome comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposome comprises a gamma-tetraglutamated antifolate. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In some embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the polyglutamated antifolate agent is an antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section. In some embodiments, the liposome comprises γ pantol containing γ -glutamyl in the form of L. In some embodiments, the liposome comprises γ pantol containing γ -glutamyl in D form. In some embodiments, the liposome comprises γ pantol containing at least one γ -glutamyl group in L form and at least one γ -glutamyl group in D form. In some embodiments, the cancer is selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, renal cancer, biliary cancer, gall bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, and hematologic malignancies (e.g., leukemia or lymphoma). In some embodiments, the cancer is selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma.
In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., TPLp- γ pantol). In some embodiments, the administered liposome composition comprises non-pegylated liposomes. In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposomes comprise a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, and other plasma cell dysplasias or dyscrasias, as well as leukemia and lymphoma and other B cell malignancies. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the liposome composition is administered to treat colorectal cancer. In some embodiments, the liposome composition is administered to treat ovarian cancer. In some embodiments, the liposome composition is administered to treat endometrial cancer. In some embodiments, the liposome composition is administered to treat pancreatic cancer. In some embodiments, the liposome composition is administered to treat liver cancer. In some embodiments, the liposome composition is administered to treat head and neck cancer. In some embodiments, the liposome composition is administered to treat osteosarcoma.
In a further embodiment, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having a cancer that expresses a folate receptor on the cell surface thereof an effective amount of a liposomal composition, wherein the liposomal composition comprises liposomes comprising (a) a gamma polyglutamated antifolate (γ pantol) and (b) a targeting moiety having specific binding affinity for a folate receptor. In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and/or folate receptor (FR-). In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha) and folate receptor beta (FR-beta). In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., TPLp- γ pantol). In some embodiments, the administered liposome composition comprises non-pegylated liposomes. In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposomes comprise a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dysplasias or dyscrasias. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the liposome composition is administered to treat colorectal cancer. In some embodiments, the liposome composition is administered to treat ovarian cancer. In some embodiments, the liposome composition is administered to treat endometrial cancer. In some embodiments, the liposome composition is administered to treat pancreatic cancer. In some embodiments, the liposome composition is administered to treat liver cancer. In some embodiments, the liposome composition is administered to treat head and neck cancer. In some embodiments, the liposome composition is administered to treat osteosarcoma.
In additional embodiments, the present disclosure provides a method for cancer maintenance therapy, the method comprising administering to a subject undergoing or having undergone cancer therapy an effective amount of a liposomal composition comprising liposomes containing a gamma polyglutamated antifolate (Lp-gamma pantol). In some embodiments, the liposome composition administered is PLp- γ pantol, NTLp- γ pantol, NTPLp- γ pantol, TLp- γ pantol or TPLp- γ pantol. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the administered liposome composition comprises a targeted liposome (e.g., TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL). In some embodiments, the administered liposome composition comprises a liposome (e.g., TPLp- γ pantol) that is pegylated and comprises a targeting moiety. In some embodiments, the liposomes of the administered liposome composition comprise a gamma polyglutamated antifolate containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposomal composition comprises gamma polyglutamate according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section.
In additional embodiments, the present disclosure provides a method for treating an immune system disorder, the method comprising administering to a subject having or at risk of having an immune system disorder an effective amount of a liposomal composition comprising liposomes containing a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol). In some embodiments, the liposome composition is administered to treat an autoimmune disease. In another embodiment, the liposome composition is administered to treat rheumatoid arthritis. In another embodiment, the liposome composition is administered to treat inflammation. In some embodiments, the immune system disorder is selected from: inflammation (e.g., acute and chronic inflammation), systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, systemic lupus erythematosus, takayasu's arteritis, and psoriasis. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the administered liposome composition comprises a targeting liposome (e.g., TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL) containing a targeting moiety with specific affinity for a surface antigen on a target cell (e.g., an immune cell) of interest. In other embodiments, the administered liposome composition comprises a liposome (e.g., TPLp- γ pantol) that is pegylated and comprises a targeting moiety. In some embodiments, the liposomes of the administered liposome composition comprise a gamma polyglutamated antifolate containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposomal composition comprises gamma polyglutamate according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section.
In additional embodiments, the present disclosure provides a method for treating an autoimmune disease, the method comprising administering to a subject having or at risk of having an autoimmune disease an effective amount of a liposomal composition comprising liposomes containing a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol). In some embodiments, the autoimmune disease is rheumatoid arthritis. In some embodiments, the autoimmune disease is a disease or disorder selected from: inflammatory Bowel Disease (IBD), crohn's disease, systemic lupus erythematosus, and psoriasis. In some embodiments, the autoimmune disease is a disease or disorder selected from: addison's disease, alopecia areata, ankylosing spondylitis, autoimmune hepatitis, autoimmune mumps, diabetes (type I), dystrophic bullous epidermolysis, epididymitis, glomerulonephritis, graves' disease, guillain-barre syndrome, hashimoto's disease, hemolytic anemia, systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, pemphigus vulgaris, psoriasis, rheumatic fever, rheumatoid arthritis, sarcoidosis, scleroderma, sjogren's syndrome, spondyloarthropathies, thyroiditis, vasculitis, vitiligo, myxoedema, pernicious anemia, and ulcerative colitis. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the administered liposome composition comprises a targeting liposome (e.g., TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL) containing a targeting moiety with specific affinity for a surface antigen on a target cell (e.g., an immune cell) of interest. In other embodiments, the administered liposome composition comprises a liposome (e.g., TPLp- γ pantol) that is pegylated and comprises a targeting moiety. In some embodiments, the liposomes of the administered liposome composition comprise a gamma polyglutamated antifolate containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposomes comprise a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section.
In additional embodiments, the present disclosure provides a method for treating an inflammatory disorder, the method comprising administering to a subject having or at risk of having an inflammatory disorder an effective amount of a liposomal composition comprising liposomes containing a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol, or TPLp-gamma pantol). In some embodiments, the inflammatory disorder is a disorder selected from: acute inflammation, chronic inflammation, systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, and systemic lupus erythematosus. In some embodiments, the inflammatory disorder is a disorder selected from: rheumatoid or other arthritic conditions (e.g., acute arthritis, acute gouty arthritis, bacterial arthritis, chronic inflammatory arthritis, degenerative arthritis (osteoarthritis), infectious arthritis, juvenile arthritis, fungal arthritis, neuropathic arthritis, polyarthritis, proliferative arthritis, psoriatic arthritis, venereal arthritis, viral arthritis), fibrositis, pelvic inflammatory disease, acne, psoriasis, actinomycosis, dysentery, biliary cirrhosis, lyme disease, heat eruption, stevens-johnson syndrome, mumps, pemphigus vulgaris, and blastomycosis. In some embodiments, the inflammatory disorder is inflammatory bowel disease. Inflammatory bowel disease is a chronic inflammatory disease of the gastrointestinal tract that includes, but is not limited to, crohn's disease, ulcerative colitis, and indeterminate colitis. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the administered liposome composition comprises a targeting liposome (e.g., TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL) containing a targeting moiety with specific affinity for a surface antigen on a target cell (e.g., an immune cell) of interest. In other embodiments, the administered liposome composition comprises a liposome (e.g., TPLp- γ pantol) that is pegylated and comprises a targeting moiety. In some embodiments, the liposomes of the administered liposome composition comprise a gamma pentaglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposomes comprise a polyglutamated antifolate agent according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section.
The present disclosure also provides a method of delivering a gamma polyglutamated antifolate to a site of inflammation in a subject, the method comprising: administering to a subject suffering from inflammation a composition comprising a gamma polyglutamated antifolate (L-gamma pantol) and a targeting moiety having specific binding affinity for an epitope located on a surface antigen on cells that are inflammatory or otherwise affect inflammation (e.g., via pro-inflammatory cytokine production). In some embodiments, the administered targeting moiety is associated with a delivery vehicle. In some embodiments, the delivery vehicle is an antibody or antigen-binding fragment of an antibody. In other embodiments, the delivery vehicle is a liposome. In other embodiments, the antibody, antigen-binding antibody fragment, or liposome is a pegylated liposome (e.g., TPLp- γ pantol). In some embodiments, the administered composition comprises a gamma polyglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered composition comprises a gamma-tetraglutamated antifolate. In some embodiments, the administered composition comprises a gamma-pentaglutaminated antifolate. In other embodiments, the administered composition comprises a gamma hexaglutaminated antifolate. In some embodiments, the γ pantol is a polyglutamated antifolate according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the delivery vehicle is a liposome according to any one of [12] to [67] of the summary of the invention section.
The present disclosure also provides a method of delivering a gamma polyglutamated antifolate to a tumor and/or cancer cell, the method comprising: administering to a subject having the tumor a composition comprising a gamma polyglutamated antifolate (L-gamma PANTIFOL) and a targeting moiety having specific binding affinity for an epitope on a surface antigen on a tumor cell or a cancer cell. In some embodiments, the administered targeting moiety is associated with a delivery vehicle. In some embodiments, the delivery vehicle is an antibody or antigen-binding fragment of an antibody. In other embodiments, the delivery vehicle is a liposome. In other embodiments, the antibody, antigen-binding antibody fragment, or liposome is a pegylated liposome (e.g., TPLp- γ pantol). In some embodiments, the administered composition comprises a gamma polyglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered composition comprises a gamma-tetraglutamated antifolate. In some embodiments, the administered composition comprises a gamma-pentaglutaminated antifolate. In other embodiments, the administered composition comprises a gamma hexaglutaminated antifolate. In some embodiments, the administered composition comprises a liposome according to any one of [1] to [11] of the summary of the invention section. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the administered composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section.
In additional embodiments, the present disclosure provides a method of making a liposome composition comprising a liposomal gamma polyglutamated antifolate (γ pantol) composition, the method comprising: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; and processing the mixture to form liposomes containing the polyglutamated antifolate agent. In some embodiments, the gamma polyglutamated antifolate agent contains 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the γ pantol comprises a γ tetraglutamated antifolate. In some embodiments, the γ pantol comprises a γ pentaglutaminated antifolate agent. In other embodiments, the γ pantol comprises a γ hexaglutaminated antifolate. In some embodiments, the γ pantol is a polyglutamated antifolate according to any one of [1] to [11] of the summary of the invention. In some embodiments, the γ pantol is a polyglutamated antifolate described in the summary section. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the summary of the invention section.
In one embodiment, the present disclosure provides a kit comprising an antifolate gamma polyglutamate composition and/or a gamma pantol delivery vehicle, such as liposomes containing gamma pantol and gamma pantol immunoconjugates (e.g., ADCs) as described herein.
Drawings
Fig. 1A-1N show the chemical formulas of antifolate pemetrexed (fig. 1A), exemplary gamma pemetrexed polyglutamate, gamma pemetrexed diglutamate (fig. 1B), gamma pemetrexed triglutamate (fig. 1C and 1D), gamma pemetrexed tetraglutamate (fig. 1E and 1F), gamma pemetrexed pentaglutamate (fig. 1G and 1H), gamma pemetrexed hexaglutamate (fig. 1I and 1J), gamma pemetrexed heptaglutamate (fig. 1K and 1L), and gamma pemetrexed octaglutamate (fig. 1M and 1N).
Figure 2 shows the relative potency of liposomal pemetrexed γ -L hexaglutamate (liposomal aG6) and its mirror liposomal γ -D hexaglutamate (liposomal aDG6) relative to pemetrexed within 48 hours after exposure to cancer cell lines SW620(CRC), HT-29 (colon cancer), H1806 (triple negative breast cancer), OAW28 (ovarian cancer), H292(NSCLC, adenocarcinoma subtype), and H2342(NSCLC, adenocarcinoma subtype).
Figure 3 shows exemplary dose response relationships of free pemetrexed L- γ -hexaglutamate (gG6), liposomal pemetrexed L- γ -hexaglutamate (liposomal gG6), pemetrexed, and folate receptor α -targeting antibody (FR1Ab) liposomal pemetrexed L- γ -hexaglutamate (liposomal gG6-FR1Ab) in HT-29 (colon cancer) at 48 hours.
FIG. 4 shows the effect of free pemetrexed L-gamma-hexaglutamate (hexa gG6) and liposomal pemetrexed L-gamma-hexaglutamate (liposomal hexa gG6) on the growth of colon cancer SW260 cells after exposure to 256nM of the corresponding dose for 48 hours. Non-targeted and targeted liposomal pemetrexed hexa gG6 were able to enter cells more efficiently than free pemetrexed hexa gG6, thereby inhibiting the growth of colon cancer SW260 cells.
Figure 5 shows the relative potency of liposomal pemetrexed L- γ hexaglutamate (liposomal gG6) and its mirror-image liposomal pemetrexed γ -D hexaglutamate (liposomal gDG6) relative to pemetrexed within 48 hours after exposure to cancer cell lines SW620(CRC), HT-29 (colon cancer), H1806 (triple negative breast cancer), OAW28 (ovarian cancer), H292(NSCLC, adenocarcinoma subtype), and H2342(NSCLC, adenocarcinoma subtype).
Figure 6 shows the therapeutic effect on HCC1806 triple negative breast cancer cells within 48 hours after exposure to liposomal pemetrexed gamma-L hexaglutamate (Lps Hexa gG6), liposomal pemetrexed gamma-D hexaglutamate (Lps Hexa gDG6), and pemetrexed.
Figure 7 shows the therapeutic effect on OAW28 ovarian cancer cells within 48 hours after exposure to liposomal pemetrexed gamma-L hexaglutamate (Lps Hexa gG6), liposomal pemetrexed gamma-D hexaglutamate (Lps Hexa gDG6) compared to pemetrexed.
Figure 8 shows the therapeutic effect on H292 non-small cell lung cancer cells within 48 hours after exposure to liposomal pemetrexed gamma-L hexaglutamate (Lps Hexa gG6), liposomal pemetrexed gamma-D hexaglutamate (Lps Hexa gDG6), and pemetrexed.
Figure 9 shows the therapeutic effect on H292 non-small cell lung cancer cells within 48 hours after exposure to various dose levels of liposomal pemetrexed gamma-L hexaglutamate (liposomal gG6), liposomal pemetrexed gamma-D hexaglutamate (liposomal gDG6), and pemetrexed in the range of 16 to 128 nM. The liposomal pemetrexed gG6 formulation outperformed pemetrexed in inhibiting H292 non-small cell lung cancer cells at each dose range tested.
Figure 10 shows the therapeutic effect on HCC1806 triple negative breast cancer cells within 48 hours after exposure to various dose levels of liposomal pemetrexed gamma-L hexaglutamate (liposomal gG6), liposomal pemetrexed gamma-D hexaglutamate (liposomal gDG6), and pemetrexed in the range of 16 to 128 nM. At each dose tested, the liposomal pemetrexed gG6 formulation outperformed pemetrexed in inhibiting HCC1806 triple negative breast cancer cells.
Figure 11 shows the therapeutic effect of liposomal pemetrexed gamma-L-hexametaphosphate (liposomal gG6), liposomal gamma-D hexametaphosphate (liposomal gDG6), and pemetrexed on OAW28 ovarian cancer cells within 48 hours post exposure after exposure to a range of concentrations. At 128nM, pemetrexed appeared to be more effective than the liposomal pemetrexed gG6 liposomal formulation, while the liposomal formulations at 32nM and 64nM had better therapeutic effects than pemetrexed; at 16nM, liposomal pemetrexed gG6 had a similar therapeutic effect as pemetrexed.
Figure 12 shows the toxicity of liposomal pemetrexed γ -L hexaglutamate (liposomal gG6), liposomal pemetrexed γ -D hexaglutamate (liposomal gDG6), and pemetrexed on differentiated human neutrophils at 64nM, 128nM, and 264 nM. The figures show that liposomal pemetrexed gG6 is significantly less toxic to differentiated human neutrophils compared to pemetrexed.
Figure 13 shows the effect of liposomal pemetrexed gamma-L-hexaglutamate (liposomal gG6), liposomal gamma-D-hexaglutamate (liposomal gDG6), and pemetrexed on neutrophils (differentiated from CD34+ cells) within 48 hours after exposure to various dose levels of the respective agents in the range of 16 to 128 nM.
Figure 14 shows the effect of liposomal pemetrexed γ -L hexaglutamate (liposomal gG6), liposomal pemetrexed γ -D hexaglutamate (liposomal gDG6), and pemetrexed on AML12 hepatocytes within 48 hours after exposure to 16nM, 32nM, and 64nM and 128nM of the respective agents. Strikingly, at the dose levels tested, there did not appear to be any toxicity to AML12 hepatocytes after treatment with liposomal pemetrexed gG6 at any liposomal dose. In contrast, pemetrexed treatment resulted in an approximately 40% reduction in AML12 hepatocyte counts at all doses studied.
Figure 15 shows the effect of liposomal pemetrexed γ -L hexaglutamate (liposomal gG6), liposomal pemetrexed γ -D hexaglutamate (liposomal gDG6), and pemetrexed on CCD841 colonic epithelial cells within 48 hours after exposure to 16nM, 32nM, and 64nM and 128nM of the respective agents. At all concentrations tested, pemetrexed resulted in a reduction in the number of CCD841 colonic epithelial cells of about > 50%, compared to about 20% or less after treatment with each liposome formulation tested.
FIG. 16 depicts the structures of polyglutamate antifolate, Cisplatin (CDDP) and two potential gG 6-cisplatin complexes. The pH-dependent formation of interchain and/or intrachain coordination between polyglutamated antifolate and the carboxyl groups of cisplatin may break down into individual molecules of gG6 and cisplatin when encountering the acidic pH of lysosomes (pH 3-5) and the presence of chloride ions inside the cell.
Figure 17 shows the hematological parameters for treatment of mice with 40mg/kg and 80mg/kg liposomal aG6 administered once per week for 4 weeks: effects of White Blood Cell (WBC) count, neutrophil count, and platelet count. No significant decrease in mean neutrophil, mean leukocyte or mean platelet counts was observed.
FIG. 18 shows the effect of treatment of mice with 40mg/kg and 80mg/kg liposomal aG6 administered once per week for 4 weeks on hemoglobin and reticulocyte indices. At higher dose levels, the mean hemoglobin concentration decreases minimally. In parallel, the mean reticulocyte proliferation index increased slightly.
Figure 19 shows the effect of treatment of mice with 40mg/kg and 80mg/kg liposomal aG6 administered once per week for 4 weeks on liver markers including serum aspartate Aminotransferase (AST) and serum alanine Aminotransferase (ALT) and serum albumin. There was no significant increase in mean AST or mean ALT levels of hepatic transaminase, and there was no observed change in mean albumin levels.
Figure 20 shows the relative tumor volumes of immunodeficient female Nu/J mice (6-8 weeks old) inoculated with NCI-H292 (non-small cell lung cancer) cells and intravenously administered 167mg/kg once every three weeks, control, pemetrexed and liposomal aG 6. As can be seen from these preliminary data, liposomal aG6 provides reduced tumor control compared to pemetrexed.
Figures 21A-21F show dose response relationships of liposomal pemetrexed α -L triglutamate (liposomal aG3), liposomal pemetrexed α -L pentaglutamate (liposomal aG5), liposomal pemetrexed α -L octaglutamate (liposomal aG7), and a combination of liposomal pemetrexed α -L hexaglutamate (aG6) and α -L dodecaglutamate (aG12) (liposomal aG6 and aG12) to H2342(NSCLC, adenocarcinoma subtype) (figure 21A), H292(NSCLC, adenocarcinoma subtype) (figure 21B), HT-29 (colon cancer) (figure 21C), HCC1806 (triple negative breast cancer) (figure 21D), MCF7(ER + breast cancer) (figure 21E), and OAW28 (ovarian cancer) (figure 21F) within 48 hours. Substantially as described in example 1, by
Figure BDA0002715408560000271
(CTG) luminescence cell viability assay to determine cell viability. As shown in all cell lines, the efficacy of each polyglutamated pemetrexed liposome composition greatly exceeded that of the liposome vehicle and empty liposome controls.
Detailed Description
The present disclosure generally relates to gamma polyglutamated antifolate compositions. The compositions provide progression over prior treatments for hyperproliferative diseases, such as cancer. Methods of making, delivering, and using the gamma polyglutamated antifolate compositions are also provided. The gamma-polyglutamated compositions have uses including, but not limited to, the treatment or prevention of hyperproliferative diseases (such as cancer), immune system disorders (including inflammatory and autoimmune diseases, such as rheumatoid arthritis), and infectious diseases (such as HIV, malaria, and schistosomiasis).
Definition of
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.
It should be understood that when the language "comprises" is used herein to describe embodiments, other similar embodiments described in terms of "comprising," "consisting of … …," and/or "consisting essentially of … …" are also provided. However, when used as a transitional phrase in a claim, each phrase should be interpreted separately and in the appropriate legal and factual context (e.g., in the claims, the transitional phrase "comprising" is more considered an open phrase than "consisting of … …" is more exclusive, and "consisting essentially of … …" takes an intermediate position).
As used herein, the singular forms "a", "an" and "the" include plural referents unless the content clearly dictates otherwise or is clear from the context.
The term "and/or" as used in phrases such as "a and/or B" herein is intended to include a and B; a or B; a (alone) and B (alone). Also, the term "and/or" as used in phrases such as "A, B and/or C" is intended to encompass each of the following embodiments: A. b and C; A. b or C; a or C; a or B; b or C; a and C; a and B; b and C; a (alone); b (alone); and C (alone).
The headings and sub-headings are used for convenience and/or form compliance only, do not limit the subject technology, and are not mentioned in connection with the explanation of the description of the subject technology. In various embodiments, features described under one heading or one subheading of the disclosure may be combined with features described under other headings or subheadings. Furthermore, not all features under a single title or a single subtitle may necessarily be used together in an embodiment.
Unless otherwise indicated, the terms "antifolate" and "anti enol" are used interchangeably to include the salt, acid and/or free base forms of the antifolate (e.g., disodium antifolate). Compositions comprising antifolate salts may also comprise any of a variety of cations, such as Na +, Mg2+, K +, NH4+, and/or Ca2 +. In certain embodiments, the salt is a pharmaceutically acceptable salt. Antifolates contain one L-gamma glutamyl group and are therefore considered monoglutamated for the purposes of this disclosure.
Although the compounds of the present invention may exist as mixtures of stereoisomers, it is preferred to resolve them into one optically active isomeric form. This requirement complicates the synthesis of the compounds and it is therefore preferred that they contain as few asymmetric carbon atoms as possible consistent with achieving the desired activity.
However, as previously mentioned, the cyclopent [ g ] quinazolines of the present invention contain at least three asymmetric carbon atoms. Among these, it is preferred to have a 6S orientation instead of a 6R orientation at the 6 positions of the ring system. Thus, preferred compounds (I) described above preferably have such a configuration at this asymmetric carbon atom, or less preferably are mixtures in which one or two of these asymmetric carbon atoms are not resolved.
The antifolate agent may be any known or future derived folate or antifolate agent that is polyglutamated. In some embodiments, the antifolate is selected from the group consisting of LV (etoposide), L-folinic acid (L-5-leucovorin); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and AG377, 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof.
In some embodiments, the antifolate is a member selected from the group consisting of: aminopterin, methotrexate, raltitrexed (also known as
Figure BDA0002715408560000281
ZD1694(RTX)), pralletrexed (also known as BGC 9331; ZD9331), pemetrexed (also known as ALIMTA, LY231514), Lometrexol (LMX) (5, 10-dideoxyntrahydrofolic acid), cyclopentyl [ g ] with dipeptide ligand]Quinazoline, CB3717, CB300945 (also known as BGC945) or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 (also known as BGC638) and BW1843U 89.
The terms "polyglutamated antifolate", "polyglutamated ANTIFOL", "ANTIFOL-PG", "PANTIFOL", and iterative forms thereof, are used interchangeably herein to refer to antifolate compositions comprising at least one glutamyl group in addition to a glutamyl group in the antifolate (i.e., ANTIFOL-PGn, where n.gtoreq.1). Glutamyl groups in antifolates are taken into account when referring herein to the number of glutamyl groups in gamma pantol (anti-PG). For example, an ANTIFOL-PG composition containing 5 glutamyl residues in addition to the glutamyl group of ANTIFOL is referred to herein as a hexaglutamylated antifolate or an antifolate hexaglutamate. The polyglutamate chain comprises an N-terminal glutamyl group and one or more C-terminal glutamyl groups. The N-terminal glutamyl group of the polyglutamate chain is not linked to another glutamyl group via its amine group, but is linked to one or more glutamyl groups via its carboxylic acid group. In some embodiments, the N-terminal glutamyl group of the polyglutamated antifolate is a glutamyl group of the antifolate. One or more C-terminal glutamyl groups of the polyglutamate chain are linked to another glutamyl group via its amine group, but not via its carboxylic acid group.
The terms "α glutamyl", "α glutamate" and "α linkage" and their iterative forms, as they relate to the linkage of a glutamyl group, refer to a glutamyl group containing an α carboxyl linkage. In some embodiments, none of the glutamyl groups of the polyglutamated antifolate provided contain an alpha linkage.
The terms "gamma glutamyl", "gamma glutamate" and "gamma linkage" as they relate to the linkage of a glutamyl group refer to a glutamyl group containing a gamma carboxyl linkage. In some embodiments, the gamma linkage is an amide linkage between the gamma carboxyl group of one glutamyl group and a second glutamyl group. The gamma linkage may be between the glutamyl group and a glutamyl group in the antifolate, or between the glutamyl group and a second glutamyl group not present in the antifolate (such as a glutamyl group in a polyglutamate chain linked to the antifolate). In some embodiments, the γ linkage refers to the amide bond of a glutamyl group of the antifolate. Unless specifically indicated or clearly understood from the context that is not intended, reference to a gamma linkage includes the gamma linkage of a glutamyl group of an antifolate. In some embodiments, the gamma glutamyl is in the L form. In some embodiments, gamma glutamyl is in the D form. As discussed herein, in antifolate therapy, antifolates enter cells and are polyglutamated by the enzyme folate poly-gamma-glutamate synthetase (FPGS), which continuously adds L-glutamyl groups to the gamma carboxyl groups of glutamate within L-glutamyl groups in the antifolate. Thus, during antifolate treatment, no D-gamma polyglutamated antifolate composition is formed intracellularly.
The terms "gamma polyglutamated antifolate", "gamma-polyglutamated antifolate", "gamma PANTIFOL", "gamma polyglutamated antifolate", "polyglutamated ANTIFOL", "gamma ANTIFOL-PG", and iterative forms thereof, are used interchangeably herein to refer to antifolate compositions comprising at least one gamma glutamyl group having a gamma carboxyl linkage in addition to the gamma glutamyl group of the antifolate (e.g., ANTIFOL-PGn, wherein n ≧ 1 gamma glutamyl group). The number of glutamyl groups in gamma pantol (gamma ANTIFOL-PG) is referred to herein taking into account the gamma-glutamyl groups in antifolates. For example, a γ anti pol-PG composition containing 5 γ -glutamyl groups in addition to a glutamyl group in an antifolate may be referred to herein as a γ hexaglutaminated antifolate or a γ antifolate hexaglutamate.
The terms "α glutamyl", "α -glutamyl" and "α linkage" as they relate to the linkage of a glutamyl group refer to a glutamyl group containing an α carboxyl linkage.
As used herein, the term "isolated" refers to a composition in a form not found in nature. Isolated gamma polyglutamic acid compositions include those that have been purified to the extent that they are no longer in the form in which they are found in nature. In some embodiments, the isolated gamma polyglutamated antifolate agent is substantially pure. Isolated compositions will be free or substantially free of materials with which they are naturally associated, such as other cellular components, such as proteins and nucleic acids, that may be present with them in nature or their environment of preparation (e.g., cell culture). The gamma polyglutamated composition may be formulated with diluents or adjuvants and still be isolated for practical purposes-for example, when used in diagnosis or therapy, the gamma polyglutamated composition will typically be mixed with a pharmaceutically acceptable carrier or diluent. In some embodiments, an isolated gamma polyglutamate composition (e.g., gamma polyglutamate and a delivery vehicle, such as a liposome containing gamma polyglutamate, contains less than 1% or less than 0.1% of undesirable DNA or protein content.
The term "targeting moiety" is used herein to refer to a molecule that provides enhanced affinity for a selected target (e.g., a cell, cell type, tissue, organ, region or compartment of the body, such as a cell, tissue or organ compartment). The targeting moiety may comprise a wide variety of entities. The targeting moiety may comprise a naturally occurring molecule, or a recombinant or synthetic molecule. In some embodiments, the targeting moiety is an antibody, antigen-binding antibody fragment, bispecific antibody, or other antibody-based molecule or compound. In some embodiments, the targeting moiety is an aptamer, an avimer, a receptor binding ligand, a nucleic acid, a biotin-avidin binding pair, a peptide, a protein, a carbohydrate, a lipid, a vitamin, a toxin, a component of a microorganism, a hormone, a receptor ligand, or any derivative thereof. Other targeting moieties are known in the art and are encompassed by the present disclosure.
The term "specific affinity" or "specifically binds" refers to some combination of more frequent, faster, longer lasting, greater affinity, or the like, reaction or association of a targeting moiety (e.g., an antibody or antigen-binding antibody fragment) with an epitope, protein, or target molecule as compared to a surrogate (including a protein not associated with the epitope of interest). Due to sequence identity between homologous proteins in different species, in some embodiments, specific affinity may include a binding agent that recognizes a protein or target in more than one species. Likewise, due to homology within certain regions of the polypeptide sequences of different proteins, the terms "specific affinity" or "specific binding" may include binders that recognize more than one protein or target. It will be appreciated that in certain embodiments, a targeting moiety that specifically binds a first target may or may not specifically bind a second target. Thus, "specific affinity" does not necessarily require (although may include) unique binding, i.e., binding to a single target. Thus, in certain embodiments, a targeting moiety may specifically bind more than one target. In certain embodiments, multiple targets may be bound by the same targeting moiety.
The term "epitope" refers to that portion of an antigen that is capable of being recognized and specifically bound by a targeting moiety (i.e., a binding moiety), such as an antibody. When the antigen is a polypeptide, the epitope may be formed by contiguous amino acids and non-contiguous amino acids juxtaposed by tertiary folding of the protein. Epitopes formed by contiguous amino acids are typically retained when the protein is denatured, while epitopes formed by tertiary folding are typically lost when the protein is denatured. Epitopes typically comprise at least 3, and more typically at least 5 or 8-10 amino acids in a unique spatial conformation.
Expressions such as "binding affinity to a target", "binding to a target" and similar expressions known in the art refer to the properties of a targeting moiety that can be directly measured by determining the affinity constant, e.g., the amount of the targeting moiety that associates and dissociates at a given antigen concentration. Different methods can be used to characterize molecular interactions, such as, but not limited to, competition assays, equilibrium assays, and microcalorimetric assays, as well as real-time interaction assays based on surface plasmon resonance interactions (e.g., using
Figure BDA0002715408560000301
Instrument). These methods are well known to those skilled in the art and are described, for example, in Neri et al, Tibtech 14: 465-.
The term "delivery vehicle" generally refers to any composition used to assist, facilitate or assist the entry of gamma-polyglutamated antifolate into a cell. Such delivery vehicles are known in the art and include, but are not limited to, liposomes, lipid spheres, polymers (e.g., polymer-conjugates), peptides, proteins such as antibodies (e.g., immunoconjugates, such as Antibody Drug Conjugates (ADCs) and antigen-binding antibody fragments and their derivatives), cellular components, cyclic oligosaccharides (e.g., cyclodextrins), micelles, microparticles (e.g., microspheres), nanoparticles (e.g., lipid nanoparticles, biodegradable nanoparticles, and core-shell nanoparticles), hydrogels, lipoprotein particles, viral sequences, viral materials, or lipid or liposome formulations, and combinations thereof. The delivery vehicle may be directly or indirectly linked to the targeting moiety. In some examples, the targeting moiety is selected from a macromolecule, a protein, a peptide, a monoclonal antibody, or a fatty acid lipid.
By "subject" is meant a human or vertebrate mammal, including but not limited to dogs, cats, horses, goats, and primates, such as monkeys. Thus, the invention may also be used to treat a disease or disorder in a non-human subject. For example, cancer is one of the leading causes of death in companion animals (e.g., cats and dogs). In some embodiments of the invention, the subject is a human. In the present disclosure, the terms "subject" and "patient" are used interchangeably and have the same meaning. It is generally preferred to use the maximum dose, i.e., the highest safe dose according to sound medical judgment.
As used herein, "effective amount" refers to a dose of an agent sufficient to provide a medically desirable result. The effective amount will vary with the desired result, the particular condition being treated or prevented, the age and physical condition of the subject being treated, the severity of the condition, the duration of the treatment, the nature of concurrent or combined therapy (if any), the particular route of administration, and like factors within the knowledge and expertise of a health practitioner. For the purposes set forth, an effective amount can be determined empirically and in a conventional manner. In the case of cancer, an effective amount of the agent can reduce the number of cancer cells; reducing the size of the tumor; inhibit (i.e., slow to some extent and preferably prevent) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably prevent) tumor metastasis; inhibit tumor growth to some extent; and/or alleviate one or more symptoms associated with the condition to some extent. To the extent the drug can prevent growth and/or kill existing cancer cells, it can be cytostatic and/or cytotoxic. For cancer therapy, in vivo efficacy can be measured, for example, by assessing duration of survival, duration of progression-free survival (PFS), response rate (e.g., RR), response duration, and/or quality of life.
The terms "hyperproliferative disorder", "proliferative disease" and "proliferative disorder" are used interchangeably herein and relate to unwanted or uncontrolled cellular proliferation of excessive or abnormal cells, which is not desired, such as tumors or proliferative growth (whether in vitro or in vivo). In some embodiments, the proliferative disease is a cancer or a tumor disease (including benign or cancerous) and/or any metastasis, regardless of where the cancer, tumor and/or metastasis is located. In some embodiments, the proliferative disease is a benign or malignant tumor. In some embodiments, the proliferative disease is a non-cancerous disease. In some embodiments, the proliferative disease is a hyperproliferative disorder, such as hyperplasia, fibrosis (especially pulmonary fibrosis, but also including other types of fibrosis, such as renal fibrosis), angiogenesis, psoriasis, atherosclerosis, and smooth muscle proliferation in blood vessels, such as stenosis or restenosis following angioplasty.
"cancer," "tumor," or "malignant disease" are used synonymously and refer to any of a variety of diseases characterized by uncontrolled, abnormal proliferation of cells, the ability of affected cells to spread locally or through the bloodstream and lymphatic system to other parts of the body (metastasis), and any of a number of characteristic structural and/or molecular features. As used herein, "tumor" refers to all neoplastic cell growth and proliferation (whether malignant or benign) as well as all precancerous and cancerous cells and tissues. By "cancerous tumor" or "malignant cell" is understood a cell that has specific structural properties, lacks differentiation and is capable of invading and metastasizing. Cancers that can be treated using the γ pantol compositions provided herein include, but are not limited to, non-hematologic malignancies, including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, biliary cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dysplasias or dyscrasias. In some embodiments, the cancer is selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. Other types of cancers and tumors that can be treated using the gamma pantol compositions are described herein or are otherwise known in the art. The term "metastasis" refers to the spread or dissemination of a tumor, cancer or neoplasia to other sites, locations, regions or organs or tissue systems within the body of a subject, wherein the sites, locations, regions or organs or tissue systems are distinct from the primary tumor, cancer or neoplasia. The terms "cancer," "cancerous," "cell proliferative disorder," "proliferative disorder," and "tumor" are not mutually exclusive when referred to herein.
Terms such as "treating" or "treatment" or "treating" refer to (a) therapeutic measures to cure, slow down, alleviate symptoms of, and/or halt the progression of the diagnosed pathological condition or disorder and (b) prophylactic or preventative measures to prevent and/or slow the development of the targeted disease or condition. Thus, a subject in need of treatment includes a subject already suffering from a cancer, disorder or disease; those subjects at risk for cancer or a condition; and those subjects in whom the infection or condition is to be prevented. Using well known medical and diagnostic techniques, a subject is identified as "having" or at risk of having a cancer, an infectious disease, an immune system disorder, a hyperproliferative disease, or another disease or disorder mentioned herein. In certain embodiments, a subject is successfully "treated" according to the methods provided herein if the subject exhibits a full, partial, or transient improvement or elimination of, for example, symptoms associated with a disease or disorder (e.g., cancer, inflammation, and rheumatoid arthritis). In particular embodiments, the terms "treating" or "treatment" refer to ameliorating at least one measurable physical parameter of a proliferative disorder, such as tumor growth, which is not necessarily discernible by the patient. In other embodiments, the terms "treating" or "treatment" refer to inhibiting the progression of a proliferative disorder in the body, for example, by stabilizing a discernible symptom or physiologically, for example, by stabilizing a physical parameter, or both. In other embodiments, the term "treating" or "treatment" refers to a reduction or stabilization of tumor size, tumor cell proliferation or survival, or cancer cell count. The gamma-pantol composition may be used alone or in combination with additional therapeutic agents for treatment.
"subject" and "patient" and "animal" are used interchangeably and refer to mammals such as human patients and non-human primates as well as laboratory animals such as rabbits, rats, and mice, among others. Animals include all vertebrates, e.g., mammals and non-mammals, such as chickens, amphibians, and reptiles. As used herein, "mammal" refers to any member of the class mammalia, including, but not limited to, humans and non-human primates, such as chimpanzees, and other apes and monkey species; livestock such as cattle, sheep, pigs, goats, and horses; domestic mammals, such as dogs and cats; laboratory animals, including rodents, such as mice, rats and guinea pigs, and other members of the class mammalia known in the art. In a particular embodiment, the patient is a human.
"treatment of a proliferative disorder" is used herein to include maintaining or reducing tumor size, inducing tumor regression (partial or complete), inhibiting tumor growth, and/or increasing the lifespan of a subject having a proliferative disorder. In one embodiment, the proliferative disorder is a solid tumor. Such tumors include, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, renal cancer, cholangiocarcinoma, gallbladder cancer, bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma. In one embodiment, the proliferative disorder is a hematologic malignancy. Such hematological malignancies include, for example, leukemias, lymphomas and other B cell malignancies, myelomas and other plasma cell dysplasias or dyscrasias.
The term "autoimmune disease" as used herein is defined as a condition caused by an autoimmune response. Autoimmune diseases are the result of inappropriate and excessive responses to self-antigens. Examples of autoimmune diseases include, but are not limited to, Addison's disease, alopecia areata, ankylosing spondylitis, autoimmune hepatitis, autoimmune mumps, Crohn's disease, diabetes (type I), dystrophic bullous epidermolysis, epididymitis, glomerulonephritis, Graves ' disease, Guillain-Barre syndrome, Hashimoto's disease, hemolytic anemia, systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, pemphigus vulgaris, psoriasis, rheumatic fever, rheumatoid arthritis, sarcoidosis, scleroderma, Sjogren's syndrome, spondyloarthropathies, thyroiditis, vasculitis, vitiligo, myxoedema, pernicious anemia, ulcerative colitis, and the like.
The terms "inflammation" and "inflammatory disease" are used interchangeably and refer to a disease or condition characterized by or caused by inflammation. "inflammation" refers to the local response to cellular injury that serves as a mechanism to begin the elimination of harmful agents and damaged tissue, and is characterized by telangiectasia, leukocyte infiltration, redness, warmth, and pain. Sites of inflammation include the lungs, pleura, tendons, lymph nodes or glands, uvula, vagina, brain, spinal cord, nasal and pharyngeal mucosa, muscles, skin, bone or bony tissue, joints, bladder, retina, cervix of uterus, canthus, intestines, vertebrae, rectum, anus, bursa, hair follicles, and the like. Such inflammatory diseases include, but are not limited to, inflammatory bowel disease, rheumatoid diseases (e.g., rheumatoid arthritis), other arthritic diseases (e.g., acute arthritis, acute gouty arthritis, bacterial arthritis, chronic inflammatory arthritis, degenerative arthritis (osteoarthritis), infectious arthritis, juvenile arthritis, fungal arthritis, neuropathic arthritis, multiple arthritis, proliferative arthritis, psoriatic arthritis, venereal arthritis, viral arthritis), fibrositis, pelvic inflammatory disease, acne, psoriasis, actinomycosis, dysentery, biliary cirrhosis, lyme disease, heat rash, stevens-johnson syndrome, mumps, pemphigus vulgaris, and blastomycosis. Inflammatory bowel disease is a chronic inflammatory disease of the gastrointestinal tract that includes, but is not limited to, crohn's disease, ulcerative colitis, and indeterminate colitis. Rheumatoid arthritis is a chronic inflammatory disease of primarily joints, usually multiple joints, characterized by inflammatory changes in the synovium and joint structure as well as muscle atrophy and osteoporosis.
The term "therapeutic agent" is used herein to refer to an agent or derivative or prodrug thereof that interacts with a hyperproliferative cell, such as a cancer cell or an immune cell, thereby reducing the proliferative state of the cell and/or killing the cell. Examples of therapeutic agents include, but are not limited to, chemotherapeutic agents, cytotoxic agents, platinum-based agents (e.g., cisplatin, carboplatin, oxaliplatin), taxanes (e.g.,
Figure BDA0002715408560000321
) Etoposide, alkylating agents (e.g., cyclophosphamide, ifosfamide), metabolic antagonists (e.g., Antifolates (ANTIFOL), 5-fluorouracil gemcitabine or derivatives thereof), antitumor antibiotics (e.g., mitomycin, doxorubicin), plant-derived antitumor agents (e.g., vincristine, vindesine, Taxol). Such agents may also include, but are not limited to, the anticancer agents trimetrexate, temozolomide, raltitrexed, S- (4-nitrobenzyl) -6-thioinosine (NBMPR), 6-benzylguanidine (6-BG), bis-chloronitrosourea (BCNU), and camptothecitm, or a therapeutic derivative of any of them. Can be used suitablyAdditional examples of therapeutic agents for use in combination with the disclosed methods include, but are not limited to, anti-restenotic agents, pro-proliferative or anti-proliferative agents, anti-inflammatory agents, anti-neoplastic agents, anti-mitotic agents, anti-platelet agents, anti-coagulant agents, anti-fibrin agents, anti-thrombin agents, cytostatic agents, antibiotics and other anti-infective agents, anti-thrombin agents, anti-metabolite agents, angiogenic agents, cytoprotective agents, Angiotensin Converting Enzyme (ACE) inhibitors, angiotensin II receptor antagonists, and/or cardioprotective agents. "therapeutic agent" also refers to salts, acids, and free base forms of the above agents.
As used herein, the term "chemotherapeutic agent," when used in relation to cancer therapy, refers to any agent that causes cancer cells to die or inhibits the growth or spread of cancer cells. Examples of such chemotherapeutic agents include alkylating agents, antibiotics, antimetabolites, plant derived agents, and hormones. In some embodiments, the chemotherapeutic agent is cisplatin. In some embodiments, the chemotherapeutic agent is carboplatin. In some embodiments, the chemotherapeutic agent is oxaliplatin. In other embodiments, the chemotherapeutic agent is gemcitabine. In other embodiments, the chemotherapeutic agent is doxorubicin.
The term "antimetabolite" is used herein to refer to an antineoplastic agent or prodrug thereof that inhibits the utilization of a metabolite. Examples of antimetabolites include antifolates, pemetrexed, 5-fluorouracil prodrugs such as capecitabine, 5-fluorodeoxyuridine monophosphate, cytarabine prodrugs such as nelarabine, 5-azacytidine, gemcitabine, mercaptopurine, thioguanine, azathioprine, adenosine, pentostatin, erythrohydroxynonyladenine (erythrohydroxynonyladenosine), and cladribine. Antimetabolites useful for practicing the methods of the present disclosure include nucleoside analogs, including purine or pyrimidine analogs. In some embodiments, the gamma polyglutamated antifolate composition is used in combination with an antimetabolite selected from the group consisting of: fluoropyrimidine 5-fluorouracil, 5-fluoro-2' -deoxycytidine, cytarabine, gemcitabine, troxacitabine, decitabine, azacytidine, pseudoisocytidine, zebularine, ancitabine, fazarabine, 6-azacytidine, capecitabine, N4-octadecyl-cytarabine, cytarabine elaeate, fludarabine, cladribine, clofarabine, nelarabine, forodesine (forodesine), and pentostatin, or derivatives thereof. In one example, a nucleoside analog is a substrate for a nucleoside deaminase, which is either adenosine deaminase or cytidine deaminase. In some examples, the nucleoside analog is selected from fludarabine, cytarabine, gemcitabine, decitabine, and azacytidine, or derivatives thereof. In certain embodiments, the antimetabolite is 5-fluorouracil.
As used herein, a "taxane" is an anti-cancer agent that interferes with or disrupts microtubule stability, formation and/or function. Taxane agents include paclitaxel and docetaxel and their derivatives, which act on microtubules in the same way as the taxane from which they are derived. In certain embodiments, the taxane is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel. In certain embodiments, the taxane is paclitaxel
Figure BDA0002715408560000331
Docetaxel
Figure BDA0002715408560000332
Albumin-bound paclitaxel (nano albumin-bound paclitaxel;
Figure BDA0002715408560000333
) DHA-paclitaxel or PG-paclitaxel.
The term "pharmaceutically acceptable carrier" refers to an ingredient of a pharmaceutical formulation other than an active ingredient that is not toxic to a subject. Pharmaceutically acceptable carriers include, but are not limited to, buffers, excipients, stabilizers, or preservatives. A pharmaceutically acceptable carrier may include, for example, one or more compatible solid or liquid fillers, diluents, or encapsulating substances suitable for administration to a human or other subject.
The present disclosure relates generally to gamma polyglutamated antifolate (gamma anti fol) compositions and methods of making and using the compositions to treat diseases, including hyperproliferative diseases (such as cancer), immune system disorders (such as rheumatoid arthritis), and infectious diseases (such as HIV, malaria, and schistosomiasis).
In some embodiments, the present disclosure provides:
[1] a composition comprising a gamma polyglutamated antifolate agent;
[2] the composition of [1], wherein the antifolate is selected from the group consisting of: pirtrexin, pralatrexate, AG2034, GW1843, and LY309887, and or stereoisomers thereof;
[3] the composition of [1], wherein the antifolate is selected from the group consisting of: PMX, MTX, RTX and LTX, or stereoisomers thereof;
[4] the composition according to any one of [1] to [3], wherein the antifolate is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof;
[5] The composition of [1], wherein the antifolate is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89;
[6] the composition of any one of [1] to [5], wherein the gamma polyglutamated antifolate agent contains 4, 5, 2-10, 4-6, or more than 5 glutamyl groups;
[7] the composition of any one of [1] - [6], wherein the gamma polyglutamated antifolate agent:
(a) is a gamma-tetraglutaminic antifolate;
(b) is a gamma-pentaglutaminated antifolate; or
(c) Is a gamma-hexaglutaminated antifolate;
[8] the composition of any one of [1] to [7], wherein the gamma polyglutamated antifolate agent comprises 1-10 glutamyl groups having gamma carboxyl linkages;
[9] the composition according to any one of [1] to [8], wherein:
(a) at least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form,
(b) each glutamyl group of the gamma polyglutamated antifolate is in the L form,
(c) At least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form,
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate, or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form;
[10] the composition of any one of [1] to [9], wherein the polyglutamate is linear;
[11] the composition of any one of [1] to [9], wherein the polyglutamate is branched;
[12] a liposomal composition comprising a gamma polyglutamated antifolate (Lp-gamma pantol) according to any one of [1] to [11 ];
[13] the Lp- γ PANTIFOL composition of [12], wherein the polyglutamated antifolate agent is selected from the group consisting of:
(a) AG2034, pirtroxine, pralatrexate, GW1843, antifolate and LY 309887; or
(b) PMX, MTX, RTX and LTX, or stereoisomers thereof;
[14] the Lp- γ PANTIFOL composition of [12] or [13], wherein the polyglutamated antifolate agent is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and AG377, 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof;
[15] The Lp- γ PANTIFOL composition of any one of [12] to [14], wherein the antifolate is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89;
[16] the Lp- γ PANTIFOL composition of any one of [12] to [15], wherein the liposome comprises a gamma polyglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 gamma glutamyl groups;
[17] the Lp- γ PANTIFOL composition of any one of [12] to [16], wherein the liposome comprises a γ tetraglutamated antifolate;
[18] the Lp- γ PANTIFOL composition of any one of [12] to [16], wherein the liposome comprises a γ -pentaglutamated antifolate;
[19] the Lp- γ PANTIFOL composition of any one of [12] to [16], wherein the liposome comprises a γ hexaglutaminated antifolate;
[20] the Lp- γ PANTIFOL composition of any one of [12] - [19], wherein the gamma polyglutamated antifolate agent comprises 1-10 glutamyl groups having a gamma carboxyl linkage;
[21] The Lp- γ PANTIFOL composition according to any one of [12] to [20], wherein:
(a) at least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form;
(b) each glutamyl group of the gamma polyglutamated antifolate agent is in the L form;
(c) at least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form;
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate; or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form;
[22] the Lp-gamma PANTIFOL composition as claimed in any one of [12] to [21], wherein
(a) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form;
(b) each glutamyl group of the gamma polyglutamated antifolate agent is in the L form;
(c) at least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form;
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate; or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form;
[23] The Lp- γ PANTIFLOL composition of any one of [12] - [22], wherein the liposome is pegylated (PLp- γ PANTIFLOL);
[24] the Lp- γ PANTIFOL composition of any one of [12] to [22], wherein the liposome is non-pegylated;
[25] the Lp- γ PANTIFOL composition of any one of [12] to [24], wherein the liposome has a diameter in the range of 20nm to 200 nm;
[26] the Lp- γ PANTIFOL composition of any one of [12] to [25], wherein the liposome has a diameter in the range of 80nm to 120 nm;
[27] the Lp- γ PANTIFOL composition of any one of [12] to [26], wherein the liposome is formed from liposome components;
[28] the Lp- γ PANTIFOL composition of [27], wherein the liposomal composition comprises at least one of anionic lipids and neutral lipids;
[29] the Lp- γ PANTIFOL composition of [27] or [28], wherein the liposome component comprises at least one selected from the group consisting of: DSPE; DSPE-PEG; DSPE-PEG-maleimide; HSPC; HSPC-PEG; cholesterol; cholesterol-PEG; and cholesterol-maleimide.
[30] The Lp- γ PANTIFOL composition of any one of [27] - [29], wherein the liposome component comprises at least one selected from the group consisting of: DSPE; DSPE-PEG; DSPE-PEG-FITC; DSPE-PEG-maleimide; cholesterol; and HSPC;
[31] The Lp- γ PANTIFOL composition of any one of [27] to [30], wherein the one or more liposomal components further comprises a steric stabilizer;
[32] the Lp- γ PANTIFLOL composition of [31], wherein the steric stabilizer is at least one selected from the group consisting of: polyethylene glycol (PEG); poly-L-lysine (PLL); monosialoganglioside (GM 1); poly (vinyl pyrrolidone) (PVP); poly (acrylamide) (PAA); poly (2-methyl-2-oxazoline); poly (2-ethyl-2-oxazoline); a phosphatidylpolyglycerol; poly [ N- (2-hydroxypropyl) methacrylamide ]; amphiphilic poly-N-vinylpyrrolidone; an L-amino acid-based polymer; oligomerization of glycerol; copolymers comprising polyethylene glycol and polypropylene oxide; poloxamer 188; and polyvinyl alcohol;
[33] the Lp- γ PANTIFLOL composition of [32], wherein the steric stabilizer is PEG, and the PEG has a number average molecular weight (Mn) of 200 to 5000 daltons;
[34] the Lp- γ PANTIFOL composition of any one of [12] to [33], wherein the liposome is anionic or neutral;
[35] the Lp- γ PANTIFOL composition of any one of [12] to [33], wherein the liposome has a zeta potential less than or equal to zero;
[36] The Lp- γ PANTIFOL composition of any one of [12] - [33], wherein the liposome has a zeta potential between 0 to-150 mV;
[37] the Lp- γ PANTIFOL composition of any one of [12] - [33], wherein the liposome has a zeta potential between-30 to-50 mV;
[38] the Lp- γ PANTIFOL composition of any one of [12] to [33], wherein the liposome is cationic;
[39] the Lp- γ PANTIFOL composition of any one of [12] - [38], wherein the liposome has an interior space comprising the γ polyglutamated antifolate agent and a pharmaceutically acceptable aqueous carrier;
[40] the Lp- γ PANTIFOL composition of [39], wherein the pharmaceutically acceptable carrier comprises a tonicity agent such as dextrose, mannitol, glycerol, potassium chloride, sodium chloride at a concentration of greater than 1%;
[41] the Lp- γ PANTIFOL composition of [39], wherein the pharmaceutically acceptable aqueous carrier is trehalose;
[42] the Lp- γ PANTIFOL composition of [41], wherein the pharmaceutically acceptable carrier comprises 1% to 50% trehalose;
[43] the Lp- γ PANTIFOL composition of any one of [39] - [42], wherein the pharmaceutically acceptable carrier comprises a dextrose solution of 1% to 50%;
[44] The Lp- γ PANTIFOL composition of any one of [39] - [43], wherein the interior space of the liposome comprises 5% dextrose suspended in HEPES buffer solution;
[45] the Lp- γ PANTIFOL composition of any one of [39] - [44], wherein the pharmaceutically acceptable carrier comprises a buffer, such as HEPES Buffered Saline (HBS) or the like, at a concentration of between 1 and 200mM and a pH of between 2 and 8;
[46] the Lp- γ PANTIFOL composition of any one of [39] - [45], wherein the pharmaceutically acceptable carrier comprises sodium acetate and calcium acetate in a total concentration between 50mM to 500 mM;
[47] the Lp- γ PANTIFOL composition of any one of [12] to [46], wherein the interior space of the liposome has a pH of 5-8 or a pH of 6-7, or any range therebetween;
[48] the Lp- γ PANTIFOL composition of any one of [12] - [47], wherein the liposome comprises less than 500,000 or less than 200,000 of the γ polyglutamated antifolate molecules;
[49] the Lp- γ PANTIFOL composition of any one of [12] - [48], wherein the liposomes comprise between 10 to 100,000 of the γ polyglutamated antifolate molecules, or any range therebetween;
[50] The Lp- γ PANTIFLOL composition of any one of [12] to [49], further comprising a targeting moiety, and wherein the targeting moiety has specific affinity for a surface antigen on a target cell of interest;
[51] the Lp- γ PANTIFLOL composition of [50], wherein the targeting moiety is attached to one or both of PEG and the exterior of the liposome, optionally wherein targeting moiety is attached to one or both of the PEG and the exterior of the liposome by a covalent bond;
[52] the Lp- γ PANTIFLOL composition of [50] or [51], wherein the targeting moiety is a polypeptide;
[53] the Lp- γ PANTIFLOL composition of any one of [50] - [52], wherein the targeting moiety is an antibody or an antigen-binding fragment of an antibody;
[54]according to [50]-[53]The Lp-gamma PANTIFOL composition of any one of the above, wherein the use is as follows
Figure BDA0002715408560000371
The targeting moiety is measured by analysis to be at 0.5x 10-10To 10x 10-6An equilibrium dissociation constant (Kd) in a range binds to the surface antigen;
[55] the Lp- γ PANTIFOL composition of any one of [50] - [54], wherein the targeting moiety specifically binds to one or more folate receptors selected from the group consisting of: folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and folate receptor (FR-);
[56] The Lp- γ PANTIFLOL composition of any one of [50] - [55], wherein the targeting moiety comprises one or more selected from the group consisting of: antibodies, humanized antibodies, antigen-binding fragments of antibodies, single chain antibodies, single domain antibodies, bispecific antibodies, synthetic antibodies, pegylated antibodies, and multimeric antibodies;
[57] the Lp- γ PANTIFOL composition of any one of [50] - [56], wherein each pegylated liposome comprises 1 to 1000 or 30 to 200 targeting moieties;
[58] the Lp- γ PANTIFLOL composition of any one of [39] - [57], further comprising one or more of an immunostimulatory agent, a detectable label, and a maleimide, wherein the immunostimulatory agent, the detectable label, or the maleimide is attached to the PEG or the exterior of the liposome;
[59] the Lp- γ PANTIFLOL composition of [58], wherein the immunostimulatory agent is at least one selected from the group consisting of: a protein immunostimulant; a nucleic acid immunostimulant; a chemical immunostimulant; a hapten; and an adjuvant;
[60] the Lp- γ PANTIFLOL composition of [58] or [59], wherein the immunostimulant is at least one selected from the group consisting of: fluorescein; fluorescein Isothiocyanate (FITC); DNP; beta glucan; beta-1, 3-glucan; beta-1, 6-glucan; resolvin (resolvin) (e.g., resolvin D such as Dn-6DPA or Dn-3DPA, resolvin E, or T series resolvin); and Toll-like receptor (TLR) modulators, such as oxidized low density lipoproteins (e.g., OXPAC, PGPC) and eritoran lipids (e.g., E5564);
[61] The Lp- γ PANTIFOL composition of any one of [58] to [60], wherein the immunostimulatory agent and the detectable label are the same;
[62] the Lp- γ PANTIFLOL composition of any one of [58] to [61], further comprising a hapten;
[63] the Lp- γ PANTIFLOL composition of [62], wherein the hapten comprises one or more of fluorescein or β 1, 6-glucan;
[64] the Lp- γ PANTIFLOL composition of any one of [12] - [63], further comprising at least one cryoprotectant selected from the group consisting of: mannitol; trehalose; sorbitol; and sucrose;
[65] a targeting composition comprising the composition according to any one of [1] to [64 ];
[66] a non-targeted composition comprising the composition according to any one of [1] to [49 ];
[67] the Lp- γ PANTIFOL composition of any one of [12] - [66], further comprising carboplatin and/or pembrolizumab (pembrolizumab);
[68] a pharmaceutical composition comprising a liposomal gamma polyglutamated antifolate composition according to any one of [12] to [67 ];
[69] A pharmaceutical composition comprising a gamma polyglutamated antifolate composition according to any one of [1] to [7 ];
[70] a composition as described in any one of [1] to [69], for use in the treatment of a disease;
[71] use of a composition as defined in any one of [1] to [70] in the manufacture of a medicament for the treatment of a disease;
[72] a method for treating or preventing a disease in a subject in need of such treatment or prevention, the method comprising administering to the subject a composition as described in any one of [1] to [70 ];
[73] a method for treating or preventing a disease in a subject in need of such treatment or prevention, the method comprising administering to the subject a liposomal gamma polyglutamated antifolate composition of any one of [12] to [69 ];
[74] a method of killing a hyperproliferative cell, said method comprising contacting a hyperproliferative cell with a composition as described in any of [1] to [69 ];
[75] a method of killing hyperproliferative cells, said method comprising contacting hyperproliferative cells with a liposomal gamma polyglutamated antifolate composition of any one of [12] to [69 ];
[76] The method of [74] or [75], wherein the hyperproliferative cell is a cancer cell, a mammalian cell, and/or a human cell;
[77] a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of the composition of any one of [1] to [69 ];
[78] a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [12] to [68 ];
[79] the method of [77] or [78], wherein the cancer is selected from: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dyscrasias;
[80] the method of [77] or [78], wherein the cancer is selected from: lung, breast, colon, pancreatic, gastric, bladder, head and neck, ovarian and cervical cancer;
[81] The method of [77] or [78], wherein the cancer is selected from: colorectal, lung, breast, head and neck, and pancreatic cancers;
[82] the method of [77] or [78], wherein the cancer is selected from: colorectal cancer, breast cancer, ovarian cancer, lung cancer, head and neck cancer, pancreatic cancer, gastric cancer, and mesothelioma;
[83] a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer cells expressing on their surface folate receptors bound by a targeting moiety an effective amount of an Lp- γ pantol composition according to any one of [50] - [66 ];
[84] a maintenance therapy for a subject undergoing or having undergone cancer therapy, the maintenance therapy comprising administering to the subject undergoing or having undergone cancer therapy an effective amount of the composition of any one of [1] - [69 ];
[85] a maintenance therapy for a subject undergoing or having undergone cancer therapy, said maintenance therapy comprising administering to a subject undergoing or having undergone cancer therapy an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [12] - [69 ];
[86] A method for treating an immune system disorder, the method comprising administering to a subject having or at risk of having an immune system disorder an effective amount of the composition of any one of [1] - [69], optionally wherein the immune system disorder is selected from: inflammation (e.g., acute and chronic inflammation), systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, systemic lupus erythematosus and takayasu's arteritis, and psoriasis;
[87] a method for treating an immune system disorder, the method comprising administering to a subject having or at risk of having an immune system disorder an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [8] - [69], optionally wherein the immune system disorder is selected from: inflammation (e.g., acute and chronic inflammation), systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, systemic lupus erythematosus and takayasu's arteritis, and psoriasis;
[88] a method for treating the following diseases:
(a) an infectious disease, the method comprising administering to a subject having or at risk of having an infectious disease an effective amount of the composition according to any one of [1] to [69 ];
(b) An infectious disease, a cardiovascular disease, a metabolic disease, or another disease, the method comprising administering to a subject having or at risk of having an infectious disease, a cardiovascular disease, or another disease an effective amount of a composition according to any one of [1] - [69], wherein the disease is a member selected from: atherosclerosis, cardiovascular disease (CVD), coronary artery disease, myocardial infarction, stroke, metabolic syndrome, gestational trophoblastic disease, and ectopic pregnancy;
(c) an autoimmune disease, the method comprising administering to a subject having or at risk of having an autoimmune disease an effective amount of a composition according to any one of [1] to [69 ];
(d) rheumatoid arthritis, the method comprising administering to a subject having or at risk of having rheumatoid arthritis an effective amount of the composition according to any one of [1] to [69 ];
(e) an inflammatory disorder, the method comprising administering to a subject having or at risk of having inflammation an effective amount of a composition according to any one of [1] - [69], optionally wherein the inflammation is acute, chronic and/or systemic inflammation; or
(f) A skin condition, the method comprising administering to a subject having or at risk of having a skin condition an effective amount of the composition according to any one of [1] - [69], optionally wherein the skin condition is psoriasis;
[89] a method for treating an infectious disease, the method comprising administering to a subject having or at risk of having an infectious disease an effective amount of a liposomal gamma polyglutamated antifolate composition of any one of [12] to [69 ];
[90] a method of delivering a gamma polyglutamated antifolate to a tumor expressing a folate receptor on the surface, the method comprising: administering to a subject having the tumor an amount of an Lp- γ PANTIFOL composition of any one of [1] - [69] to deliver a therapeutically effective dose of a γ polyglutamated antifolate to the tumor;
[91] a method of making a gamma polyglutamated antifolate composition comprising a liposomal gamma polyglutamated antifolate composition according to any one of [12] to [69], the method comprising: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; and processing the mixture to form liposomes containing a gamma polyglutamated antifolate agent;
[92] A method of making a gamma polyglutamated antifolate composition comprising a liposomal gamma polyglutamated antifolate composition according to any one of [12] to [69], the method comprising: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; and processing the mixture to form liposomes containing a gamma polyglutamated antifolate agent,
[93] the method of [92], wherein processing the mixture comprises homogenizing the mixture to form liposomes in the solution,
[94] a method of making the composition of any one of [50] to [69], the method comprising the steps of: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; processing the mixture to form liposomes encapsulating and/or encapsulating the gamma polyglutamated antifolate agent; and providing a targeting moiety on the surface of the liposome, the targeting moiety having specific affinity for at least one of folate receptor alpha (FR-a), folate receptor beta (FR- β), and folate receptor (FR-);
[95] A method of making the composition of any one of [50] to [69], the method comprising the steps of: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; processing the mixture to form liposomes encapsulating and/or encapsulating the gamma polyglutamated antifolate agent; and providing a targeting moiety on the surface of the liposome, the targeting moiety having specific affinity for at least one of folate receptor alpha (FR-a), folate receptor beta (FR- β), and folate receptor (FR-);
[96] the method of [95], wherein the processing step comprises homogenizing the mixture to form liposomes in the solution;
[97] the method of [92], wherein the processing step comprises one or more of: film hydration, extrusion, online mixing, an ethanol injection technology, a freeze thawing technology, reversed phase evaporation, dynamic high-pressure micro-jet, micro-jet mixing, a multiple emulsion method, a freeze drying multiple emulsion method, 3D printing, a membrane contactor method and stirring; and/or
[98] The method of any of [95] to [97], wherein the processing step comprises one or more steps of altering the size of the liposomes by one or more of extrusion, high pressure microfluidization, and/or sonication steps; and/or
[99] The method of any one of [91] to [98], wherein at least 1% of starting material of a gamma polyglutamated antifolate is encapsulated or embedded in the liposomes.
I. Gamma polyglutamated antifolate agent (gamma PANTIFOL)
The present disclosure generally relates to gamma polyglutamated antifolate (gamma pantol) compositions. The gamma pantol composition comprises at least one glutamyl group having a gamma linkage. These compositions are structurally distinct from the L- γ polyglutamated form of antifolates (L γ 1 pantol) produced by folyl polyglutamate synthase (FPGS) in cells during antifolate treatment.
In some embodiments, the gamma pantol composition contains 2-20, 2-15, 2-10, 2-5 or more than 5 glutamyl groups (including glutamyl groups of antifolates). In some embodiments, each glutamyl group in the gamma pantol has a gamma linkage in addition to the glutamyl group of the antifolate. In some embodiments, 2 or more glutamyl groups in the γ pantol have a γ linkage. In some embodiments, each glutamyl group in the γ pantol is in the L form. In some embodiments, each glutamyl group in the γ pantol is in the D form in addition to a glutamyl group in the antifolate. In some embodiments, the γ pantol comprises two or more glutamyl groups in L form and one or more glutamyl groups in D form.
In some embodiments, the antifolate is selected from the group consisting of: PMX, MTX, RTX and LTX, or stereoisomers thereof.
In some embodiments, the antifolate is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and AG377, 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof.
In some embodiments, the antifolate is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89.
In some embodiments, the antifolate is a 6-substituted pyrrolo [2,3-d ] pyrimidine benzoyl antifolate. In some embodiments, the antifolate is a 6-substituted pyrrolo [2,3-d ] pyrimidine benzoyl antifolate having a carbon bridge length of 1 to 6 carbons (e.g., a compound having the structure of formula (I), wherein n1 ═ 1-6). In some embodiments, the antifolate is a 6-substituted thieno [2,3-d ] pyrimidine benzoyl antifolate having a bridge with a bridge length of 2-8 carbons (e.g., a compound having the structure of formula (II), wherein n2 ═ 7-13). In some embodiments, the antifolate is a 6-substituted pyrrolo [2,3-d ] pyrimidine antifolate having a thienyl group replacing a benzoyl moiety (e.g., a compound having the structure of formula (III)) with a bridge length of 2-8 carbons, wherein n1 ═ 1-6. In some embodiments, the antifolate has a structure according to any one of formulas (I) - (III), wherein x ═ 4, 5, 2-10, 4-6, or greater than 5.
Figure BDA0002715408560000411
In some embodiments, the antifolate is selected from the group consisting of: methotrexate derivatives bearing an indoline ring and a modified ornithine, methotrexate derivatives bearing an indoline ring and a modified glutamic acid, methotrexate derivatives bearing an alkyl-substituted phenyl ring C, methotrexate derivatives bearing a benzoxazine moiety, methotrexate derivatives bearing a benzothiazine moiety, 10-deazaaminopterin analogs, 5-deazaaminopterin methotrexate analogs, 5, 10-deazaaminopterin methotrexate analogs, methotrexate derivatives bearing an indoline moiety, lipophilic amide methotrexate derivatives, methotrexate analogs containing L-threo- (2S,4S) -4-fluoro-glutamic acid, methotrexate analogs containing DL-3, 3-difluoroglutamic acid, methotrexate tetrahydroquinazoline analogs, N- (ac-aminoacyl) methotrexate derivatives, biotin methotrexate derivatives, D-glutamic methotrexate analogs, D-erythro, threo-4-fluoroglutamic methotrexate analogs, beta, gamma-methanomethotrexate analogs, 10-deazaaminopterin (10-EDAM) analogs, gamma-tetrazole methotrexate analogs, N- (L-alpha-aminoacyl) methotrexate derivatives, meta-isomers of aminopterin, ortho-isomers of aminopterin, hydroxymethyl methotrexate, gamma-fluoro methotrexate, polyglutamyl methotrexate derivatives, gem-diphosphonic acid methotrexate analogs (see, e.g., WO1988/06158, the contents of which are incorporated herein by reference in their entirety), alpha-substituted methotrexate analogs, Gamma-substituted methotrexate analogs, 5-methyl-5-deaminomethotrexate analogs (see, e.g., U.S. Pat. No. 4,725,687, the contents of which are incorporated herein by reference in their entirety), N-acyl-Nalpha- (4-amino-4-deoxypteroyl) -L-ornithine derivatives, 8-deazamethotrexate analogs, acivicin methotrexate analogs, polymeric cisplatin methotrexate derivatives, methotrexate-gamma-dimyristoyl phosphatidylethanolamine, methotrexate polyglutamate analogs, poly-gamma-glutamyl methotrexate derivatives, deoxyuridylate methotrexate derivatives, iodoacetyl lysine methotrexate analogs, 2, omega-diaminoalkanoic acid-containing methotrexate analogs-polyglutamate methotrexate derivatives, 5-methyl-5-deaza analogs, quinazoline methotrexate analogs, pyrazine methotrexate analogs, cysteine or homocysteine methotrexate analogs (see, e.g., U.S. Pat. No. 4,490,529 and EPA 0142220, the contents of each of which are incorporated herein by reference in their entirety), gamma-tert-butyl methotrexate ester, fluorinated methotrexate analogs, folic acid methotrexate analogs, phosphonoglutamic acid analogs, poly (L-lysine) methotrexate conjugates, dilysine or trilysine methotrexate derivatives, 7-hydroxy methotrexate, poly-gamma-glutamyl methotrexate analogs, 3 ', 5' -dichloro methotrexate, diazoketone or chloromethyl ketone methotrexate analogs, 10-propargyl aminopterin, alkyl methotrexate homologs, lectin derivatives of methotrexate, Polyglutamate methotrexate derivatives, halogenated methotrexate derivatives, 8-alkyl-7, 8-dihydro analogs, 7-methyl methotrexate derivatives, dichloro methotrexate, lipophilic methotrexate derivatives, 3 ', 5' -dichloro methotrexate, deazaaminopterin analogs and MX 068; or a stereoisomer thereof.
In some embodiments, the antifolate agent has formula (IV):
Figure BDA0002715408560000421
wherein X is CH2、C2H4Or O (CH)2)3O; r1 ═ Me or Et; r2 ═ H, Cl, F, OH, or R2 ═ R3; and R3 ═ H, Cl, F, OH, Me, or Br.
In some embodiments, the antifolate agent has formula (IV), wherein X ═ CH 2; r1 ═ Me or Et; r2 ═ H, Cl, F, OH, or R2 ═ R3; and R3 ═ H, Cl, F, OH, Me, or Br. In some embodiments, X ═ CH2(ii) a R1 ═ Me; r2 ═ H, Cl, F, OH; and R3 ═ H, Cl, Me, or Br. In some embodiments, X ═ O (CH)2)3O; r1 ═ Me; and R2 ═ R3 ═ H.
In some embodiments, the antifolate agent has formula (V):
Figure BDA0002715408560000422
wherein X is C2H4、C4H8、C6H12、O(CH2)2O or O (CH)2)3O; r1 ═ H or Cl, or R2 ═ R3; and R3 ═ H or Cl.
In some embodiments, the antifolate agent has formula (V), wherein X ═ C2H4(ii) a And R1 ═ R2 ═ H or CL. In some embodiments, X ═ C2H4(ii) a R1 ═ Cl; and R2 ═ H. In some embodiments, X ═ C4H8(ii) a And R1 ═ R2 ═ H. In some embodiments, X=C6H12(ii) a And R1 ═ R2 ═ H.
In some embodiments, the antifolate agent has formula (VI):
Figure BDA0002715408560000431
wherein X is CH2Or C2H4(ii) a Y ═ 2, 5-thiophene; and R is CH 2F. Cn, Et, Me or CH2OH。
In some embodiments, the antifolate agent has formula (VI), wherein X ═ CH2(ii) a Y ═ 2, 5-thiophene; and R ═ H2F. Cn, Et or CH2And (5) OH. In some embodiments, X ═ C2H4(ii) a Y ═ 2, 5-thiophene; and R ═ Me.
In some embodiments, the antifolate agent has formula (VII):
Figure BDA0002715408560000432
wherein X is N or CH; y is NH2;CH3Or H; and R is CH3CHO or H.
In some embodiments, the antifolate is of formula (VII), wherein (a) X ═ N; y is NH2(ii) a And R ═ H; (b) x is N; y is NH2(ii) a And R is CH3;(c)X=N,Y=NH2(ii) a And R ═ CHO; (d) x is CH and Y is NH2R ═ H; (e) x ═ CH, Y ═ H, R ═ H; or (f) X ═ CH, Y ═ CH3And R ═ H.
In some embodiments, the antifolate agent has formula (VIII):
Figure BDA0002715408560000433
wherein A ═ NH, NCH3Or CH2
In some embodiments, the antifolate agent has formula (IX):
Figure BDA0002715408560000434
wherein, (a) X ═ OH; r is H; and Y ═ GIu, (b) X ═ OCH3(ii) a R is H; and Y ═ GIu, (c) X ═ OH, R ═ H; and Y ═ valine; (d) x ═ OH; r is H; and Y ═ suberate; or (e) X ═ OH; r is CH3(ii) a And Y ═ GIu.
In a further embodiment, the antifolate is a cyclopent [ g ] quinazoline derivative. In some embodiments, the cyclopenta [ g ] quinazoline derivative is N- {4- [ N- (2-methyl-4-oxo-3, 4,7, 8-tetrahydro-6H-cyclopenta [ g ] quinazolin-6-yl) -N- (prop-2-ynyl) amino ] benzoyl } -L- γ -glutamyl } -D-glutamic acid; or N- {4- [ N- (2-hydroxymethyl-4-oxo-3, 4,7, 8-tetrahydro-6H-cyclopenta [ g ] -quinazolin-6-yl) -N- (prop-2-ynyl) amino ] benzoyl } -L-y-glutamyl } -D-glutamic acid; or a pharmaceutically acceptable salt or ester thereof.
In some embodiments, the antifolate agent has formula (X):
Figure BDA0002715408560000441
wherein R1 is H, amino, C1-4 alkyl, C1-4 alkoxy, C1-4 hydroxyalkyl, or C1-4 fluoroalkyl;
r2 is hydrogen, C1-4 alkyl, C3-4 alkenyl, C3-4 alkynyl, C2-4 hydroxyalkyl, C2-4 haloalkyl or C1-4 cyanoalkyl;
ar is phenylene, thiophenediyl, thiazolediyl, pyridinediyl or pyrimidinediyl, which may optionally bear one or two substituents selected from the group consisting of halo, hydroxy, amino, nitro, cyano, trifluoromethyl, C1-4 alkyl and C1-4 alkoxy; and is
R3 is a group of one of the following formulae: -NHCH (CO2H) -A1-Y1-NH-A3-Y3 or R3 is L-or D-glutamyl linked to an alpha or gamma carboxyl group.
In some embodiments, the antifolate agent has the formula (X), wherein R1 is C1-4 alkyl or C1-4 hydroxyalkyl (e.g., methyl or hydroxymethyl); r2 is (a) methyl, ethyl, propyl, prop-2-enyl, prop-2-ynyl, 2-hydroxy-ethyl, 2-fluoroethyl, 2-bromoethyl or 2-cyanoethyl, (b) methyl, or (c) prop-2-ynyl; and Ar is 1, 4-phenylene or 1, 4-phenylene having one or two substituents selected from chloro and fluoro (e.g., a 2-fluoro substituent such as 2-fluoro-1, 4-phenylene or 2, 6-difluoro-1, 4-phenylene), thiophene-2, 5-diyl, thiazole-2, 5-diyl, or pyridine-2, 5-diyl.
In some embodiments, the antifolate agent is of formula (X), wherein R1 is methyl or hydroxymethyl; r2 is methyl or prop-2-ynyl; and Ar is 1, 4-phenylene or 1, 4-phenylene having a 2-fluoro substituent (such as in 2, 6-difluoro-1, 4-phenylene or especially 2-fluoro-1, 4-phenylene), or is pyridine 2, 5-diyl. In some embodiments, Ar is 1, 4-phenylene or 2-fluoro-1, 4-phenylene.
In other embodiments, the gamma polyglutamated antifolate agent is disclosed in WO 2009/115776; WO 2003/020300; WO 2003/020706, WO 2003/020748; gibbs et al, Cancer Research 65(15):11721-11728 (2005); and cyclopenta [ g ] quinazolines disclosed in Bavetsias et al, Tetrahedron 63(7):1537-1543(2007), the contents of each of which are incorporated herein by reference in their entirety.
In some embodiments, the gamma polyglutamated antifolate agent is diglutamated. That is, the gamma polyglutamated antifolate contains 1 gamma-glutamyl group (gamma ANTIFOL-PG1) in addition to the glutamyl group in the antifolate, and the additional glutamyl group is linked to the glutamyl group in the antifolate by a gamma bond. In some embodiments, each glutamyl group of the gamma-diglutamated antifolate agent is in the L form. In other embodiments, the gamma-diglutamated antifolate agent comprises a glutamyl group in the D form.
In some embodiments, the gamma polyglutamated antifolate agent is triglutaminated. That is, the gamma polyglutamated antifolate contains 2 gamma-glutamyl groups (gamma ANTIFOL-PG2) in addition to glutamyl groups in the antifolate. In some embodiments, each of the 2 additional glutamyl groups has a gamma linkage. In other embodiments, one of the 2 glutamyl groups has a gamma linkage and the other glutamyl group has a gamma linkage. In some embodiments, each glutamyl group of the gamma triglutamated antifolate agent is in the L form. In other embodiments, the gamma tri-glutamated antifolate comprises a glutamyl group in the D form. In other embodiments, each glutamyl group of the gamma-triglutamated antifolate is in the D form in addition to the gamma-glutamyl group in the antifolate. In further embodiments, the gamma-triglutamated antifolate agent comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is tetraglutamated, and thus contains 3 gamma-glutamyl groups (gamma ANTIFOL-PG3) in addition to the glutamyl groups of the antifolate agent. In some embodiments, the gamma tetraglutamylated antifolate comprises two or more gamma-glutamyl groups in the L form. In other embodiments, each γ -glutamyl group of the γ tetraglutamylated antifolate is in the L form. In other embodiments, the gamma tetraglutamylated antifolate comprises a gamma-glutamyl group in the D form. In some embodiments, the gamma tetraglutamylated antifolate comprises 2 gamma-glutamyl groups in the D form. In other embodiments, each glutamyl group of the gamma tetraglutamylated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the tetraglutamated antifolate comprises a γ -glutamyl group in the D form and two or more γ -glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is pentaglutamated (gamma ANTIFOL-PG4) and contains a chain of 4 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the gamma pentaglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma pentaglutaminated antifolate is in the L form. In other embodiments, the gamma pentaglutaminated antifolate comprises a glutamyl group in the D form. In some embodiments, the gamma tetraglutamylated antifolate comprises 2 or 3 gamma-glutamyl groups in D form. In other embodiments, each γ -glutamyl group of the gamma pentaglutaminated antifolate is in the D form in addition to a glutamyl group in the antifolate. In further embodiments, the pentaglutamated antifolate comprises a γ -glutamyl group in the D form and two or more γ -glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is hexaglutamated (gamma ANTIFOL-PG5) and contains a chain of 5 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the gamma hexaglutamylated antifolate comprises two or more gamma-glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma hexaglutaminated antifolate is in the L form. In other embodiments, the gamma hexaglutaminated antifolate comprises a gamma-glutamyl group in the D form. In some embodiments, the gamma tetraglutamated antifolate comprises 2, 3, 4 or 5 gamma-glutamyl groups in D form. In other embodiments, each glutamyl group of the gamma hexaglutaminated antifolate is in the D form in addition to a glutamyl group in the antifolate. In further embodiments, the hexaglutamated antifolate comprises a γ -glutamyl group in the D form and two or more γ -glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is heptaglutamated (gamma anti ol-PG6), and thus contains a chain of 6 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the gamma heptaglutamated antifolate comprises two or more gamma-glutamyl groups in the L form. In other embodiments, each γ -glutamyl group of the γ heptaglutamylated antifolate is in the L form. In other embodiments, the gamma heptaglutamated antifolate comprises a gamma-glutamyl group in D form. In some embodiments, the gamma tetraglutamated antifolate comprises 2, 3, 4, 5 or 6 gamma-glutamyl groups in D form. In other embodiments, each γ -glutamyl group of the γ heptaglutamylated antifolate is in the D form in addition to a glutamyl group in the antifolate. In further embodiments, the heptaglutamated antifolate comprises a γ -glutamyl group in the D form and two or more γ -glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate is octaglutamated (gamma anti col-PG 7), and thus contains a chain of 7 gamma-glutamyl groups linked to glutamyl groups in the antifolate. In some embodiments, the gamma octaglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma octaglutamated antifolate is in the L form. In other embodiments, the gamma octaglutamated antifolate comprises a glutamyl group in D form. In some embodiments, the gamma octaglutamated antifolate agent comprises 2, 3, 4, 5, 6 or 7 gamma-glutamyl groups in D form. In other embodiments, each glutamyl group of the gamma octaglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the octaglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is nonaglutamated (gamma ANTIFOL-PG8) and contains a chain of 8 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the gamma nonaglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma nonaglutamated antifolate is in the L form. In other embodiments, the gamma nonaglutamated antifolate comprises a glutamyl group in the D form. In other embodiments, each glutamyl group of the gamma nonaglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the nonaglutamated antifolate comprises a γ -glutamyl group in the D form and two or more γ -glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate is decaglutamated (gamma anti col-PG 9) (i.e., a chain containing 9 gamma-glutamyl groups linked to glutamyl groups in the antifolate). In some embodiments, the gamma decaglutamated antifolate agent comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma decaglutamated antifolate is in the L form. In other embodiments, the gamma decaglutamated antifolate comprises a glutamyl group in the D form. In other embodiments, each glutamyl group of the gamma decaglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the decaglutamated antifolate agent comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate is undeglutamated (gamma ANTIFOL-PG10) and contains a chain of 10 gamma-glutamyl groups linked to glutamyl groups in the antifolate. In some embodiments, the gamma undecaplutamated antifolate agent comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma undeglutamated antifolate is in the L form. In other embodiments, the gamma undecaplutamated antifolate agent comprises a D glutamyl group. In other embodiments, each glutamyl group of the gamma undeglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the undecaplutamated antifolate agent comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate is dodecaglutamated (gamma ANTIFOL-PG11) and contains a chain of 11 gamma-glutamyl groups linked to glutamyl groups in the antifolate. In some embodiments, the gamma dodecaglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma dodecaglutamated antifolate is in the L form. In other embodiments, the gamma dodecaglutamated antifolate comprises a glutamyl group in the D form. In other embodiments, each glutamyl group of the gamma dodecaglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the dodecaglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is tridecyl glutamated (gamma ANTIFOL-PG12) and contains a chain of 12 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the γ tridecyl glutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the γ tridecyl glutamated antifolate is in the L form. In other embodiments, the gamma tridecyl glutamated antifolate comprises a glutamyl group in D form. In other embodiments, each glutamyl group of the γ tridecyl glutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the tridecyl-glutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate is tetradecaglutated (gamma ANTIFOL-PG13) and contains a chain of 13 gamma-glutamyl groups linked to glutamyl groups in the antifolate. In some embodiments, the gamma tetradecylglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma tetradecylglutamated antifolate is in the L form. In other embodiments, the gamma tetradecylglutamated antifolate comprises a glutamyl group in D form. In other embodiments, each glutamyl group of the gamma tetradecylglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the tetradecylglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is pentadeclutamated (gamma ANTIFOL-PG14) and contains a chain of 14 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the γ pentadecyl glutamated antifolate comprises two or more glutamyl groups in L form. In other embodiments, each glutamyl group of the γ pentadecyl glutamated antifolate is in the L form. In other embodiments, the γ pentadecyl glutamated antifolate comprises a glutamyl group in D form. In other embodiments, each glutamyl group of the γ pentadecglutamated antifolate is in the D form in addition to a glutamyl group in the antifolate. In further embodiments, the pentadecyl glutamated antifolate comprises a glutamyl group in D form and two or more glutamyl groups in L form.
In some embodiments, the gamma polyglutamated antifolate is hexaglutamated (gamma ANTIFOL-PG15) and contains a chain of 15 gamma-glutamyl groups linked to glutamyl groups in the antifolate. In some embodiments, the gamma hexadecagoylated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma hexadecagoylated antifolate is in the L form. In other embodiments, the gamma hexadecagoylated antifolate comprises a glutamyl group in the D form. In other embodiments, each glutamyl group of the gamma hexadecagoylated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the hexadecagoylated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate is heptadecaglutamated (gamma anti pol-PG 16) and contains a chain of 16 gamma-glutamyl groups linked to glutamyl groups in the antifolate. In some embodiments, the gamma heptadecaglutamic acid resistant agent comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma heptadecaglutamic antifolate is in the L form. In other embodiments, the gamma heptadecaglutamic acid antifolate comprises a D glutamyl group. In other embodiments, each glutamyl group of the gamma heptadecaglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the heptadecaglutamated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate is octadecylglutamated (gamma ANTIFOL-PG17) and contains a chain of 17 gamma-glutamyl groups linked to glutamyl groups in the antifolate. In some embodiments, the gamma octadecylglutamated antifolate comprises two or more glutamyl groups in L form. In other embodiments, each glutamyl group of the γ octadecylglutamated antifolate is in the L form. In other embodiments, the gamma-octadecylglutamated antifolate comprises D-glutamyl. In other embodiments, each glutamyl group of the γ octadecylglutamated antifolate is in the D form in addition to a glutamyl group in the antifolate. In further embodiments, the octadecaglutamated antifolate agent comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is nonadecanoylated (gamma ANTIFOL-PG18) and contains a chain of 18 gamma glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the gamma nineteen glutamated antifolate agent comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the γ nineteen glutamated antifolate is in the L form. In other embodiments, the gamma nineteen glutamated antifolate comprises D glutamyl. In other embodiments, each glutamyl group of the gamma nonadecanoylated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the nineteen glutamated antifolate agent comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is icosanylated (gamma ANTIFOL-PG19) and contains a chain of 19 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the gamma eicosylglutamated antifolate comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma eicosylglutamated antifolate is in the L form. In other embodiments, the gamma eicosylglutamated antifolate comprises a D glutamyl group. In other embodiments, each glutamyl group of the gamma eicosylglutamated antifolate is in the D form in addition to the glutamyl group in the antifolate. In further embodiments, the icosyl-glutamated antifolate agent comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate agent is heneicosanmate (gamma anti col-PG 20) and contains a chain of 20 gamma-glutamyl groups linked to glutamyl groups in the antifolate agent. In some embodiments, the gamma heneicosylated antifolate agent comprises two or more glutamyl groups in the L form. In other embodiments, each glutamyl group of the gamma heneicosylated antifolate is in the L form. In other embodiments, the gamma heneicosylated antifolate comprises a D-glutamyl group. In other embodiments, each glutamyl group of the gamma heneicosylated antifolate is in the D form in addition to a glutamyl group in the antifolate. In further embodiments, the heneicosylated antifolate comprises a glutamyl group in the D form and two or more glutamyl groups in the L form.
In some embodiments, the gamma polyglutamated antifolate contains a chain of 4-7 glutamyl groups linked to the antifolate (i.e., gamma anti l-PGn, where n ═ 4-7), and each of the 4-7 linked glutamyl groups has a gamma linkage. In some embodiments, each of the 4-7 linked glutamyl groups is in the L form. In other embodiments, each of the 4-7 linked glutamyl groups is in the D form. In other embodiments, the 4-7 linked glutamyl groups are in the L form and the D form.
In some embodiments, the gamma polyglutamated antifolate (gamma pantol) contains a total of 1-15, 1-10, 2-15, 2-10, 3-15, 3-10, 3-6, 3-5, 4-10, 4-7, or 4-6 glutamyl groups, including glutamyl groups of an antifolate, or any range therebetween. In some embodiments, each glutamyl group in the γ pantol has a γ linkage in addition to a glutamyl group in the antifolate. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 glutamyl groups in the gamma pantol have a gamma linkage. In some embodiments, the γ pantol comprises γ glutamyl in L form and D form. In some embodiments, each glutamyl group in the polyglutamate structure of the polyglutamated antifolate is in the L form. In some embodiments, each glutamyl group in the γ pantol is in the D form in addition to a glutamyl group in the antifolate. In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 glutamyl groups in the gamma pantol are in the L form. In another embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 glutamyl groups in the gamma pantol are in the form of D.
In some embodiments, the gamma polyglutamated antifolate (gamma pantol) contains a total of 2-20, 2-15, 2-10, 2-5 glutamyl groups, including glutamyl groups of antifolate, or any range therebetween. In some embodiments, each glutamyl group in the γ pantol is in the L form. In some embodiments, each glutamyl group in the γ pantol is in the D form in addition to the glutamyl group of the antifolate. In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 glutamyl groups in the γ pantol are in the L form. In another embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19 glutamyl groups in the γ pantol are in the form of D.
In some embodiments, the gamma polyglutamated antifolate contains a total of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 glutamyl groups in addition to glutamyl groups of the antifolate).
In some embodiments, a total of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 glutamyl groups in the gamma polyglutamated antifolate agent are in the L form, the D form, or in the L form and the D form. In some embodiments, each glutamyl group of the gamma polyglutamated antifolate is in the L form. In other embodiments, each glutamyl group of the gamma polyglutamated antifolate is in the D form in addition to a glutamyl group in the antifolate. In an alternative embodiment, at least two glutamyl groups in the gamma polyglutamated antifolate agent are in the L form and at least one glutamyl group in the gamma polyglutamated antifolate agent is in the D form. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16 glutamyl groups in the gamma polyglutamated antifolate agent are in the L form. In other embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 glutamyl groups in the gamma polyglutamated antifolate agent are in the D form.
In further embodiments, the gamma polyglutamated antifolate agent contains 20-100, 20-75, 20-50, 20-40, 20-30, 20-25, or more than 100 gamma glutamyl groups, or any range therebetween. In some embodiments, each glutamyl group of the gamma polyglutamated antifolate is in the L form. In other embodiments, each glutamyl group of the gamma polyglutamated antifolate is in the D form in addition to a glutamyl group in the antifolate. In an alternative embodiment, at least two glutamyl groups in the gamma polyglutamated antifolate agent are in the L form and at least one glutamyl group in the gamma polyglutamated antifolate agent is in the D form.
In further embodiments, provided compositions comprise a gamma polyglutamated antifolate comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 1-10, or 1-20 glutamyl groups having a gamma linkage. In some embodiments, the gamma polyglutamated antifolate agent contains 1, 2, 3, 4, 5, 6, 7, 8, 9, 1-10, or 1-20 glutamyl groups in the L form. In some embodiments, the gamma polyglutamated antifolate agent contains 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 1-10, or 1-20 glutamyl groups in D form. In some embodiments, the gamma polyglutamated antifolate agent contains 1, 2, 3, 4, 5, 6, 7, 8, 9, 1-10, or 1-20 glutamyl groups in L form and 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 1-10 or 1-20 glutamyl groups in D form.
In some embodiments, the gamma polyglutamated antifolate compositions provided herein are capable of adding one or more additional glutamyl groups, i.e., the compositions are capable of acting as substrates for FPGS (folate polyglutamate synthetase). Reagents and assays and reagents for determining the ability of a gamma polyglutamated antifolate composition to act as a substrate for FPGS (e.g., human FPGS or rat liver FPGS) are readily available and can be routinely performed.
In some embodiments, the uptake rate of naked γ pantol compositions disclosed herein (e.g., γ pantol not associated with a delivery vehicle) by hepatocytes is significantly reduced compared to the uptake rate of an antifolate under physiological conditions. In some embodiments, the naked γ pantol composition has a hepatocyte uptake rate of less than 30%, 20%, 15% or 10% compared to the rate of antifolate. In other embodiments, the efflux (export) rate of the γ pantol compositions disclosed herein from hepatocytes occurs at a significantly reduced rate (e.g., less than 30%, 20%, 15% or 10%) compared to the rate of antifolates.
In some embodiments, the gamma polyglutamated antifolate compositions provided herein are more cytotoxic to hyperproliferative cells than antifolates. In some embodiments, the hyperproliferative cell is a cancer cell. In some embodiments, the hyperproliferative cell is a colorectal cancer cell, a colon cancer cell, a breast cancer cell, or an ovarian cancer cell. In some embodiments, the cancer cell is a mesothelioma cell or a non-small cell lung cancer cell. In some embodiments, cytotoxicity is measured in an in vitro assay. In some embodiments, the gamma polyglutamated antifolate is a hexaglutamated antifolate.
In some embodiments, the gamma polyglutamated antifolate compositions provided herein have lower toxic side effects compared to antifolates. In some embodiments, the gamma polyglutamated antifolate compositions provided herein are less toxic to non-hyperproliferative cells than antifolates. In some embodiments, the gamma polyglutamated antifolate compositions provided herein are less toxic to neutrophils, hepatocytes, or colonic epithelial cells as compared to the antifolate. In some embodiments, the neutrophil is a human neutrophil, a differentiated human neutrophil, or a neutrophil differentiated from a CD34+ cell. In some embodiments, the hepatocyte is an AML12 hepatocyte. In some embodiments, the colonic epithelial cells are CCD841 colonic epithelial cells. In some embodiments, toxicity is measured in an in vitro assay. In some embodiments, the gamma polyglutamated antifolate is a hexaglutamated antifolate.
In some embodiments, the gamma polyglutamated antifolate compositions provided herein have lower toxic side effects compared to antifolates. In some embodiments, the gamma polyglutamated antifolate compositions provided herein cause fewer or less severe side effects in an in vivo assay compared to an antifolate. In some embodiments, the in vivo assay is an in vivo murine model. In some embodiments, the gamma polyglutamated antifolate compositions provided herein cause fewer or less severe hematologic or hepatotoxic side effects as compared to antifolates. In some embodiments, hematological side effects are assessed by measuring mean neutrophil, mean leukocyte, or mean platelet counts. In some embodiments, hepatotoxic side effects are assessed by measuring serum aspartate transaminase (AST), serum alanine transaminase (ALT), and/or serum albumin levels. In some embodiments, the in vivo assay comprises administering 40mg/kg or 80mg/kg of a gamma polyglutamated antifolate composition once per week for 4 weeks. In some embodiments, the gamma polyglutamated antifolate is a hexaglutamated antifolate.
In some embodiments, treatment with the gamma polyglutamated antifolate compositions provided herein does not induce significant hematologic or hepatotoxic side effects in an in vivo murine model. In some embodiments, hematological side effects are assessed by measuring mean neutrophil, mean leukocyte, or mean platelet counts. In some embodiments, hepatotoxic side effects are assessed by measuring serum aspartate transaminase (AST), serum alanine transaminase (ALT), and/or serum albumin levels. In some embodiments, the gamma polyglutamated antifolate compositions provided herein do not significantly reduce mean neutrophil, mean leukocyte or mean platelet counts. In some embodiments, the gamma polyglutamated antifolate compositions provided herein do not significantly increase serum aspartate transaminase (AST) and serum alanine transaminase (ALT) levels. In some embodiments, the gamma polyglutamated antifolate compositions provided herein do not significantly reduce serum albumin levels. In some embodiments, the in vivo assay comprises administering 40mg/kg or 80mg/kg of a gamma polyglutamated antifolate composition once per week for 4 weeks. In some embodiments, the gamma polyglutamated antifolate is a hexaglutamated antifolate.
In some embodiments, the gamma polyglutamated antifolate composition is free of fluorine atoms. In some embodiments, the gamma polyglutamated antifolate composition is free of 4-fluoro glutamyl.
Gamma polyglutamated antifolate (gamma PANTIFOL) compositions and uses thereof may be further described in International application Ser. No. PCT/US2017/046667, as well as U.S. patent application Ser. Nos. 62/630,824, 62/630,613, 62/630,713, 62/630,620, 62/627,733, 62/630,625, 62/630,652, 62/627,732, 62/636,289, 62/630,751, 62/630,821, 62/627,741, and 62/583,432, the disclosures of each of which are incorporated herein by reference in their entirety.
A. Gamma polyglutamated antifolate analogs and derivatives
The present disclosure also encompasses gamma polyglutamated antifolate derivatives and analogs. It is contemplated that the compositions and methods disclosed herein are applicable to any and every known derivative or analog of polyglutamated antifolates. In some embodiments, the analogs correspond to modified forms of the antifolate wherein the glutamyl group of the antifolate is not linked to the remainder of the antifolate molecule by a gamma peptide bond linkage. In some embodiments, the analog is a variant form of an antifolate, wherein the glutamyl group in the antifolate is in the D form. In some embodiments, the polyglutamated form of the antifolate or the polyglutamated antifolate analog or derivative is not fluorinated.
In some embodiments, the antifolate is selected from the group consisting of: methotrexate derivatives bearing an indoline ring and a modified ornithine, methotrexate derivatives bearing an indoline ring and a modified glutamic acid, methotrexate derivatives bearing an alkyl-substituted phenyl ring C, methotrexate derivatives bearing a benzoxazine moiety, methotrexate derivatives bearing a benzothiazine moiety, 10-deazaaminopterin analogs, 5-deazaaminopterin methotrexate analogs, 5, 10-deazaaminopterin methotrexate analogs, methotrexate derivatives bearing an indoline moiety, lipophilic amide methotrexate derivatives, methotrexate analogs containing L-threo- (2S,4S) -4-fluoro-glutamic acid, methotrexate analogs containing DL-3, 3-difluoroglutamic acid, methotrexate tetrahydroquinazoline analogs, N- (ac-aminoacyl) methotrexate derivatives, biotin methotrexate derivatives, D-glutamic methotrexate analogs, D-erythro, threo-4-fluoroglutamic methotrexate analogs, beta, gamma-methanomethotrexate analogs, 10-deazaaminopterin (10-EDAM) analogs, gamma-tetrazole methotrexate analogs, N- (L-alpha-aminoacyl) methotrexate derivatives, meta-isomers of aminopterin, ortho-isomers of aminopterin, hydroxymethyl methotrexate, gamma-fluoro methotrexate, polyglutamyl methotrexate derivatives, gem-diphosphonic acid methotrexate analogs (see, e.g., WO1988/06158, the contents of which are incorporated herein by reference in their entirety), alpha-substituted methotrexate analogs, Gamma-substituted methotrexate analogs, 5-methyl-5-deaminomethotrexate analogs (see, e.g., U.S. Pat. No. 4,725,687, the contents of which are incorporated herein by reference in their entirety), N-acyl-Nalpha- (4-amino-4-deoxypteroyl) -L-ornithine derivatives, 8-deazamethotrexate analogs, acivicin methotrexate analogs, polymeric cisplatin methotrexate derivatives, methotrexate-gamma-dimyristoyl phosphatidylethanolamine, methotrexate polyglutamate analogs, poly-gamma-glutamyl methotrexate derivatives, deoxyuridylate methotrexate derivatives, iodoacetyl lysine methotrexate analogs, 2, omega-diaminoalkanoic acid-containing methotrexate analogs-polyglutamate methotrexate derivatives, 5-methyl-5-deaza analogs, quinazoline methotrexate analogs, pyrazine methotrexate analogs, cysteine or homocysteine methotrexate analogs (see, e.g., U.S. Pat. No. 4,490,529 and EPA 0142220, the contents of each of which are incorporated herein by reference in their entirety), gamma-tert-butyl methotrexate ester, fluorinated methotrexate analogs, folic acid methotrexate analogs, phosphonoglutamic acid analogs, poly (L-lysine) methotrexate conjugates, dilysine or trilysine methotrexate derivatives, 7-hydroxy methotrexate, poly-gamma-glutamyl methotrexate analogs, 3 ', 5' -dichloro methotrexate, diazoketone or chloromethyl ketone methotrexate analogs, 10-propargyl aminopterin, alkyl methotrexate homologs, lectin derivatives of methotrexate, Polyglutamate methotrexate derivatives, halogenated methotrexate derivatives, 8-alkyl-7, 8-dihydro analogs, 7-methyl methotrexate derivatives, dichloro methotrexate, lipophilic methotrexate derivatives, 3 ', 5' -dichloro methotrexate, deazaaminopterin analogs, and MX068, or their stereoisomers.
In a further embodiment, the gamma polyglutamated antifolate derivative or analog has a variant polyglutamate chain. In some embodiments, the polyglutamate chains contain one or more natural or synthetic residues other than glutamate. In some embodiments, the polyglutamate chains contain one or more glutamyl groups that do not contain an amide linkage. In other embodiments, one or more glutamyl groups of the polyglutamate chain are derivatized.
B. Gamma ANTIFOL-PG synthesis
The antifolate polyglutamate compositions provided herein can be obtained by the following synthetic procedures known in the art. Procedures for the synthesis of antifolates (including various pharmaceutically acceptable salts or acids (e.g., disodium antifolate) as well as crystalline and amorphous forms) and intermediates for the synthesis of antifolates include, but are not limited to, U.S. patent nos. 2,512,572; 3,892,801, respectively; 3,989,703, respectively; 4,057,548, respectively; 4,067,867, respectively; 4,079,056, respectively; 4,080,325, respectively; 4,106,488, respectively; 4,136,101, respectively; 4,224,446, respectively; 4,306,064, respectively; 4,374,987, respectively; 4,421,913, respectively; 4,558,690, respectively; 4,662,359, respectively; and 4,767,859; and those described in Calvert, Semin. Oncol.26:3-10 (1999)).
The antifolate polyglutamate compositions provided herein can be obtained by the following synthetic procedure using available reagents and synthetic intermediates. Glutamyl residues can be added to the glutamyl residues of antifolates using synthetic procedures known in the art. In some embodiments, the glutamyl residue is added continuously to the glutamyl residue of the antifolate. In further embodiments, polyglutamate is added to the glutamyl residues of the antifolate using a "click chemistry" method or other bioconjugate chemistry known to those skilled in the art. Alternatively, peptides of glutamyl residues of the desired length can be produced and added to precursors of antifolates without glutamyl residues. Peptides may be produced using synthetic methods known in the art. In some embodiments, the initial glutamyl residue is bonded to the Wang resin, and additional glutamyl residues are added sequentially via solid phase peptide synthesis using F-moc chemistry. After addition of the final glutamyl residue, the antifolate precursor is coupled to the peptide and the molecule is cleaved from the resin.
C. Gamma polyglutamated antifolate complex
The present inventors have surprisingly found that polyglutamated antifolates such as antifolates (γ pantol) are capable of forming complexes with other compositions comprising therapeutic agents, including cytotoxic compounds such as platinum-based compounds. Thus, in some embodiments, the present disclosure provides a complex of γ pantol (e.g., γ pantol disclosed herein) and a therapeutic agent or a salt or acid thereof. In some embodiments, the present disclosure provides a complex of γ pantol according to any one of [1] to [11] of the detailed description section and a therapeutic agent or a salt or acid thereof. In some embodiments, the γ pantol/complex comprises γ pantol and a therapeutic agent. In some embodiments, the therapeutic agent is a cytotoxic compound, such as a chemotherapeutic agent. In other embodiments, the γ pantol/complex contains a platinum-based drug, such as a platinum-based chemotherapeutic (e.g., cisplatin, carboplatin, and oxaliplatin). In other embodiments, the γ pantol/complex contains a taxane-based chemotherapeutic (e.g., carboplatin and cisplatin). In other embodiments, the γ pantol/complex comprises a cyclodextrin. In other embodiments, the γ pantol/complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] [67 ].
In other embodiments, the gamma pantol/therapeutic agent complex comprises one or more gamma pantols containing 2 to 150, 2 to 100, 2 to 75, 2 to 50, 2 to 24, 2 to 30, 2 to 20, 2 to 19, 2 to 15, 2 to 10, or 2 to 5 glutamyl groups. In some embodiments, the gamma pantol/therapeutic agent complex comprises one or more gamma pantols containing 3-10, 3-9, 3-8, or 3-7 glutamyl groups or any range therebetween. In other embodiments, the gamma pantol/therapeutic agent complex comprises one or more gamma pantols containing 4-10, 4-9, 4-8, 4-7, 4-6, or 4-5 glutamyl groups or any range therebetween. In a particular embodiment, the complex comprises one or more gamma pantols containing 3-10 glutamyl groups. In other embodiments, the gamma pantol/therapeutic agent complex comprises one or more gamma pantols containing 3-7 glutamyl groups. In another embodiment, the γ pantol/therapeutic agent complex comprises one or more γ pantol containing 5 glutamyl groups. In another embodiment, the γ pantol/therapeutic agent complex comprises one or more γ pantol containing 6 glutamyl groups. In some embodiments, the therapeutic agent is a cytotoxic compound or a salt or acid thereof. In another embodiment, the therapeutic agent is a chemotherapeutic agent or a salt or acid thereof. In another embodiment, the therapeutic agent is a platinum-based drug. In another embodiment, the therapeutic agent is a taxane-based drug. In further embodiments, the molar ratio of gamma pantol/therapeutic agent in the complex is in the range of 1-10: 1. In some embodiments, the molar ratio of gamma pantol/therapeutic agent in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the γ pantol/therapeutic agent complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art). In some embodiments, the molar ratio of gamma pantol/therapeutic agent in the complex is 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the molar ratio of γ pantol/therapeutic agent in the complex is: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In some embodiments, the γ pantol/therapeutic agent complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art). In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In an alternative embodiment, the γ pantol complex comprises γ pantol and cyclodextrin. In some embodiments, the γ pantol complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises an antifolate described in part I. In some embodiments, the molar ratio of γ pantol (e.g., γ pantol salt)/cyclodextrin in the complex is in the range of 1-20:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/cyclodextrin in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/cyclodextrin in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/cyclodextrin in the complex is: 1. 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/cyclodextrin in the complex is: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In other embodiments, the molar ratio of gamma pantol/cyclodextrin in the complex is in the range of 1:1 to 20, 1:1 to 10, or 1:2 to 8, or any range therebetween. In some embodiments, the molar ratio of γ pantol/cyclodextrin in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/cyclodextrin in the complex is: 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the γ pantol/cyclodextrin complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art). In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In some embodiments, the present disclosure provides a composition comprising a γ pantol/platinum-based chemotherapeutic complex. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the platinum-based chemotherapeutic agent is selected from the group consisting of: cisplatin, carboplatin, and oxaliplatin, or salts or acids thereof. In other embodiments, the γ pantol/platinum-based chemotherapeutic complex comprises an analog of cisplatin, carboplatin, oxaliplatin, or a salt or acid thereof. In some embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is in the range of 1-20:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/platinum-based analog in the complex is 11:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/platinum-based analog in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In other embodiments, the molar ratio of gamma pantol/platinum-based chemotherapeutic agent in the complex is in the range of 1:1-20, 1:1-10, or 1:2-8, or any range therebetween. In some embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In further embodiments, the γ PANTIFOL// platinum-based agent complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In additional embodiments, the γ pantol/platinum-based chemotherapeutic complex comprises an analog of cisplatin, carboplatin, oxaliplatin, or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the molar ratio of γ pantol/platinum-based analog in the complex is in the range of 1-20:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/platinum-based analog in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/platinum-based analog in the complex is 11:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/platinum-based analog in the complex is 11:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/platinum-based agent in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In further embodiments, the γ PANTIFOL// platinum-based analog complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In other embodiments, the present disclosure provides a complex comprising γ pantol and cisplatin or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises an antifolate described in part I. In some embodiments, the molar ratio of γ pantol/cisplatin (or cisplatin salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/cisplatin (or cisplatin salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/cisplatin (or cisplatin salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/cisplatin (or cisplatin salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/cisplatin (or cisplatin salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/cisplatin (or cisplatin salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/cisplatin (or cisplatin salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In additional embodiments, the γ PANTIFOL// cisplatin (or cisplatin salt or acid) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In another embodiment, the present disclosure provides a complex comprising γ pantol and carboplatin or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I herein. In some embodiments, the molar ratio of γ pantol/carboplatin (or carboplatin salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In other embodiments, the molar ratio of gamma pantol/carboplatin (or carboplatin salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/carboplatin (or carboplatin salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/carboplatin (or carboplatin salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/carboplatin (or carboplatin salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/carboplatin in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/carboplatin in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In further embodiments, the γ pantol/carboplatin (or carboplatin salt or acid) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In another embodiment, the present disclosure provides a complex comprising γ pantol and oxaliplatin or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the molar ratio of γ pantol/oxaliplatin (or oxaliplatin salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/oxaliplatin (or oxaliplatin salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/oxaliplatin (or oxaliplatin salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/oxaliplatin (or oxaliplatin salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/oxaliplatin (or oxaliplatin salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/oxaliplatin (or oxaliplatin salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/oxaliplatin (or oxaliplatin salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In a further embodiment, the γ pantol/oxaliplatin (or oxaliplatin salt or acid) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In additional embodiments, the present disclosure provides a complex comprising γ pantol and a platinum-based chemotherapeutic agent ("platinum") selected from the group consisting of: nedaplatin, heptaplatin, lobaplatin, satraplatin, carboplatin (paratoplatin), cisplatin (platinol), cycloplatin, dexormaplatin, spiroplatin, picoplatin, triplatin, tetraplatin, iproplatin, ormaplatin, zeniplatin, platinum-triamine, traplatin, enloplatin, JM216, NK121, CI973, DWA 2114R, NDDP, and dedaplatin, or salts or acids thereof. In other embodiments, the γ PANTIFOL/platinum-based chemotherapeutic complex comprises an analog of nedaplatin, heptaplatin, lobaplatin, satraplatin, carboplatin, cisplatin, cycloplatin, dexormaplatin, spiroplatin, picoplatin, triplatin, tetraplatin, iproplatin, ormaplatin, zeniplatin, platinum-triamine, traplatin, enloplatin, JM216, NK121, CI973, DWA 2114R, NDDP, or dedaplatin, or a salt or acid thereof. In some embodiments, the molar ratio of γ pantol/platinum-based chemotherapeutic ("platinum") (or platinum-based chemotherapeutic salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In other embodiments, the molar ratio of γ pantol/platinum (or platinum salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/platinum (or platinum salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/platinum (or platinum salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/platinum (or platinum salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/platinum (or platinum salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/platinum (or platinum salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In further embodiments, the γ pantol/platinum (or salt or acid or analog thereof) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In some embodiments, the present disclosure provides a composition comprising a γ pantol/taxane-based chemotherapeutic (taxane) complex. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the taxane-based chemotherapeutic agent is selected from: paclitaxel (PTX), Docetaxel (DTX), Larotaxel (LTX) and Cabazitaxel (CTX), or salts or acids thereof. In some embodiments, the molar ratio of γ pantol/taxane (or taxane salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/taxane (or taxane salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/taxane (or taxane salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/taxane (or taxane salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/taxane (or taxane salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/taxane (or taxane salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/taxane (or taxane salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In further embodiments, the γ pantol/taxane (or taxane salt or acid) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In additional embodiments, the present disclosure provides a complex comprising γ pantol and Paclitaxel (PTX) or a salt or acid thereof. In other embodiments, the gamma pantol/paclitaxel (or paclitaxel salt or acid) complex comprises an analog of Paclitaxel (PTX) or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the molar ratio of γ pantol/paclitaxel (or paclitaxel salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/paclitaxel (or paclitaxel salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/paclitaxel (or paclitaxel salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/paclitaxel (or paclitaxel salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/paclitaxel (or paclitaxel salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/paclitaxel (or paclitaxel salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/paclitaxel (or paclitaxel salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In further embodiments, the γ pantol/paclitaxel (or paclitaxel salt or acid) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In additional embodiments, the present disclosure provides a complex comprising γ pantol and Docetaxel (DTX) or salts or acids thereof. In other embodiments, the γ PANTIFOL/docetaxel complex comprises an analog of Docetaxel (DTX) or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the molar ratio of γ pantol/docetaxel (or docetaxel salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/docetaxel (or docetaxel salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/docetaxel (or docetaxel salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/docetaxel (or docetaxel salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/docetaxel (or docetaxel salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of γ pantol/docetaxel (or docetaxel salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/docetaxel (or docetaxel salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In additional embodiments, the γ pantol/docetaxel (or docetaxel salt or acid) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In additional embodiments, the present disclosure provides a complex comprising γ pantol and raloxitol (LTX) or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the molar ratio of gamma pantol/raloxitol (or raloxitol salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In other embodiments, the molar ratio of gamma pantol/raloxitol (or raloxitol salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of gamma pantol/raloxitol (or raloxitol salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of gamma pantol/raloxitol (or raloxitol salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of gamma pantol/raloxitol (or raloxitol salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1, or >50: 1. In some embodiments, the molar ratio of γ pantol/raloxitol (or raloxitol salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/raloxitol (or raloxitol salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In additional embodiments, the γ pantol/raloxitol (or raloxitol salt or acid) complex is encapsulated in liposomes. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In additional embodiments, the present disclosure provides a complex comprising γ pantol and Cabazitaxel (CTX) or salts or acids thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. In some embodiments, the molar ratio of γ pantol/cabazitaxel (or cabazitaxel salt or acid) in the complex is in the range of 1-20:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/cabazitaxel (or cabazitaxel salt or acid) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of γ pantol/cabazitaxel (or cabazitaxel salt or acid) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of γ pantol/cabazitaxel (or cabazitaxel salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of γ pantol/cabazitaxel (or cabazitaxel salt or acid) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1, or >50: 1. In some embodiments, the molar ratio of γ pantol/cabazitaxel (or cabazitaxel salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of γ pantol/cabazitaxel (or cabazitaxel salt or acid) in the complex is: 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In additional embodiments, the γ pantol/cabazitaxel (or cabazitaxel salt or acid) complex is encapsulated in liposomes. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In a further embodiment, the present disclosure provides a complex comprising γ pantol and another antimetabolite, or a salt or acid thereof. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol complex comprises a polyglutamated antifolate agent described in section I. An antimetabolite is a chemical substance that has a structure similar to a metabolite required for normal biochemical reactions, but differs from the metabolite sufficiently to interfere with one or more normal functions of a cell (such as cell division). In some embodiments, the present disclosure provides a complex comprising γ pantol and an Antifolate (ANTIFOL) or a salt or acid thereof. In some embodiments, the present disclosure provides a complex comprising γ pantol and an antimetabolite selected from the group consisting of: gemcitabine, fluorouracil, capecitabine, antifolates (e.g., antifolate, raltitrexed), tegafur, cytarabine, thioguanine, 5-azacytidine, 6-mercaptopurine, azathioprine, 6-thioguanine, pentostatin, fludarabine phosphate and cladribine, and pharmaceutically acceptable salts or acids, acids or derivatives of any of these. In some embodiments, the molar ratio of gamma pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) in the complex is in the range of 1-20:1, or any range therebetween. In other embodiments, the molar ratio of gamma pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) in the complex is in the range of 1-10:1, or any range therebetween. In other embodiments, the molar ratio of gamma pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) in the complex is in the range of 2-8:1, or any range therebetween. In some embodiments, the molar ratio of gamma pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, or 20: 1. In some embodiments, the molar ratio of gamma pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50: 1 or >50: 1. In some embodiments, the molar ratio of gamma pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) in the complex is 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, or 1: 20. In some embodiments, the molar ratio of gamma pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) in the complex is 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In additional embodiments, the γ pantol/antimetabolite (or antimetabolite salt or acid, or prodrug) complex is encapsulated in a liposome. In some embodiments, the liposome is Lp- α pantol according to any one of [12] - [67] of the detailed description section.
In additional embodiments, the present disclosure provides complexes of γ pantol (e.g., γ pantol disclosed herein) and cyclodextrins. Cyclodextrins (CDs) are groups of cyclic oligosaccharides that have been shown to improve the physicochemical properties of many drugs by forming complexes. CD is a cyclic oligosaccharide consisting of several D-glucose units linked by alpha- (1,4) bonds. This annular configuration provides a hydrophobic interior cavity and renders the CD frustoconical. Many hydroxyl groups are located at the edge of the ring, which makes CD lipophilic and water soluble. As a result, CD is able to form complexes with a wide variety of hydrophobic agents and thus alter the physico-chemical properties of these complexing agents. In some embodiments, the complex comprises γ pantol according to any one of [1] to [11] of the detailed description section.
Unless otherwise indicated herein, the term "cyclodextrin" or "CD" generally refers to a parent or derived cyclic oligosaccharide containing a variable number of (α -1,4) -linked D-glucopyranoside units, which is capable of forming a complex with an antifolate-PG. Each cyclodextrin glucopyranoside subunit has secondary hydroxyl groups at the 2 and 3 positions, and a primary hydroxyl group at the 6 position. The terms "parent", "underivatized" or "inert" cyclodextrin refer to a cyclodextrin containing D-glucopyranoside units having the basic formula C6H12O6 and a glucose structure without any additional chemical substitution (e.g., an α -cyclodextrin consisting of 6D-glucopyranoside units, a β -cyclodextrin consisting of 7D-glucopyranoside units, and a γ -cyclodextrin consisting of 8D-glucopyranoside units). The physical and chemical properties of the parent cyclodextrins can be modified by derivatizing the hydroxyl groups with other functional groups. Any substance that is located within the internal phase of a cyclodextrin is said to "complex" with the cyclodextrin, or form a complex (inclusion complex) with the cyclodextrin.
As used herein, there is no particular limitation on the cyclodextrin component of the γ pantol/cyclodextrin complex, as long as the cyclodextrin can form a complex with γ pantol. In particular embodiments, the cyclodextrin has been derivatized to carry ionizable (e.g., weakly basic and/or weakly acidic) functional groups to facilitate complex formation with γ pantol and/or liposome encapsulation.
It is known that modifying the hydroxyl groups of cyclodextrins with ionizable chemical groups (e.g., those that face away from the cyclodextrin internal phase) can facilitate loading of cyclodextrins and therapeutic agents complexed with cyclodextrins. In some embodiments, the cyclodextrin of the γ pantol/cyclodextrin complex has at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 hydroxyl groups substituted with an ionizable chemical group. The term "charged cyclodextrin" refers to a cyclodextrin having one or more of its hydroxyl groups substituted with a charged moiety. Such a moiety may itself be a charged group, or it may comprise an organic moiety (e.g., a C1-C6 alkyl or C1-C6 alkyl ether moiety) substituted with one or more charged moieties.
In some embodiments, the "ionizable" or "charged" moiety of the CD derivative is weakly ionizable. Weakly ionizable moieties are those moieties that are weakly basic or weakly acidic. According to CH3-W, the weakly basic functional group (W) has a pKa between about 6.0-9.0, 6.5-8.5, 7.0-8.0, 7.5-8.0, and any range therebetween (inclusive). Similarly, the weak acidic functional group (X) has a logarithmic dissociation constant (pKa) according to CH3-X of between about 3.0-7.0, 4.0-6.5, 4.5-6.5, 5.0-6.0, 5.0-5.5, and any range therebetween, inclusive. Representative anionic moieties include, but are not limited to, carboxylate, carboxymethyl, succinyl, sulfonyl, phosphate, sulfoalkyl ether, sulfatocarbonate, thiocarbonate, dithiocarbonate, phosphate, phosphonate, sulfonate, nitrate, and borate groups. Representative cationic moieties include, but are not limited to, amino, guanidine, and quaternary ammonium groups.
In another embodiment, the derivatized cyclodextrin is a "polyanion" or "polycation". Polyanions are derivatized cyclodextrins having more than one negatively charged group resulting in a net negative ionic charge of more than two units. Polycations are derivatized cyclodextrins that have more than one positively charged group, resulting in a net positive ionic charge of more than two units.
In another embodiment, the derivatized cyclodextrin is a "chargeable amphiphile". By "chargeable" is meant that the pK of the amphiphile is in the range of pH 4 to pH 8 or 8.5. Thus, the chargeable amphiphile may be a weak acid or a weak base. "amphoteric" herein refers to a derivatized cyclodextrin having ionizable groups of both anionic and cationic character, wherein: (a) at least one and optionally both of the cationic and anionic amphiphiles are chargeable, having at least one charged group with a pK between 4 and 8 to 8.5; (b) the cationic charge predominates at pH 4, and (c) the anionic charge predominates at pH 8 to 8.5.
In some embodiments, the derivatized cyclodextrin, whether polyionic, amphiphilic or otherwise, "ionizable" or "charged" as a whole is weakly ionizable (i.e., has a pKai between about 4.0-8.5, 4.5-8.0, 5.0-7.5, 5.5-7.0, 6.0-6.5, and any range therebetween, inclusive)).
Any, some, or all of the hydroxyl groups in any, some, or all of the α -D-glucopyranoside units of the cyclodextrin can be modified to an ionizable chemical group as described herein. Because of the different chemical reactivity of each cyclodextrin hydroxyl group, reaction with the modifying moiety can produce an amorphous mixture of positional and optical isomers. Alternatively, certain chemical methods can react a pre-modified α -D-glucopyranoside unit to form a homogeneous product.
The aggregation substitution that occurs with cyclodextrin derivatives in a mixture is described in terms of the degree of substitution. For example, a 6-ethylenediamino- β -cyclodextrin having a degree of substitution of seven would consist of an isomeric distribution of 6-ethylenediamino- β -cyclodextrin in which the average number of ethylenediamino groups per 6-ethylenediamino- β -cyclodextrin molecule is seven. The degree of substitution of the mixture of cyclodextrin derivatives can be routinely determined using mass spectrometry or nuclear magnetic resonance spectrometry.
In one embodiment, at least one hydroxyl moiety facing away from the cyclodextrin interior is substituted with an ionizable chemical group. For example, the C2, C3, C6, C2 and C3, C2 and C6, C3 and C6, and all three C2-C3-C6 hydroxyl groups of at least one α -D-glucopyranoside unit are substituted with an ionizable chemical group. Any such combination of hydroxyl groups can be similarly combined with at least two, three, four, five, six, seven, eight, nine, ten, eleven, up to all of the α -D-glucopyranoside units in the modified cyclodextrin, as well as with any degree of substitution described herein. One such derivative is sulfoalkyl ether cyclodextrin (SAE-CD). Sulfobutyl ether derivatives of beta cyclodextrin (SBE-beta-CD) have demonstrated significantly improved water solubility compared to the parent cyclodextrin.
Additional cyclodextrin derivatives that can be complexed with the therapeutic agent in the disclosed liposome compositions include sugammadex or Org-25969, wherein the 6-hydroxy group on the gamma-CD has been replaced with a carboxythioacetate ether linkage and a hydroxybutenyl-beta-CD. Alternatives to cyclodextrins include: 2, 6-di-O-methyl- β -CD (DIMEB), 2-hydroxypropyl-3-cyclodextrin (HP- β -CD), randomly methylated- β -cyclodextrin (RAMEB), sulfobutyl ether β -cyclodextrin (SBE- β -CD), and sulfobutyl ether- γ -cyclodextrin (SBE γ CD), sulfobutylated β -cyclodextrin sodium salt, (2-hydroxypropyl) - α -cyclodextrin, (2-hydroxypropyl) - β -cyclodextrin, (2-hydroxypropyl) - γ -cyclodextrin, 2, 6-di-O-methyl) - β -cyclodextrin (DIMEB-50 hepta), 2,3, 6-tri-O-methyl) - β -cyclodextrin (TRIMEB hepta), Methyl-beta-cyclodextrin, octa (6-deoxy-6-iodo) -gamma-cyclodextrin, and octa (6-deoxy-6-bromo) -gamma-cyclodextrin.
In some embodiments, the one or more cyclodextrins have a high solubility in water, such that a greater amount of the cyclodextrins is entrapped in the liposome internal phase. In some embodiments, the water solubility of the cyclodextrin is at least 10mg/mL, 20mg/mL, 30mg/mL, 40mg/mL, 50mg/mL, 60mg/mL, 70mg/mL, 80mg/mL, 90mg/mL, 100mg/mL or higher. In some embodiments, the water solubility of the one or more cyclodextrins is in the range of 10-150mg/mL, 20-100mg/mL, 20-75mg/mL, and any range therebetween (including endpoints).
In some embodiments, large association constants between cyclodextrin and γ pantol and/or other therapeutic agents complexed with cyclodextrin are preferred and can be obtained by selecting the number of glucose units in the cyclodextrin based on the size of the therapeutic agent (see, e.g., Albers et al, crit. rev. therapy. drug Carrier system.12: 311-337 (1995); Stella et al, toxicol. pathol.36:30-42 (2008). when the association constant is dependent on pH, the cyclodextrin can be selected such that the association constant becomes larger at pH in the liposome phase 200. 200-1,000, 300-750, and any range therebetween.
In some embodiments, the cyclodextrin of the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex is underivatized.
In some embodiments, the cyclodextrin of the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex is derivatized. In other embodiments, the cyclodextrin derivative of the complex has the structure of formula I:
Figure BDA0002715408560000591
Wherein: n is 4, 5 or 6;
wherein R is1、R2、R3、R4、R5、R6、R7、R8And R9Each independently is-H, straight or branched chain C1-C8Alkylene or optionally substituted straight or branched C1-C6Group, wherein R1、R2、R3、R4、R5、R6、R7、R8And R9At least one of which is a straight or branched chain C1-C8Alkylene (e.g. C)1-C8- (alkylene) -SO3 -A group);
in some embodiments, the cyclodextrin derivative of the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex has the structure of formula II:
Figure BDA0002715408560000601
wherein: n is 4, 5 or 6;
wherein R is1、R2、R3、R4、R5、R6、R7、R8And R9Each independently is-O-or-O- (C)2-C6Alkylene) -SO3-a group; wherein R is1And R2At least one of which is independently-O- (C)2-C6Alkylene) -SO3 -A group; and S1、S2、S3、S4、S5、S6、S7、S8And S9Each independently is a pharmaceutically acceptable cation. In other embodiments, the pharmaceutically acceptable cation is selected from: alkali metals, e.g. Li+、Na+Or K+(ii) a Alkaline earth metals, e.g. Ca+2Or Mg+2And ammonium ions and amine cations, such as the cations of (C1-C6) -alkylamines, piperidines, pyrazines, (C1-C6) -alkanolamines and (C4-C8) -cycloalkanolamines. In some embodiments, at least one of R1 and R2 is independently a-O- (C2-C6 alkylene) -SO 3-group that is-O- (CH)2)mA SO 3-group, wherein m is 2 to 6, preferably 2 to 4 (e.g., -O-CH 2S 03-or-O-CH 2S 03-); and S 1、S2、S3、S4、S5、S6、S7、S8And S9Each independently is H or a pharmaceutically acceptable cation including, for example, an alkali metal (e.g., Li)+、Na+、K+) Alkaline earth metal (e.g., Ca)+2、Mg+2) Ammonium ions and amine cations, e.g. (C1-C6) -alkylamines, piperidines, pyrazines, (C6)1-C6) -alkanol-amine and (C)4-C8) -cation of cycloalkanolamine:
in some embodiments, the cyclodextrin derivative of the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex is any one of the cyclodextrin derivatives of U.S. patent nos. 6,133,248, 5,874,418, 6,046,177, 5,376,645, 5,134,127, 7,034,013, 6,869,939; and cyclodextrins disclosed in international application publication No. WO 02005/117911, the contents of each of which are incorporated herein by reference in their entirety for all purposes.
In some embodiments, the cyclodextrin derivative of the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex is a sulfoalkyl ether cyclodextrin. In some embodiments, the cyclodextrin derivative of the complex is sulfobutyl ether-3-cyclodextrin, e.g.
Figure BDA0002715408560000603
(CyDex Pharma.Inc., Lenexa, Kansas). Methods for preparing sulfobutyl ether-3-cyclodextrin and other sulfoalkyl ether cyclodextrins are known in the art.
In some embodiments, the cyclodextrin derivative in the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex is a compound of formula III:
Figure BDA0002715408560000602
Wherein R is equal to:
(a)(H)21-Xor (- (CH)2)4-SO3Na) x, and x ═ 1.0 to 10.0, 1.0 to 5.0, 6.0 to 7.0, or 8.0 to 10.0;
(b)(H)21-Xor (- (CH)2CH(OH)CH3) x, and x ═ 1.0 to 10.0, 1.0 to 5.0, 6.0 to 7.0, or 8.0 to 10.0;
(c)(H)21-Xor (sulfoalkyl ether) x, and x ═ 1.0 to 10.0, 1.0 to 5.0, 6.0 to 7.0, or 8.0 to 10.0; or
(d)(H)21-XOr (- (CH)2)4-SO3Na) x, and x ═ 1.0-10.0, 1.0-5.0, 6.0-7.0, or 8.0-10.0.
In additional embodiments, the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
D. Gamma PANTIFOL delivery vehicle
In alternative embodiments, the present disclosure provides a γ pantol delivery system and its use to deliver a payload of γ pantol to one or more cells in vitro or in vivo. In some embodiments, the γ pantol is complexed with or incorporated into a delivery vehicle. Such delivery vehicles are known in the art and include, but are not limited to, liposomes, lipid spheres, polymers, peptides, proteins, antibodies (e.g., ADCs, such as antibody- γ panantipol conjugates), cellular components, cyclic oligosaccharides (e.g., cyclodextrins), nanoparticles (e.g., lipid nanoparticles, biodegradable nanoparticles, and core-shell nanoparticles), lipoprotein particles, and combinations thereof. In a particular embodiment, the delivery vehicle is a liposome. In other particular embodiments, the delivery vehicle is an antibody or antigen-binding antibody fragment. In some embodiments, the γ pantol delivery system comprises γ pantol according to any one of [1] to [11] of the detailed description section.
E. Liposomes
In some embodiments, the present disclosure provides liposome compositions comprising liposomes encapsulating (i.e., filled with) a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein). In some embodiments, the liposome composition comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome compositions comprise the polyglutamated antifolate agent described in section I. In some embodiments, the liposome is any one of [12] to [67] according to the detailed description. In some embodiments, the liposomes in the liposome composition comprise γ pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups (including glutamyl groups of antifolates). In some embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises two or more glutamyl groups in the L form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in D form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in the D form and two or more glutamyl groups in the L form. In further embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises two or more glutamyl groups having a gamma carboxy linkage. In some embodiments, the liposome composition comprises liposomes comprising a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises an L-gamma-pentaglutaminated antifolate, a D-gamma-pentaglutaminated antifolate, or an L-and D-gamma-pentaglutaminated antifolate. In some embodiments, the liposome composition comprises liposomes comprising a gamma hexaglutaminated antifolate (Lp-gamma pantol). In other embodiments, the liposome comprises an L-gamma-hexaglutaminated antifolate, a D-gamma-hexaglutaminated antifolate, or an L-and D-gamma-hexaglutaminated antifolate. In some embodiments, the liposome composition comprises anionic or neutral liposomes. In some embodiments, the liposome composition comprises cationic liposomes. In some embodiments, the Lp- γ pantol composition is non-pegylated. In some embodiments, the Lp- γ pantol composition is non-targeted (NTLp- γ pantol). In other embodiments, the Lp- γ PANTIFLOL composition comprises a targeting moiety (TLp- γ PANTIFLOL). In some embodiments, the liposome composition comprises liposomes having a diameter in the range of 20nm to 500nm or any range therebetween. In some embodiments, the liposome composition comprises liposomes having a diameter in the range of 20nm to 400nm or any range therebetween. In some embodiments, the liposome composition comprises liposomes having a diameter in the range of 20nm to 200nm or any range therebetween. In other embodiments, the liposome composition comprises liposomes having a diameter in the range of 20nm to 150nm or any range therebetween. In other embodiments, the liposome composition comprises liposomes having a diameter in the range of 80nm to 120nm or any range therebetween. In further embodiments, 30% -70%, 30% -60% or 30% -50% w/w or any range therebetween of gamma polyglutamated antifolate is encapsulated (embedded) in Lp-gamma pantol during the process of preparing liposomes. In some embodiments, the Lp- γ pantol composition comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75% or more than 75% (w/w) of a γ polyglutamated antifolate. In some embodiments, at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75% or more than 75% (w/w) of the gamma polyglutamated antifolate is encapsulated in Lp-gamma pantol during the process of making the liposomes.
In some embodiments, provided liposomes further comprise an immunostimulant, a detectable label, or both disposed on the exterior thereof. The immunostimulatory agent or detectable label can be ionically or covalently bound to the exterior of the liposome, including, for example, a steric stabilizer component optionally bound to the liposome.
The term "immunostimulating agent", also known as "immunostimulant" and "immunostimulator" refers to a substance that stimulates immunity (including a pre-existing immune response) by inducing activation or increasing activity of any component of the immune system. These immune stimulators may include one or more of haptens, adjuvants, protein immune stimulators, nucleic acid immune stimulators, and chemical immune stimulators. Many adjuvants contain substances intended to stimulate an immune response, such as lipid a, proteins of bordetella pertussis or mycobacterium tuberculosis origin. Certain adjuvants are commercially available, for example, Freund's incomplete and complete adjuvants (Difco Laboratories, Detroit, Mich.); merck adjuvant 65(Merck and Company, inc., Rahway, n.j.); AS-2(SmithKline Beecham, Philadelphia, Pa.); aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; a cationically or anionically derivatized polysaccharide; polyphosphazene; biodegradable microspheres; monophosphoryl lipid a and quil a; IFN-alpha, IFN-gamma, FLT 3-ligand; and immunostimulatory antibodies (e.g., anti-CTLA-4, anti-CD 28, anti-CD 3). Cytokines such as GM-CSF, interleukins-2, -7, -12, and-15, and other similar growth factors may also be used as adjuvants. In a preferred embodiment, the immunostimulant may be at least one selected from the group consisting of: fluorescein, DNP, beta glucan, beta-1, 3-glucan and beta-1, 6-glucan. In another preferred embodiment, the immunostimulant is a Toll-like receptor (TLR) modulator. In other embodiments, the Toll-like receptor (TLR) modulator is one or more of: OXPAC, PGPC, eritoran lipids (e.g., E5564), and resolvin.
In some embodiments, provided liposomes further comprise an agent that increases uptake of the liposome into a target cellular compartment comprising cytosol. In some embodiments, the agent provides liposome contents with the ability to bypass lysosomes (e.g., chloroquine). In some embodiments, the agent improves turnover of liposome contents by mitochondria (e.g., sphingomyelin and components of mitochondria).
The detectable label may, for example, comprise at least a radioisotope, a fluorescent compound, a bioluminescent compound, a chemiluminescent compound, a metal chelator, an enzyme, a dye, an ink, a magnetic compound, a biocatalyst or a pigment that is detectable by any suitable method known in the art, such as Magnetic Resonance Imaging (MRI), optical imaging, fluorescence/luminescence imaging or nuclear imaging techniques.
In some embodiments, the immunostimulatory agent and/or detectable marker is attached to the exterior by co-incubating it with the liposome. For example, the immunostimulatory agent and/or detectable label may be associated with the liposome membrane by hydrophobic interactions or by ionic bonds such as avidin/biotin bonds or metal chelate bonds (e.g., Ni-NTA). Alternatively, the immunostimulant or detectable label may be covalently bonded to the exterior of the liposome, for example by covalent bonding to a liposome component or as a steric stabilizer for PEG.
One exemplary reagent is Fluorescein Isothiocyanate (FITC), which unexpectedly acts as both an immune stimulator and a detectable label based on our experiments.
In some embodiments, the liposome further comprises an agent that increases uptake of the liposome into a target cellular compartment that includes cytosol.
In some embodiments, the liposome comprises a mitochondrial targeting agent. In some embodiments, the liposome comprises Triphenylphosphonium (TPP). Methods and mechanisms for surface functionalization of liposomes with TPP are known in the art (e.g., TPP is linked to a lipid anchor via a peg spacer and TPP is modified with stearyl (stearyltriphenylphosphonium (STPP)). Comprising RQIKIWFQNRRMK WKKRKKRRQRRR, RKKRRXRRRGC), or a mitochondrion-penetrating fragment thereof. In some embodiments, the liposome comprises a mitochondrial penetrating polynucleotide sequence selected from the group consisting of: RQIKIWFQNRRMKWKKRKKRRQR RR (SEQ ID NO:1), RKKRRXR RRGC wherein X is any natural or non-natural amino acid (SEQ ID NO:2), CCGCCAAGAAGCG (SEQ ID NO:3), GCGTGCACACGCGCGTAGACTTCCCCCGCAAGTCACTCGTTAGCCCGCCAAGAAGCGACCCCTCCGGGGCGAGCTGAGCGGCGTGGCGCGGGGGCGTCAT (SEQ ID NO:4), ACGTGCATACGCACGTAGACATTCCCCGCTTCCCACTCCAAAGTCCGCCAAGAAGCGTATC CCGCTGAGCGGCGTGGCGCGGGGGCGTCATCCGTCAGCTC (SEQ ID NO:5) or ACTTCCCCCGCAAGTCACTCGTTAGCCCGCCAAGAAGCGACCCCTCCGGGGCG AGCTG (SEQ ID NO:6)), or a mitochondrion-penetrating fragment thereof.
In some embodiments, the liposomes in the provided liposome compositions comprise a mitochondrial penetrating agent selected from the group consisting of: guanidine-rich peptoids, tetraguanidines, triguanidines, bis-monoguanidines, guanidine-rich polyurethanes, beta-oligoarginine, proline-rich dendrimers, and phosphonium salts (e.g., methyltriphenylphosphonium and/or tetraphenylphosphonium).
In some embodiments, the liposomes in provided liposome compositions comprise sphingomyelin and/or stearyl-octa-arginine. In some embodiments, the liposome comprises sphingomyelin and/or stearyl-octa-arginine. In some embodiments, the liposome comprises DOPE, sphingomyelin, stearyl-octa-arginine sphingomyelin, and stearyl-octa-arginine. In some embodiments, the liposomes comprise DOPE, sphingomyelin, stearyl-octa-arginine sphingomyelin, and stearyl-octa-arginine in a 9:2:1 molar ratio. In some embodiments, the liposome comprises
Figure BDA0002715408560000631
A system or a variant thereof.
In some embodiments, the liposomes in the provided liposome compositions comprise an agent, such as a cell penetrating agent, that facilitates delivery of the liposome across the cell membrane and provides the liposome with the ability to bypass the endocytic pathway and the harsh environment of the lysosome. Cell penetrating agents are known in the art and are routinely used and suitable for making and using the provided liposome compositions. In some embodiments, the cell penetrating/lysosomal bypassing agent is chloroquine. In some embodiments, the cell penetrating agent is a cell penetrating peptide. In some embodiments, the liposomes in the provided liposome compositions comprise a cell penetrating agent selected from the group consisting of: RKKRRQRRR (SEQ ID NO:7), GRKKRRQRRRTPQ (SEQ ID NO:8), YGRKKRRQRRR (SEQ ID NO:9), AAVAL LPAVLLALLA (SEQ ID NO:10), MGLGLHLLVLAAALQ (SEQ ID NO:11), GALFL GFLGAAGSTM (SEQ ID NO:12), AGYLLGKINLKALAALAKKIL (SEQ ID NO:13), RVIRVWFQNKRCKDKK (SEQ ID NO:14), RQIKFQNRRMKWKK (SEQ ID NO:15), GLFEAIAGFIENGWEGMIDG (SEQ ID NO:16), GWTLNSAGYLLGKIN (SEQ ID NO:17), RSQSRSRYYRQRQRS (SEQ ID NO:18), LAIPEQEY (SEQ ID NO:19), LGIAEQEY (SEQ ID NO:20), LGIPAQAQEY (SEQ ID NO:21), IPAEY (SEQ ID NO:22), IPAQAY (SEQ ID NO:23), EAIALGELGY (SEQ ID NO:24), IPAY (SEQ ID NO:25), SEQ ID NO: EQAY (SEQ ID NO:26), AY LLIILRRRIRKQAHAHSK), EALGEALGEALGEALGELGE (SEQ ID NO:24), EPAY (SEQ ID NO: LLIILRRRIRKQAHAHSK), SEQ ID NO:24, KLALKLALKALKAALKLA (SEQ ID NO:30), KETWWETWWTEWSQPKKKRKV (SEQ ID NO:31), DHQLNPAF (SEQ ID NO:32), DPKGDPKG (SEQ ID NO:33), VTVTVTVTVTGKGDPKPD (SEQ ID NO:34), RQIKIWFQNRRMKWKK (SEQ ID NO:35), GRKKRRQRRPPQ (SEQ ID NO:36), GWTLNSAGYLLGKINLKALAAL AKKIL (SEQ ID NO:37), GRKKRRQRRRR (SEQ ID NO:38), RRRRRRRRRRRRRRR (SEQ ID NO:39), RRRRRRRRRRRRRRRRRRRRRR (SEQ ID NO:40), RRRRRRRRR (SEQ ID NO:41), RRRRRRRR RR (SEQ ID NO:42), RRRRRRRRRRR (SEQ ID NO:43) and YTIWMPENPRPGT PCDIFTNSRGKRASNGGG G (R) n, wherein n is 2-15R in L-and/or D form (SEQ ID NO:44), or a cell penetrating fragment thereof.
As noted above, liposomes may contain steric stabilizers which extend their circulation life. For those embodiments incorporating a steric stabilizer, the steric stabilizer may be at least one member selected from the group consisting of: polyethylene glycol (PEG); poly-L-lysine (PLL); monosialoganglioside (GM 1); poly (vinyl pyrrolidone) (PVP); poly (acrylamide) (PAA); poly (2-methyl-2-oxazoline); poly (2-ethyl-2-oxazoline); a phosphatidylpolyglycerol; poly [ N- (2-hydroxypropyl) methacrylamide ]; amphiphilic poly-N-vinylpyrrolidone; an L-amino acid-based polymer; oligomerization of glycerol; copolymers comprising polyethylene glycol and polypropylene oxide; poloxamer 188; and polyvinyl alcohol. In some embodiments, the steric stabilizer or population of steric stabilizers is PEG. In one embodiment, the steric stabilizer is PEG. In another embodiment, the PEG has a number average molecular weight (Mn) of 200 to 5000 daltons. These PEGs may have any structure, such as linear, branched, star-shaped, or comb-shaped structures, and are commercially available.
In some embodiments, the present disclosure provides liposome compositions comprising pegylated liposomes (PLp- γ pantol). In some embodiments, the pegylated liposomes comprise γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the pegylated liposomes comprise a polyglutamated antifolate described in part I. In some embodiments, the pegylated liposomes in the liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises two or more glutamyl groups in the L form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in D form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in the D form and two or more glutamyl groups in the L form. In some embodiments, the liposome composition comprises pegylated liposomes comprising a gamma-tetraglutamated antifolate. In other embodiments, the liposome comprises an L-gamma-tetraglutamylated antifolate, a D-gamma-tetraglutamylated antifolate, or both an L-and a D-gamma-tetraglutamylated antifolate. In additional embodiments, the liposome composition comprises pegylated liposomes comprising a gamma pentaglutaminated antifolate. In other embodiments, the liposome comprises an L-gamma-pentaglutaminated antifolate, a D-gamma-pentaglutaminated antifolate, or an L-and D-gamma-pentaglutaminated antifolate. In some embodiments, the liposome composition comprises a pegylated liposome comprising a gamma hexaglutaminated antifolate. In other embodiments, the liposome comprises an L-gamma-hexaglutaminated antifolate, a D-gamma-hexaglutaminated antifolate, or an L-and D-gamma-hexaglutaminated antifolate. In some embodiments, the liposome composition comprises a pegylated liposome according to any one of [23] and [25] to [64] of the specific embodiments. In some embodiments, the liposome composition comprises an anionic or neutral pegylated liposome. In some embodiments, the liposome composition comprises a cationic pegylated liposome. In some embodiments, the PLp- γ pantol composition is non-targeted (NTPLp- γ pantol). In other embodiments, the PLp- γ pantol composition comprises a targeting moiety (TPLp- γ pantol). In further embodiments, the liposome composition comprises pegylated liposomes comprising 30% -70%, 30% -60%, or 30% -50% liposome-entrapped gamma polyglutamated antifolate agent, or any range therebetween. In some embodiments, the liposome composition comprises pegylated liposomes comprising at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75% liposome-embedded gamma polyglutamated antifolate. In some embodiments, the liposome composition comprises pegylated liposomes having a diameter in the range of 20nm to 200 nm. In other embodiments, the liposome composition comprises pegylated liposomes having a diameter in the range of 80nm to 120 nm.
In some embodiments, provided liposome compositions have greater than 70%, 80%, or 90% of the polyglutamated antifolate agent is pentaglutamated. In some embodiments, provided compositions are provided in which greater than 70%, 80%, or 90% of the polyglutamated antifolate agent is hexaglutamated. In some embodiments, greater than 70%, 80% or 90% of the polyglutamated antifolate agents in the composition have 4-10, 4-6 or more than 5 γ -glutamyl groups.
In some embodiments, greater than 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the polyglutamated antifolate agents in provided liposome compositions are tetraglutamated. In some embodiments, greater than 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the polyglutamated antifolate agents in provided liposome compositions are pentaglutamated. In some embodiments, greater than 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the polyglutamated antifolate agents in provided liposome compositions are hexaglutamated.
In some embodiments, the gamma polyglutamated antifolate composition (e.g., polyglutamate and a delivery vehicle, such as a liposome containing polyglutamate) is in an aqueous solution. In some embodiments, the polyglutamated antifolate composition is administered in the form of a liposomal composition at between about 0.005 to about 5000mg/M2 (square meter body surface area) or between about 0.1 to about 1000mg/M2 or any range therebetween. In some embodiments, the gamma pantol composition is administered in the form of a liposome composition at about 1mg/kg to about 500mg/kg, 1mg/kg to about 250mg/kg, 1mg/kg to about 200mg/kg, 1mg/kg to about 150mg/kg, 1mg/kg to about 100mg/kg, 1mg/kg to about 50mg/kg, about 1mg/kg to about 25mg/kg, about 1mg/kg to about 20mg/kg, about 1mg/kg to about 15mg/kg, about 1mg/kg to about 10mg/kg, or about 1mg/kg to about 5mg/kg, or any range therebetween.
(1) Liposome composition
The lipids and other components of the liposomes contained in the liposome composition may be any lipids, the combination and proportions of lipids or the combination of lipids and other liposome components and their respective proportions are known in the art. However, one skilled in the art will appreciate that liposomal encapsulation of any particular drug, such as, but not limited to, the gamma polyglutamated antifolate agents discussed herein, may involve a great deal of routine experimentation to achieve a useful and functional liposomal formulation. In general, the provided liposomes can have any liposomal structure, e.g., a structure having an interior space isolated from an external medium by one or more lipid bilayers, or any microcapsule having a semi-permeable membrane with a lipophilic central portion, wherein the membrane isolates the interior. The lipid bilayer may be any arrangement of amphipathic molecules characterized by hydrophilic portions (hydrophilic portions) and hydrophobic portions (hydrophobic portions). In general, the amphipathic molecules in the bilayer are arranged in a two dimensional sheet with the hydrophobic portions oriented inward and the hydrophilic portions oriented outward. The amphipathic molecules forming the provided liposomes can be any known or later discovered amphipathic molecules, such as synthetic or naturally derived lipids or biocompatible lipids. Liposomes can also be formed from amphiphilic polymers and surfactants, for example, polymeric bodies and vesicles. For the purposes of this disclosure, but not by way of limitation, these liposome-forming materials are also referred to as "lipids".
The liposome composition formulations provided herein can be in liquid or dry form, such as a dry powder or a dry cake. The dry powder or dry cake can be subjected to primary drying, for example, under lyophilization conditions, or optionally, the dry cake or dry powder can be subjected to only primary drying or both primary and secondary drying. In dry form, the powder or filter cake may for example have a moisture of 1% to 6%, for example between 2% to 5% or between 2% to 4%. An exemplary method of drying is lyophilization (also known as freeze-drying or lyophilization). Any of the compositions and methods of the present disclosure can include liposomes, lyophilized liposomes, or liposomes reconstituted from lyophilized liposomes. In some embodiments, the disclosed compositions and methods include one or more lyoprotectants or cryoprotectants. These protective agents are generally polyhydroxy compounds, such as sugars (mono-, di-and polysaccharides), polyols and their derivatives, glycerol or polyethylene glycols, trehalose, maltose, sucrose, glucose, lactose, dextran, glycerol or aminoglycosides. In other embodiments, the lyoprotectant or cryoprotectant comprises up to 10% or up to 20% of the solution outside the liposome, inside the liposome, or both outside and inside the liposome.
In some embodiments, the liposomes include a steric stabilizer that extends their lifetime in circulation. One or more steric stabilizers such as hydrophilic polymers (polyethylene glycol (PEG)), glycolipids (monosialoganglioside (GM1)) or other substances occupy the space immediately adjacent to the liposome surface and exclude other macromolecules from this space. Thus, access to and binding of plasma opsonin to the liposome surface is hindered, and thus interaction of macrophages with such liposomes or any other clearance mechanism is inhibited, and the lifetime of the liposomes in circulation is enhanced. In some embodiments, the steric stabilizer or population of steric stabilizers is PEG or a combination comprising PEG. In other embodiments, the steric stabilizer is PEG or a combination comprising PEG having a number average molecular weight (Mn) of 200 to 5000 daltons. These PEGs may have any structure, such as linear, branched, star-shaped, or comb-shaped structures, and are commercially available.
In some embodiments, the liposome composition comprises liposomes having a diameter in the range of 20nm to 150nm or any range therebetween. In some embodiments, the liposome composition comprises liposomes containing the γ pantol according to any one of [1] to [11] of the detailed description section and having a diameter in the range of 20nm to 150 nm. In some embodiments, the liposome is a liposome composition according to any one of [12] to [67] of the detailed description section, and has a diameter in the range of 20nm to 150 nm. In other embodiments, the liposome composition comprises liposomes having a diameter in the range of 30nm to 150nm or any range therebetween. In some embodiments, the liposome composition comprises liposomes containing the γ pantol according to any one of [1] to [11] of the detailed description section and having a diameter in the range of 30nm to 150 nm. In some embodiments, the liposome is a liposome composition according to any one of [12] to [67] of the detailed description section, and has a diameter in the range of 30nm to 150nm, or any range therebetween. In other embodiments, the liposome composition comprises liposomes having a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the liposome composition comprises liposomes containing the γ pantol according to any one of [1] to [11] of the detailed description section and having a diameter in the range of 80nm to 120 nm. In some embodiments, the liposome is a liposome composition according to any one of [12] to [67] of the detailed description section, and has a diameter in the range of 80nm to 120 nm. In other embodiments, the liposome composition comprises liposomes having a diameter in the range of 40nm to 70nm or any range therebetween. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section and has a diameter in the range of 40nm to 70 nm. In some embodiments, the liposome is a liposome composition according to any one of [12] to [67] of the detailed description section, and has a diameter in the range of 40nm to 70 nm.
The properties of liposomes are influenced by the properties of the lipids used to prepare the liposomes. A wide variety of lipids have been used to prepare liposomes. These include cationic lipids, anionic lipids and neutral lipids. In some embodiments, the liposomes comprising the gamma polyglutamated antifolate are anionic or neutral. In other embodiments, provided liposomes are cationic. The charge (e.g., anionic, neutral, or cationic) can be determined conventionally by measuring the zeta potential of the liposome. The zeta potential of the liposomes can be positive, zero, or negative. In some embodiments, the zeta potential of the liposome is less than or equal to zero. In some embodiments, the zeta potential of the liposome is in the range of 0 to-150 mV. In another embodiment, the zeta potential of the liposomes is in the range of-30 to-50 mV.
In some embodiments, cationic lipids are used to prepare cationic liposomes, which are typically used as gene transfection agents. The positive charge on the cationic liposome can interact with the negative charge on the cell surface. After the cationic liposome is bound to the cell, the liposome is transported within the cell by endocytosis.
In some preferred embodiments, neutral to anionic liposomes are used. In a preferred embodiment, anionic liposomes are used. The use of a mixture of, for example, neutral lipids such as HSPC and anionic lipids such as PEG-DSPE, allows the formation of anionic liposomes that are less likely to bind non-specifically to normal cells. Specific binding to tumor cells can be achieved by using tumor targeting antibodies, such as folate receptor antibodies, including, for example, folate receptor alpha antibodies, folate receptor beta antibodies, and/or folate receptor antibodies.
As an example, at least one (or some) lipid is an amphiphilic lipid, defined as having a hydrophilic portion and a hydrophobic portion (typically a hydrophilic head and a hydrophobic tail). The hydrophobic portion is generally oriented as the hydrophobic phase (e.g., within the bilayer), while the hydrophilic portion is generally oriented as the aqueous phase (e.g., outside the bilayer). The hydrophilic moiety may comprise polar or charged groups such as carbohydrates, phosphates, carboxylic acids, sulfates, amino groups, sulfhydryls, nitro groups, hydroxyl groups, and other similar groups. The hydrophobic moiety may comprise a non-polar group including, but not limited to, long chain saturated and unsaturated aliphatic hydrocarbon groups and groups substituted with one or more aromatic, alicyclic, or heterocyclic groups. Examples of amphiphilic compounds include, but are not limited to, phospholipids, amino lipids, and sphingolipids.
Typically, for example, the lipid is a phospholipid. Phospholipids include, but are not limited to, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidylinositol, phosphatidylserine, and the like. It will be appreciated that other lipid membrane components may be used, such as cholesterol, sphingomyelin and cardiolipin.
The lipids comprising the liposomes provided herein can be anionic and neutral (including zwitterionic and polar) lipids, including anionic and neutral phospholipids. At a selected pH, neutral lipids exist in the uncharged or neutral zwitterionic form. At physiological pH, such lipids include, for example, Dioleoylphosphatidylglycerol (DOPG), diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide, sphingomyelin, cephalin, cholesterol, cerebrosides, and diacylglycerol. Examples of zwitterionic lipids include, but are not limited to, Dioleoylphosphatidylcholine (DOPC), Dimyristoylphosphatidylcholine (DMPC), and Dioleoylphosphatidylserine (DOPS). Anionic lipids are negatively charged at physiological pH. These lipids include, but are not limited to, phosphatidylglycerol, cardiolipin, diacylphosphatidylserine, diacylphosphatidic acid, N-dodecanoylphosphatidylethanolamine, N-succinylphosphatidylethanolamine, N-glutarylphosphatidylethanolamine, lysylphosphatidylglycerol, palmitoyloleoyl-phosphatidylglycerol (POPG), and other anionic modifying groups attached to neutral lipids.
Collectively, anionic lipids and neutral lipids are referred to herein as non-cationic lipids. Such lipids may contain phosphorus, but are not limited thereto. Examples of non-cationic lipids include lecithin, lysolecithin, phosphatidylethanolamine, lysophosphatidylethanolamine, Dioleoylphosphatidylethanolamine (DOPE), Dipalmitoylphosphatidylethanolamine (DPPE), Dimyristoylphosphatidylethanolamine (DMPE), distearoylphosphatidyl1-ethanolamine (DSPE), palmitoylphosphatidylethanolamine (POPE), palmitoyl-oleoylphosphatidylcholine (POPC), Egg Phosphatidylcholine (EPC), Distearoylphosphatidylcholine (DSPC), Dioleoylphosphatidylcholine (DOPC), Dipalmitoylphosphatidylglycerol (DPPC), Dioleoylphosphatidylglycerol (DOPG), Dipalmitoylphosphatidylglycerol (DPPG), palmitoylphosphatidylglycerol (POPG), 16-0-methyl PE, 16-0-dimethyl PE, 18-1-trans PE, palmitoyl oleoyl-phosphatidylethanolamine (POPE), 1-stearoyl-2-oleoyl phosphatidylethanolamine (SOPE), phosphatidylserine, phosphatidylinositol, sphingomyelin, cephalin, cardiolipin, phosphatidic acid, cerebroside, dicetyl phosphate, and cholesterol.
Liposomes can be assembled using any liposome assembly method known in the art that uses a liposome component (also known as a liposome component). Liposomal components include, for example, lipids such as DSPE, HSPC, cholesterol, and derivatives of these components. Other suitable Lipids are commercially available, for example, from Avanti Polar Lipids, Inc (Alabaster, Alabama, USA). A partial list of useful negatively or neutrally charged lipids suitable for preparing anionic liposomes can be, for example, at least one of: DLPC, DMPC, DPPC, DSPC, DOPC, DMPE, DPPE, DOPE, DMPA. Na, DPPA. Na, DOPA. Na, DMPG. Na, DPPG. Na, DOPG. Na, DMPS. Na, DPPS. Na, DOPS. Na, DOPE-glutaryl (Na)2, tetramyristoyl cardiolipin (Na)2, DSPE-mPEG-2000. Na, DSPE-mPEG-5000. Na, and DSPE-maleimide PEG-2000. Na.
In some embodiments, the γ pantol compositions provided herein are formulated in liposomes comprising cationic lipids. In one embodiment, the cationic lipid is selected from, but is not limited to, the cationic lipids described in international application publication nos. WO2012/040184, WO2011/153120, WO2011/149733, WO2011/090965, WO2011/043913, WO2011/022460, WO2012/061259, WO2012/054365, WO2012/044638, WO2010/080724, WO2010/21865, and WO2008/103276, U.S. patent nos. 7,893,302, 7,404,969, and 8,283,333, and U.S. application publication nos. US20100036115 and US20120202871, each of which is incorporated herein by reference in its entirety. In another embodiment, the cationic lipid may be selected from, but is not limited to, formula a described in international application publication nos. WO2012/040184, WO2011/153120, WO201/1149733, WO2011/090965, WO2011/043913, WO2011/022460, WO2012/061259, WO2012/054365, and WO2012/044638, each of which is incorporated herein by reference in its entirety. In another embodiment, the cationic lipid may be selected from, but is not limited to, those of formula CLI-CLXXIX of international publication No. WO2008103276, CLI-CLXXIX of U.S. patent No. 7,893,302, CLI-clxxxii of U.S. patent No. 7,404,969, and formula I-VI of U.S. patent publication No. US 20100036115; each of which is incorporated herein by reference in its entirety. As a non-limiting example, the cationic lipid may be selected from (20Z,23Z) -N, N-dimethyl-nonacosane-20, 23-dien-10-amine, (17Z,20Z) -N, N-dimethyl-hexacosane-17, 20-dien-9-amine, (1Z,19Z) -N5N-dimethyl pentacosane-16, 19-dien-8-amine, (13Z,16Z) -N, N-dimethyl docosane-13, 16-dien-5-amine, (12Z,15Z) -N, N-dimethyl heneicosane-12, 15-dien-4-amine, (14Z,17Z) -N, N-dimethyl-tricosane-14, 17-dien-6-amine, (15Z,18Z) -N, N-dimethylditetradec-15, 18-dien-7-amine, (18Z,21Z) -N, N-dimethylheptacosan-18, 21-dien-10-amine, (15Z,18Z) -N, N-dimethylditetradec-15, 18-dien-5-amine, (14Z,17Z) -N, N-dimethyl-tricosane-14, 17-dien-4-amine, (19Z,22Z) -N, N-dimethyldioctadecyl-19, 22-dien-9-amine, (18Z,21Z) -N, N-dimethylheptacosa-18, 21-dien-8-amine, (17Z,20Z) -N, N-dimethylhexacosan-17, 20-dien-7-amine, (16Z,19Z) -N, N-dimethylpentacosan-16, 19-dien-6-amine, (22Z,25Z) -N, N-dimethylhentriacontan-22, 25-dien-10-amine, (21Z,24Z) -N, N-dimethyl-triaconta-21, 24-dien-9-amine, (18Z) -N, N-dimethylheptacosan-18-en-10-amine, (17Z) -N, N-dimethylhexacosan-17-en-9-amine, (19Z,22Z) -N, N-dimethyldioctadecyl-19, 22-dien-7-amine, N-dimethylheptacosan-10-amine, (20Z,23Z) -N-ethyl-N-methyl-nonacosan-20, 23-dien-10-amine, 1- [ (11Z,14Z) -1-nonyleicosa-11, 14-dien-1-yl ] pyrrolidine, (20Z) -N, N-dimethyl-heptacosan-20-en-10-amine, (15Z) -N, N-dimethylheptacosan-15-en-10-amine, (14Z) -N, N-dimethylnonacosan-14-en-10-amine, (17Z) -N, N-dimethylnonacosan-17-en-10-amine, N-dimethylnonacosan-10-amine, N-dimethylheptacosan-10-amine, N-dimethyloctacosan-1, (24Z) -N, N-dimethyltridec-24-en-10-ylamine, (20Z) -N, N-dimethyl-nonacosan-20-en-10-ylamine, (22Z) -N, N-dimethylhentriacont-22-en-10-ylamine, (16Z) -N, N-dimethylpentacosan-16-en-8-ylamine, (12Z,15Z) -N, N-dimethyl-2-nonylheneicosyl-12, 15-dien-1-ylamine, (13Z,16Z) -N, N-dimethyl-3-nonyldidodeca-13, 16-dien-1-ylamine, N-dimethyl-1- [ (1S,2R) -2-octylcyclopropyl ] heptadec-8-amine, 1- [ (1S,2R) -2-hexylcyclopropyl ] -N, N-dimethylnona-10-amine, N-dimethyl-1- [ (1S,2R) -2-octylcyclopropyl ] nonadeca-10-amine, N-dimethyl-21- [ R1S,2R) -2-octylcyclopropyl ] heneico-10-amine, N-dimethyl-1- [ (1S,2S) -2- { [ (1R,2R) -2-pentylcyclopropyl ] methyl } cyclopropyl ] nona-10-amine, N-dimethyl-1- [ (1S,2R) -2-octylcyclopropyl ] hexadec-8-amine, N-dimethyl- [ (1R,2S) -2-undecyl-cyclopropyl ] tetradec-5-amine, N-dimethyl-3- {7- [ (1S,2R) -2-octylcyclopropyl ] heptyl } dodeca-1-amine, 1- [ (1R,2S) -2-heptylcyclopropyl ] -N, N-dimethyloctadeca-9-amine, 1- [ (1S,2R) -2-decylcyclopropyl ] -N, N-dimethylpentadec-6-amine, N-dimethyl-1- [ (1S,2R) -2-octylcyclopropyl ] pentadecan-8-amine, N-dimethylcy-1-ylpropyl ] -N, N-dimethylpentadecan-6-amine, N-dimethylcy-1- [ (1S,2R) -2-octylcyclopropyl, R-N, N-dimethyl-1- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] -3- (octyloxy) propan-2-amine, S-N, N-dimethyl-1- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] -3- (octyloxy) propan-2-amine, 1- {2- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] -1- [ (octyloxy) methyl ] ethyl } pyrrolidine, (2S) -N, N-dimethyl-1- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] -3- [ (5Z) -oct-5-en-1-yloxy ] propan-2-amine, 1- {2- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] -1- [ (octyloxy) methyl ] ethyl } azetidine, (2S) -1- (hexyloxy) -N, N-dimethyl-3- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] propan-2-amine, (2S) -1- (heptyloxy) -N, N-dimethyl-3- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] propan-2-amine, N-dimethyl-1- (nonyloxy) -3- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] propan-2-amine, N-dimethyl-1- [ (9Z) -octadec-9-en-1-yloxy ] -3- (octyloxy) propan-2-amine; (2S) -N, N-dimethyl-1- [ (6Z,9Z,12Z) -octadeca-6, 9, 12-trien-1-yloxy ] -3- (octyloxy) propan-2-amine, (2S) -1- [ (11Z,14Z) -eicosa-11, 14-dien-1-yloxy ] -N, N-dimethyl-3- (pentyloxy) propan-2-amine, (2S) -1- (hexyloxy) -3- [ (11Z,14Z) -eicosa-11, 14-dien-1-yloxy ] -N, N-dimethylpropan-2-amine, 1- [ (11Z,14Z) -eicosa-11, 14-dien-1-yloxy ] -N, N-dimethyl-3- (octyloxy) propan-2-amine, 1- [ (13Z,16Z) -docosan-13, 16-dien-1-yloxy ] -N, N-dimethyl-3- (octyloxy) propan-2-amine, (2S) -1- [ (13Z,16Z) -docosan-13, 16-dien-1-yloxy ] -3- (hexyloxy) -N, N-dimethyl-propan-2-amine, (2S) -1- [ (13Z) -docosan-13-en-1-yloxy ] -3- (hexyloxy) -N, n-dimethyl-propan-2-amine, 1- [ (13Z) -docosan-13-en-1-yloxy ] -N, N-dimethyl-3- (octyloxy) propan-2-amine, 1- [ (9Z) -hex-9-en-1-yloxy ] -N, N-dimethyl-3- (octyloxy) propan-2-amine, (2R) -N, N-dimethyl-H (1-formyloctyl) oxy ] -3- [ (9Z,12Z) -octadec-9, 12-dien-1-yloxy ] propan-2-amine, (2R) -1- [ (3, 7-dimethyloctyl) oxy ] -N, N-dimethyl-3-R9Z, 12Z) -octadeca-9, 12-dien-1-yloxy ] propan-2-amine, N-dimethyl-1- (octyloxy) -3- ({8- [ (1S,2S) -2- { [ (1R,2R) -2-pentylcyclopropyl ] methyl } cyclopropyl ] octyl } oxy) propan-2-amine, N-dimethyl-1- { [ - (2-octylcyclopropyl) octyl ] oxy } -3- (octyloxy) propan-2-amine and (11E,20Z,23Z) -N, N-dimethyltwenty-nine-11, 20, 2-trien-10-amine or a pharmaceutically acceptable salt or stereoisomer thereof.
In one embodiment, the lipid may be a cleavable lipid, such as those described in international publication No. WO2012/170889, which is incorporated herein by reference in its entirety.
Cationic lipids can be conventionally synthesized using methods known in the art and/or as described in international publication nos. WO2012/040184, WO2011/153120, WO2011/149733, WO2011/090965, WO201/1043913, WO2011/022460, WO2012/061259, WO2012/054365, WO2012/044638, WO2010/080724, and WO2010/21865, each of which is incorporated herein by reference in its entirety.
The lipid derivative may comprise, for example, at least a bonding (preferably covalent bonding) of one or more steric stabilizers and/or functional groups to the liposomal composition, after which the steric stabilizers and/or functional groups should be considered as part of the liposomal composition. Functional groups include groups that can be used to attach a liposome component to another moiety, such as a protein. Such functional groups include at least maleimide. These steric stabilizers include at least one from the group consisting of: polyethylene glycol (PEG); poly-L-lysine (PLL); monosialoganglioside (GM 1); poly (vinyl pyrrolidone) (PVP); poly (acrylamide) (PAA); poly (2-methyl-2-oxazoline); poly (2-ethyl-2-oxazoline); a phosphatidylpolyglycerol; poly [ N- (2-hydroxy-propyl) methacrylamide ]; amphiphilic poly-N-vinylpyrrolidone; an L-amino acid based polymer; and polyvinyl alcohol.
In some embodiments, the γ pantol composition is formulated in a lipid-polycation complex. Formation of the lipid-polycation complex may be accomplished using methods known in the art and/or as described in U.S. publication No. 20120178702, which is incorporated herein by reference in its entirety. As non-limiting examples, the polycation may include cationic peptides or polypeptides such as, but not limited to, polylysine, polyornithine, and/or polyarginine and cationic peptides described in international publication No. WO 2012/013326; the publication is incorporated by reference herein in its entirety. In another embodiment, the γ pantol is formulated in a lipid-polycation complex further comprising a neutral lipid, such as but not limited to cholesterol or dioleoyl phosphatidylethanolamine (DOPE).
As the components of the liposomes can include any molecule (i.e., chemical/agent/protein) bound thereto, in some embodiments, the components of the provided liposomes include at least a member selected from the group consisting of: DSPE, DSPE-PEG, DSPE-maleimide, HSPC; HSPC-PEG; HSPC-maleimide; cholesterol; cholesterol-PEG; and cholesterol-maleimide. In some embodiments, provided components of liposomes include DSPE, DSPE-PEG, DSPE-maleimide, HSPC; HSPC-PEG; HSPC maleimides; cholesterol; cholesterol-PEG; and cholesterol-maleimide. In a preferred embodiment, the liposome components making up the liposomes comprise DSPE; DSPE-FITC; DSPE-maleimide; cholesterol; and HSPC.
In further embodiments, the liposomes of the liposome compositions provided herein comprise an oxidized phospholipid. In some embodiments, the liposome comprises an oxidized phospholipid selected from the group consisting of: phosphatidylserine, phosphatidylinositol, phosphatidylethanolamine, phosphatidylcholine, and 1-palmitoyl-2-arachidonoyl-sn-glycero-2-phosphate. In some embodiments, the phospholipid has an unsaturated bond. In some embodiments, the phospholipid is an arachidonic acid-containing phospholipid. In further embodiments, the phospholipid is sn-2-oxygenated. In other embodiments, the phospholipid is not fragmented.
In some embodiments, the liposomes of the disclosed liposome compositions comprise oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (oxpac). As used herein, the term "oxPAPC" refers to lipids produced by oxidation of 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine (PAPC) resulting in a mixture of oxidized phospholipids containing fragmented or full length oxygenated sn-2 residues. Well characterized oxidatively fragmented species contain five carbon sn-2 residues carrying either an omega-aldehyde or an omega-carboxyl group. Oxidation of arachidonic acid residues also produces phospholipids containing esterified isoprostane. In some embodiments, the oxPAPC includes HOdia-PC, KOdia-PC, HOOA-PC, and KOOA-PC species in addition to other oxidation products present in the oxPAPC. In other embodiments, the oxPAPC is an epoprostane-containing phospholipid. In other embodiments, the oxPAPC is 1-palmitoyl-2- (5, 6-epoxyisoprostane E2) -sn-glycero-3-phosphocholine (5,6-PEIPC), 1-palmitoyl-2- (epoxy-cyclopentenone) -sn-glycero-3-phosphorylcholine (PECPC) and/or 1-palmitoyl-2- (epoxy-isoprostane E2) -sn-glycero-4-phosphocholine (PEIPC). In some embodiments, the phospholipid has an unsaturated bond. In some embodiments, the phospholipid is an arachidonic acid-containing phospholipid. In further embodiments, the phospholipid is sn-2-oxygenated. In other embodiments, the phospholipid is not fragmented.
In some embodiments, the liposomal gamma-polyglutamated antifolate composition is pegylated (i.e., pegylated liposomal gamma-polyglutamated (e.g., pentaglutamated or hexaglutamated) antifolate (PLp-gamma pantol or TPLp-gamma pantol).
In some embodiments, the liposomes of the disclosed liposome compositions comprise a lipid selected from the group consisting of: 1-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine (PGPC); 1-palmitoyl-2- (9' oxo-nonanoyl) -sn-glycero-3-phosphocholine; 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine; 1-palmitoyl-2-myristoyl-sn-glycero-3-phosphocholine; 1-palmitoyl-2-hexadecyl-sn-glycero-3-phosphocholine; 1-palmitoyl-2-nonanedioyl-sn-glycero-3-phosphocholine; and 1-palmitoyl-2-acetyl-sn-glycero-3-phosphocholine. In other embodiments, the liposome comprises PGPC.
In some embodiments, the pH of the solution comprising the liposome composition is pH 2 to 8, or any range therebetween. In some embodiments, the pH of the solution comprising the liposome composition is pH 5 to 8 or pH 2 to 6, or any range therebetween. In some embodiments, the pH of the solution comprising the liposome composition is pH 5 to 8, or any range therebetween. In some embodiments, the pH of the solution comprising the liposome composition is pH 6 to 7, or any range therebetween. In some embodiments, the pH of the solution comprising the liposome composition is 6 to 7.5, 6.5 to 7.5, 6.7 to 7.5, or 6.3 to 7.0, or any range therebetween.
In some embodiments, at least one component of the liposomal lipid bilayer is functionalized (or reactive). As used herein, a functionalized component is a component that comprises reactive groups that can be used to crosslink reagents and moieties to lipids. If the lipid is functionalized, any liposomes it forms are also functionalized. In some embodiments, the reactive group is a group that will react with the crosslinker (or other moiety) to form a crosslink. The reactive group in the liposomal lipid bilayer is located anywhere on the lipid that allows it to contact the cross-linking agent and cross-link with another moiety (e.g., a steric stabilizer or targeting moiety). In some embodiments, the reactive group is in the head group of the lipid, including, for example, phospholipids. In some embodiments, the reactive group is a maleimide group. The maleimide groups may be crosslinked to each other in the presence of dithiol crosslinkers, including but not limited to Dithiothreitol (DTT).
It is to be understood that the use of other functionalized lipids, other reactive groups, and other cross-linking agents than those described above are further contemplated. In addition to maleimide groups, other examples of reactive groups contemplated include, but are not limited to, other thiol-reactive groups, amino groups such as primary and secondary amines, carboxyl groups, hydroxyl groups, aldehyde groups, alkynyl groups, azido groups, carbonyl groups, haloacetyl groups (e.g., iodoacetyl groups), imidate groups, N-hydroxysuccinimide esters, sulfhydryl groups, and pyridyl disulfide groups.
Functionalized and non-functionalized Lipids are available from a number of commercial sources, including Avanti Polar Lipids (Alabaster, AL) and Lipoid LLC (Newark, NJ).
(2) Liposome interior space
In other non-limiting embodiments, provided liposomes enclose an interior space. In some embodiments, the interior space includes, but is not limited to, an aqueous solution. In some embodiments, the interior space comprises a gamma polyglutamated antifolate agent as provided herein. In further embodiments, the interior space of the liposome comprises a tonicity agent. In some embodiments. In some embodiments, the concentration (weight percent) of the tonicity agent is 0.1% to 20%, 1% to 20%, 0.5% to 15%, 1% to 15%, or 1% to 50%, or any range therebetween. In some embodiments, the interior space of the liposome comprises a sugar (e.g., trehalose, maltose, sucrose, lactose, mannose, mannitol, glycerol, dextrose, fructose, etc.). In other embodiments, the concentration (weight percent) of the sugar is 0.1% -20%, 1% -20%, 0.5% -15%, 1% -15%, or 1% -50%, or any range therebetween. In some embodiments, the pH of the liposome interior space is pH 2 to 8, or any range therebetween. In some embodiments, the pH of the solution comprising the liposome composition is pH 5 to 8, or any range therebetween. In some embodiments, the pH of the solution comprising the liposome composition is pH 6 to 7, or any range therebetween. In some embodiments, the pH of the solution comprising the liposome composition is 6 to 7.5, 6.5 to 7.5, 6.7 to 7.5, or 6.3 to 7.0, or any range therebetween. In some embodiments, the interior space comprises a buffer. In other embodiments, the buffer is a buffer selected from HEPES, citrate, or sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the buffer is HEPES. In some embodiments, the buffer is citrate. In some embodiments, the buffer is sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the concentration of the buffer is 15 to 200mM, or any range therebetween. In other embodiments, the buffer has a concentration between 5 to 200mM, 15 to 200, between 5 to 100mM, between 15 to 100mM, between 5 to 50mM, between 15 to 50mM, between 5 to 25mM, between 5 to 20mM, between 5 to 15mM, or any range therebetween. In some embodiments, the buffer is HEPES at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is citrate at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is sodium phosphate at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration between 5mM to 500mM, or 50mM to 500mM, or any range therebetween.
In some embodiments, the interior space of the liposome comprises glutamine, glutamate, and/or polyglutamate (e.g., diglutamate, triglutamate, tetraglutamate, and/or pentaglutamate containing one or more gamma glutamyl linkages or 1 or more alpha glutamyl linkages). In other embodiments, the concentration of glutamine, glutamate, and/or polyglutamate is 0.1% -20%, 1% -20%, 0.5% -15%, 1% -15%, 5% -20%, or 1% -50% by weight, or any range therebetween. In some embodiments, the interior space of the liposome comprises glutamine. In some embodiments, the interior space of the liposome comprises glutamate. In some embodiments, the interior space of the liposome comprises polyglutamate. In some embodiments, the concentration (weight percent) of glutamine, glutamate, and/or polyglutamate is 1% to 15% or any range therebetween. In another embodiment, the glutamine, glutamate, and/or polyglutamate is present at about 5% to 20% by weight of glutamine, glutamate, and/or polyglutamate, or any combination of one or more lyoprotectants or cryoprotectants, at a total concentration of 5% to 20%. In some embodiments, the interior space comprises a buffer. In other embodiments, the buffer is a HEPES buffer or a citrate buffer. In other embodiments, the concentration of citrate buffer is between 5 and 200 mM. In some embodiments, the pH of the interior space is between 2.8 and 6. In some embodiments, the pH of the solution comprising the liposome composition is 6 to 7.5, 6.5 to 7.5, 6.7 to 7.5, or 6.3 to 7.0, or any range therebetween. In some embodiments, the interior space comprises a buffer. In some embodiments, the buffer is a buffer selected from HEPES, citrate, or sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the buffer is HEPES. In some embodiments, the buffer is citrate. In some embodiments, the buffer is sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the concentration of the buffer is 15 to 200mM, or any range therebetween. In other embodiments, the buffer has a concentration between 5 to 200mM, 15 to 200, between 5 to 100mM, between 15 to 100mM, between 5 to 50mM, between 15 to 50mM, between 5 to 25mM, between 5 to 20mM, between 5 to 15mM, or any range therebetween. In some embodiments, the buffer is HEPES at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is citrate at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is sodium phosphate at a concentration of 15 to 200mM or any range therebetween. In further embodiments, the interior space of the liposome comprises sodium acetate and/or calcium acetate. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration between 5mM to 500mM, or 50mM to 500mM, or any range therebetween. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration of between 50mM to 500 mM.
In some embodiments, the interior space of the liposome comprises glutamine. In other embodiments, the concentration of glutamine is 0.1% -20%, 1% -20%, 0.5% -15%, 1% -15%, 5% -20%, or 1% -50% by weight, or any range therebetween. In some embodiments, the concentration (weight percent) of glutamine is 1% -15% or any range therebetween. In another embodiment, glutamine is present at about 5% to 20% by weight of glutamine or one or more lyoprotectants or any combination of cryoprotectants, at a total concentration of 5% to 20%. In some embodiments, the interior space comprises a buffer. In other embodiments, the buffer is a HEPES buffer or a citrate buffer. In other embodiments, the concentration of citrate buffer is between 5 and 200 mM. In some embodiments, the pH of the interior space is between 2.8 and 6. In some embodiments, the pH of the solution comprising the liposome composition is 6 to 7.5, 6.5 to 7.5, 6.7 to 7.5, or 6.3 to 7.0, or any range therebetween. In some embodiments, the interior space comprises a buffer. In some embodiments, the buffer is a buffer selected from HEPES, citrate, or sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the buffer is HEPES. In some embodiments, the buffer is citrate. In some embodiments, the buffer is sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the concentration of the buffer is 15 to 200mM, or any range therebetween. In other embodiments, the buffer has a concentration between 5 to 200mM, 15 to 200, between 5 to 100mM, between 15 to 100mM, between 5 to 50mM, between 15 to 50mM, between 5 to 25mM, between 5 to 20mM, between 5 to 15mM, or any range therebetween. In some embodiments, the buffer is HEPES at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is citrate at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is sodium phosphate at a concentration of 15 to 200mM or any range therebetween. In further embodiments, the interior space of the liposome comprises sodium acetate and/or calcium acetate. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration between 5mM to 500mM, or 50mM to 500mM, or any range therebetween. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration of between 50mM to 500 mM.
In some embodiments, the interior space of the liposome comprises trehalose. In other embodiments, the concentration of trehalose is 0.1% -20%, 1% -20%, 0.5% -15%, 1% -15%, 5% -20%, or 1% -50% by weight, or any range therebetween. In other embodiments, the concentration (weight percent) of trehalose is 1% -15% or any range therebetween. In another embodiment, trehalose is present at about 5% to 20% weight percent of trehalose or any combination of one or more lyoprotectants or cryoprotectants, at a total concentration of 5% to 20%. In some embodiments, the interior space comprises a buffer. In other embodiments, the buffer is a HEPES buffer or a citrate buffer. In other embodiments, the concentration of citrate buffer is between 5 and 200 mM. In some embodiments, the pH of the interior space is between 2.8 and 6. In some embodiments, the pH of the solution comprising the liposome composition is 6 to 7.5, 6.5 to 7.5, 6.7 to 7.5, or 6.3 to 7.0, or any range therebetween. In some embodiments, the interior space comprises a buffer. In some embodiments, the buffer is a buffer selected from HEPES, citrate, or sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the buffer is HEPES. In some embodiments, the buffer is citrate. In some embodiments, the buffer is sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the concentration of the buffer is 15 to 200mM, or any range therebetween. In other embodiments, the buffer has a concentration between 5 to 200mM, 15 to 200, between 5 to 100mM, between 15 to 100mM, between 5 to 50mM, between 15 to 50mM, between 5 to 25mM, between 5 to 20mM, between 5 to 15mM, or any range therebetween. In some embodiments, the buffer is HEPES at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is citrate at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is sodium phosphate at a concentration of 15 to 200mM or any range therebetween. In further embodiments, the interior space of the liposome comprises sodium acetate and/or calcium acetate. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration between 5mM to 500mM, or 50mM to 500mM, or any range therebetween. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration of between 50mM to 500 mM.
In some embodiments, the interior space of the liposome comprises dextrose. In other embodiments, the concentration of dextrose is 0.1% -20%, 1% -20%, 0.5% -15%, 1% -15%, 5% -20%, or 1% -50% by weight, or any range therebetween. In other embodiments, the concentration (weight percent) of dextrose is 1% -15% or any range therebetween. In another embodiment, dextrose is present at about 5% to 20% weight percent of dextrose or one or more lyoprotectants or any combination of cryoprotectants, for a total concentration of 5% to 20%. In some embodiments, the pH of the solution comprising the liposome composition is 6 to 7.5, 6.5 to 7.5, 6.7 to 7.5, or 6.3 to 7.0, or any range therebetween. In some embodiments, the interior space comprises a buffer. In some embodiments, the buffer is a buffer selected from HEPES, citrate, or sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the buffer is HEPES. In some embodiments, the buffer is citrate. In some embodiments, the buffer is sodium phosphate (e.g., sodium dihydrogen phosphate and/or disodium hydrogen phosphate). In some embodiments, the concentration of the buffer is 15 to 200mM, or any range therebetween. In other embodiments, the buffer has a concentration between 5 to 200mM, 15 to 200, between 5 to 100mM, between 15 to 100mM, between 5 to 50mM, between 15 to 50mM, between 5 to 25mM, between 5 to 20mM, between 5 to 15mM, or any range therebetween. In some embodiments, the buffer is HEPES at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is citrate at a concentration of 15 to 200mM or any range therebetween. In some embodiments, the buffer is sodium phosphate at a concentration of 15 to 200mM or any range therebetween. In further embodiments, the interior space of the liposome comprises sodium acetate and/or calcium acetate. In some embodiments, the interior space of the liposome comprises sodium acetate and calcium acetate at a total concentration between 5mM to 500mM, or 50mM to 500mM, or any range therebetween.
In additional embodiments, the present disclosure provides liposome compositions comprising liposomes encapsulating (i.e., filled with) a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein). In some embodiments, the liposome composition comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome comprises a polyglutamated antifolate described in section I. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the liposomes in the liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups (including glutamyl groups of antifolates). In some embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises two or more glutamyl groups in the L form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in D form. In other embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises a glutamyl group in the D form and two or more glutamyl groups in the L form. In further embodiments, the gamma polyglutamated antifolate agent in said Lp-gamma pantol comprises two or more glutamyl groups having a gamma carboxy linkage. In some embodiments, the liposome composition comprises liposomes comprising a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises an L-gamma-pentaglutaminated antifolate, a D-gamma-pentaglutaminated antifolate, or an L-and D-gamma-pentaglutaminated antifolate. In some embodiments, the liposome composition comprises liposomes comprising a gamma hexaglutaminated antifolate (Lp-gamma pantol). In other embodiments, the liposome comprises an L-gamma-hexaglutaminated antifolate, a D-gamma-hexaglutaminated antifolate, or an L-and D-gamma-hexaglutaminated antifolate.
In some embodiments, the present disclosure provides a liposome composition comprising targeted and pegylated liposomes comprising a gamma polyglutamated antifolate (TPLp-gamma pantol). In some embodiments, the liposome composition comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposomal composition comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the liposome composition is a targeted pegylated liposome composition according to any one of [50] to [69] of the specific embodiments. In some embodiments, the targeted pegylated liposomal gamma polyglutamated (e.g., pentaglutamated or hexaglutamated) antifolate comprises a medium comprising liposomes comprising an interior space; a gamma polyglutamated antifolate aqueous solution disposed within the interior space; and a targeting moiety comprising a protein having specific affinity for at least one folate receptor, and wherein the targeting moiety is disposed on the exterior of the liposome. In some embodiments, the medium is an aqueous solution. In some embodiments, the interior space, the exterior space (e.g., medium), or both the interior space and the medium contain one or more of the above-listed lyoprotectants or cryoprotectants. In some embodiments, the cryoprotectant is mannitol, trehalose, sorbitol, or sucrose.
In some embodiments, the liposomes encapsulating the gamma polyglutamated antifolate (e.g., Lp-gamma pantol, including PLp-gamma pantol, TPLp-gamma pantol, TLp-gamma pantol and NTLp-gamma pantol) have an interior space containing less than 500,000 or less than 200,000 gamma polyglutamated antifolate molecules. In some embodiments, the liposome interior space contains between 10 to 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposome interior space contains between 10,000 to 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are non-pegylated and have an interior space containing less than 500,000 or less than 200,000 gamma polyglutamated antifolate molecules. In some embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10 and 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10,000 to 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes comprise a targeting moiety, are non-pegylated (TLp- γ pantol), and have an interior space containing less than 500,000 or less than 200,000 γ polyglutamated antifolate molecules. In some embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10 and 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10,000 and 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are free of targeting moieties, are non-pegylated (NTLp- γ panantifol), and have an interior space containing less than 500,000 or less than 200,000 gamma polyglutamated antifolate molecules. In some embodiments, the liposomes are free of a targeting moiety, are non-pegylated, and the interior space of the liposomes contains between 10 and 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposomes are free of targeting moieties, are non-pegylated, and the interior space of the liposomes contains between 10,000 and 100,000 gamma polyglutamated antifolate molecules, or any range therebetween.
In some embodiments, the liposome encapsulates a gamma polyglutamated antifolate containing 2-10 glutamyl groups (i.e., Lp-gamma pantol, including PLp-gamma pantol, TPLp-gamma pantol, TLp-gamma pantol and NTLp-gamma pantol), and has an interior space containing less than 500,000 or less than 200,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups. In some embodiments, the liposome interior space contains between 10 and 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween. In other embodiments, the liposome interior space contains between 10,000 and 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween. In some embodiments, the liposomes are non-pegylated and have an interior space containing less than 500,000 or less than 200,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups. In some embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10 and 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween. In other embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10,000 and 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween. In some embodiments, the liposomes comprise a targeting moiety, are non-pegylated (TLp- γ pantol), and have an interior space containing less than 500,000 or less than 200,000 γ polyglutamated antifolate molecules containing 2-10 glutamyl groups. In some embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10 and 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween. In other embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10,000 and 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween. In some embodiments, the liposomes are non-targeted and non-pegylated (NTLp- γ panantifol) and have an interior space containing less than 500,000 or less than 200,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups. In some embodiments, the liposomes are free of targeting moieties, are non-pegylated, and the interior space of the liposomes contains between 10 and 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween. In other embodiments, the liposomes are non-targeted and non-pegylated, and the internal space of the liposomes contains between 10,000 to 100,000 gamma polyglutamated antifolate molecules containing 2-10 glutamyl groups, or any range therebetween.
In some embodiments, the liposome encapsulates a gamma-tetraglutamated antifolate (i.e., Lp-gamma pantol, including PLp-gamma pantol, TPLp-gamma pantol, TLp-gamma pantol and NTLp-gamma pantol) and has an interior space containing less than 500,000 or less than 200,000 gamma-tetraglutamated antifolate molecules. In some embodiments, the liposome interior space contains between 10 to 100,000 gamma-tetraglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposome interior space contains between 10,000 to 100,000 gamma-tetraglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are non-pegylated and have an interior space containing less than 500,000 or less than 200,000 gamma tetraglutamated antifolate molecules. In some embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10 and 100,000 gamma tetraglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10,000 to 100,000 gamma tetraglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes comprise a targeting moiety, are non-pegylated (TLp- γ pantol), and have an interior space containing less than 500,000 or less than 200,000 γ tetraglutamated antifolate molecules. In some embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10 and 100,000 gamma-tetraglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10,000 and 100,000 gamma tetraglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are free of a targeting moiety, are non-pegylated (NTLp- γ panantifol), and have an interior space containing less than 500,000 or less than 200,000 γ tetraglutamated antifolate molecules. In some embodiments, the liposomes are free of a targeting moiety, are non-pegylated, and the interior space of the liposomes contains between 10 and 100,000 gamma tetraglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposomes are free of targeting moieties, are non-pegylated, and the interior space of the liposomes contains between 10,000 and 100,000 gamma tetraglutamated antifolate molecules, or any range therebetween.
In some embodiments, the liposome encapsulates a gamma pentaglutamated antifolate (e.g., Lp-gamma pantol, including PLp-gamma pantol, TPLp-gamma pantol, TLp-gamma pantol and NTLp-gamma pantol) and has an internal space containing less than 500,000 or less than 200,000 gamma pentaglutamated antifolate molecules. In some embodiments, the liposome interior space contains between 10 to 100,000 gamma pentaglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposome interior space contains between 10,000 to 100,000 gamma pentaglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are non-pegylated and have an interior space containing less than 500,000 or less than 200,000 gamma pentaglutaminated antifolate molecules. In some embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10 and 100,000 gamma pentaglutaminated antifolate molecules, or any range therebetween. In other embodiments, the liposome is non-pegylated, and the interior space of the liposome contains between 10,000 and 100,000 gamma pentaglutaminated antifolate molecules, or any range therebetween. In some embodiments, the liposomes comprise a targeting moiety, are non-pegylated (TLp-gamma PANTIFOL), and have an interior space containing less than 500,000 or less than 200,000 gamma pentaglutamated antifolate molecules. In some embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10 and 100,000 gamma pentaglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10,000 and 100,000 gamma pentaglutamated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are non-targeted and non-pegylated (NTLp- γ panantifol) and have an interior space containing less than 500,000 or less than 200,000 γ pentaglutamated antifolate molecules. In some embodiments, the liposomes are free of targeting moieties and are non-pegylated, and the interior space of the liposomes contains between 10 and 100,000 gamma polyglutamated antifolate molecules, or any range therebetween. In other embodiments, the liposomes are non-targeted and non-pegylated, and the internal space of the liposomes contains between 10,000 to 100,000 gamma pentaglutamated antifolate molecules, or any range therebetween.
In some embodiments, the liposome encapsulates a gamma hexaglutaminated antifolate (i.e., Lp-gamma pantol, including PLp-gamma pantol, TPLp-gamma pantol, TLp-gamma pantol and NTLp-gamma pantol) and has an internal space containing less than 500,000 or less than 200,000 gamma hexaglutaminated antifolate molecules. In some embodiments, the liposome interior space contains between 10 to 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween. In other embodiments, the liposome interior space contains between 10,000 to 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are non-pegylated and have an interior space containing less than 500,000 or less than 200,000 gamma hexaglutaminated antifolate molecules. In some embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10 and 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween. In other embodiments, the liposomes are non-pegylated and the internal space of the liposomes contains between 10,000 to 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween. In some embodiments, the liposomes comprise a targeting moiety, are non-pegylated (TLp- γ pantol), and have an interior space containing less than 500,000 or less than 200,000 γ hexaglutaminated antifolate molecules. In some embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10 and 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween. In other embodiments, the liposome comprises a targeting moiety, is non-pegylated, and the interior space of the liposome contains between 10,000 and 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween. In some embodiments, the liposomes are non-targeted and non-pegylated (NTLp- γ panantifol) and have an interior space containing less than 500,000 or less than 200,000 γ hexaglutaminated antifolate molecules. In some embodiments, the liposomes are free of a targeting moiety, are non-pegylated, and the interior space of the liposomes contains between 10 and 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween. In other embodiments, the liposomes are free of a targeting moiety, are non-pegylated, and the interior space of the liposomes contains between 10,000 and 100,000 gamma hexaglutaminated antifolate molecules, or any range therebetween.
In some embodiments, the present disclosure provides a liposomal gamma-polyglutamated antifolate composition, wherein the liposomes encapsulate a gamma-polyglutamated antifolate or a salt or acid thereof, and one or more pharmaceutically acceptable aqueous carriers. In some embodiments, the liposome interior space contains trehalose. In some embodiments, the liposome interior space contains 5% to 20% trehalose by weight. In some embodiments, the liposome interior space contains HBS at a concentration of between 1 and 200mM and a pH of between 2 and 8. In some embodiments, the liposome interior space has a pH of 5-8, or any range therebetween. In some embodiments, the liposome interior space has a pH of 6-7, or any range therebetween. In some embodiments, the liposome interior space has a total concentration of sodium acetate and calcium acetate between 50mM to 500mM or any range therebetween.
A non-polyglutamated antifolate agent
In some embodiments, the liposomal gamma-polyglutamated antifolate (i.e., Lp-gamma pantol, including PLp-gamma pantol, TPLp-gamma pantol, TLp-gamma pantol and NTLp-gamma pantol) composition comprises a gamma-polyglutamated antifolate (e.g., gamma pantol as disclosed herein) and one or more non-polyglutamated polyglutamable antifolate compositions.
In some embodiments, the Lp- γ pantol (e.g., PLp- γ pantol, TPLp- γ pantol, TLp- γ pantol and NTLp- γ pantol) comprises a γ -polyglutamated antifolate (e.g., γ pantol disclosed herein) and an antifolate (anti). In some embodiments, the Lp- γ pantol (i.e., liposomal γ polyglutamated antifolate) comprises a γ polyglutamated antifolate and a polyglutamated antifolate selected from the group consisting of: methotrexate (MTX), Pemetrexed (PMX), Lometrexol (LMX), Raltitrexed (RTX), pralatrexate, AG2034, GW1843, aminopterin and LY 309887. In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and lometrexol. In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and pemetrexed. In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and folinic acid. In some embodiments, the Lp- γ pantol comprises a γ polyglutamated antifolate and a triazine antifolate derivative (e.g., a sulfonyl fluorotriazine, such as NSC 127755). In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and a serine hydroxymethyltransferase (SHMT2) inhibitor. In some embodiments, the SHMT2 inhibitor is an antifolate (e.g., a polyglutamated or non-polyglutamated antifolate). In some embodiments, the SHMT2 inhibitor is an antifolate.
B non-polyglutamated antifolate agent
In some embodiments, the Lp- γ pantol (e.g., PLp- γ pantol, TPLp- γ pantol, TLp- γ pantol and NTLp- γ pantol) comprises a γ -polyglutamated antifolate (e.g., γ pantol as disclosed herein) and a so-called "non-polyglutamated" antifolate. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome comprises a polyglutamated antifolate described in section I. In some embodiments, the liposomes comprise a gamma polyglutamated antifolate and a non-polyglutamated antifolate that inhibits one or more enzymes in the folate cycle metabolic pathway. In other embodiments, the non-polyglutamated antifolate inhibits one or more enzymes selected from the group consisting of: thymidylate Synthase (TS), dihydrofolate reductase (DHFR), glycinamide ribonucleotide (GAR) converting enzyme, and aminoimidazole carboxamide ribonucleotide (AICAR) converting enzyme. In some embodiments, the liposomes comprise a gamma polyglutamated antifolate and a non-polyglutamated antifolate that inhibits DHFR. In some embodiments, the liposomes comprise a gamma polyglutamated antifolate and a non-polyglutamated antifolate that inhibits TS. In some embodiments, the liposome comprises a gamma polyglutamated antifolate and a non-polyglutamated antifolate that inhibits a GAR or AICAR converting enzyme. In other embodiments, the non-polyglutamated antifolate is selected from the group consisting of: trimetrexate (TMQ), pirtrexin (BW301U), and talotrexin (PT 523). In other embodiments, the non-polyglutamated antifolate is selected from the group consisting of: nolatrexed (AG337), prallexed (ZD9331, BGC9331), and BGC 945(ONX 0801) or pharmaceutically acceptable salts thereof.
C platinum
In some embodiments, the liposomes comprise a gamma polyglutamated antifolate (Lp-gamma pantol, such as PLp-gamma pantol, TPLp-gamma pantol, TLp-gamma pantol and NTLp-gamma pantol), comprise a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein) and a platinum-based chemotherapeutic agent or a salt or acid thereof. In some embodiments, the alpha polyglutamated antifolate/platinum-based agent complex comprises gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the complex comprises a polyglutamated antifolate described in section I.
In some embodiments, the Lp- γ pantol comprises a platinum-based chemotherapeutic agent selected from: cisplatin, carboplatin, and oxaliplatin, or salts or acids thereof. In other embodiments, the Lp- γ pantol comprises an analog of a platinum-based chemotherapeutic agent selected from the group consisting of: cisplatin, carboplatin, or oxaliplatin, or a salt or acid thereof.
In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and cisplatin or a salt or acid thereof. In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and a cis-platinum analog or salt or acid thereof.
In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and carboplatin or a salt or acid thereof. In some embodiments, the liposomes comprise a gamma polyglutamated antifolate and a carboplatin analog or salt or acid thereof.
In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and oxaliplatin or a salt or acid thereof. In some embodiments, the liposome comprises a gamma polyglutamated antifolate and an oxaliplatin analog or salt or acid thereof.
In some embodiments, the liposomes comprise a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein) and a platinum-based chemotherapeutic agent selected from: nedaplatin, heptaplatin and lobaplatin, or salts or acids thereof. In some embodiments, the Lp- γ pantol comprises a gamma polyglutamated antifolate and an analog of a platinum-based chemotherapeutic selected from the group consisting of: nedaplatin, heptaplatin and lobaplatin, or salts or acids thereof.
In some embodiments, the Lp- γ pantol comprises a γ -polyglutamated antifolate and a platinum-based chemotherapeutic selected from the group consisting of: satraplatin, carboplatin, cisplatin, cycloplatin, dexormaplatin, spiroplatin, picoplatin, triplatin, tetraplatin, iproplatin, ormaplatin, zeniplatin, platinum-triamine, traplatin, enloplatin, JM216, 254-S, NK121, CI-973, DWA 2114R, NDDP, and dedaplatin, or salts or acids thereof. In some embodiments, the Lp- γ pantol comprises a gamma polyglutamated antifolate and an analog of a platinum-based chemotherapeutic selected from the group consisting of: satraplatin, carboplatin, cisplatin, cycloplatin, dexormaplatin, spiroplatin, picoplatin, triplatin, tetraplatin, iproplatin, ormaplatin, zeniplatin, platinum-triamine, traplatin, enloplatin, JM216, 254-S, NK121, CI-973, DWA 2114R, NDDP, and dedaplatin, or salts or acids thereof.
In some embodiments, the liposome composition comprises a liposome further comprising one or more of an immunostimulatory agent, a detectable label, and a maleimide disposed on at least one of the PEG and an exterior of the liposome.
D Cyclodextrin
In additional embodiments, the liposome comprises γ pantol (e.g., γ pantol disclosed herein) and a cyclodextrin (e.g., cyclodextrin in section IB herein). In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome comprises a polyglutamated antifolate described in section I. In some embodiments, the liposome is a liposome composition according to any one of [12] to [67] of the detailed description section. In some embodiments, the γ pantol liposome is a targeted liposome composition according to any one of [12] to [67] of the detailed description section.
In some embodiments, the γ pantol liposome comprises a complex formed from a cyclodextrin and a therapeutic agent. In some embodiments, the therapeutic agent is a cytotoxic compound or a salt or acid thereof. In another embodiment, the therapeutic agent is a chemotherapeutic agent or a salt or acid thereof. In another embodiment, the therapeutic agent is a platinum-based drug. In another embodiment, the therapeutic agent is a taxane-based drug. In additional embodiments, the therapeutic agent of the cyclodextrin/therapeutic agent complex is selected from the group consisting of: gemcitabine, gemcitabine-based therapeutics, doxorubicin, antifolates, antifolate-based chemotherapeutics or salts or acid, acid or free base forms thereof. In some embodiments, the γ pantol liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome comprises a polyglutamated antifolate described in section I. In some embodiments, the γ pantol liposome is a liposome composition according to any one of [12] to [67] of the detailed description section. In some embodiments, the γ pantol liposome is a targeted liposome composition according to any one of [12] to [67] of the detailed description section. In further embodiments, the complex has a cyclodextrin/therapeutic agent molar ratio in the range of 1-10: 1. In some embodiments, the molar ratio of gamma pantol/therapeutic agent in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the molar ratio of gamma pantol/therapeutic agent in the complex is 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a cyclodextrin/therapeutic agent molar ratio of: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In further embodiments, the cyclodextrin// platinum-based agent complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In some embodiments, the γ pantol liposome comprises γ pantol and a cyclodextrin/platinum-based chemotherapeutic complex. In some embodiments, the platinum-based chemotherapeutic agent is selected from the group consisting of: cisplatin, carboplatin, and oxaliplatin, or salts or acids thereof. In other embodiments, the cyclodextrin/platinum-based chemotherapeutic agent complex comprises an analog of cisplatin, carboplatin, oxaliplatin, or a salt or acid thereof. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome comprises a polyglutamated antifolate described in section I. In some embodiments, the γ pantol liposome is a liposome composition according to any one of [12] to [67] of the detailed description section. In some embodiments, the γ pantol liposome is a targeted liposome composition according to any one of [12] to [67] of the detailed description section. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio of: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In further embodiments, the cyclodextrin// platinum-based agent complex is encapsulated in a liposome.
In some embodiments, the platinum-based chemotherapeutic agent is selected from the group consisting of: cisplatin, carboplatin, and oxaliplatin, or salts or acids thereof. In other embodiments, the cyclodextrin/platinum-based chemotherapeutic agent complex comprises an analog of cisplatin, carboplatin, oxaliplatin, or a salt or acid thereof. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a cyclodextrin/platinum-based agent molar ratio of: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1.
In other embodiments, the present disclosure provides a complex comprising cyclodextrin and cisplatin or a salt or acid thereof. In some embodiments, the complex has a cyclodextrin/cisplatin (or cisplatin salt or acid) molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/cisplatin (or cisplatin salt or acid) molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the cyclodextrin/cisplatin (or cisplatin salt or acid) molar ratio in the complex is 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a cyclodextrin/cisplatin (or cisplatin salt or acid) molar ratio of: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In further embodiments, the cyclodextrin// cisplatin (or cisplatin salt or acid) complex is encapsulated in a liposome.
In another embodiment, the present disclosure provides a complex comprising a cyclodextrin and carboplatin or a salt or acid thereof. In some embodiments, the complex has a cyclodextrin/carboplatin (or carboplatin salt or acid) molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/carboplatin (or carboplatin salt or acid) molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the molar ratio of cyclodextrin/carboplatin (or carboplatin salt or acid) in the complex is 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a molar ratio of cyclodextrin/carboplatin (or carboplatin salt or acid) of: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In further embodiments, the cyclodextrin/carboplatin (or carboplatin salt or acid) complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In another embodiment, the present disclosure provides a complex comprising cyclodextrin and oxaliplatin or a salt or acid thereof. In some embodiments, the cyclodextrin/oxaliplatin (or oxaliplatin salt or acid) molar ratio in the complex is in the range of 1-10: 1. In some embodiments, the cyclodextrin/oxaliplatin (or oxaliplatin salt or acid) molar ratio in the complex is 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the cyclodextrin/oxaliplatin (or oxaliplatin salt or acid) molar ratio in the complex is 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the molar ratio of cyclodextrin/oxaliplatin (or oxaliplatin salt or acid) in the complex is: 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In further embodiments, the cyclodextrin/oxaliplatin (or oxaliplatin salt or acid) complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In additional embodiments, the present disclosure provides a complex comprising a cyclodextrin and a platinum-based chemotherapeutic agent selected from the group consisting of: nedaplatin, heptaplatin, lobaplatin, satraplatin, carboplatin, cisplatin, cycloplatin, dexormaplatin, spiroplatin, picoplatin, triplatin, tetraplatin, iproplatin, ormaplatin, zeniplatin, platinum-triamine, traplatin, enloplatin, JM216, NK121, CI973, DWA 2114R, NDDP, and dedaplatin, or salts or acids thereof. In other embodiments, the cyclodextrin/platinum-based chemotherapeutic complex comprises an analog of nedaplatin, heptaplatin, lobaplatin, satraplatin, carboplatin, cisplatin, cycloplatin, dexormaplatin, spiroplatin, picoplatin, triplatin, tetraplatin, iproplatin, ormaplatin, zeniplatin, platinum-triamine, traplatin, enloplatin, JM216, NK121, CI973, DWA 2114R, NDDP, or dedaplatin, or a salt or acid thereof. In some embodiments, the cyclodextrin/oxaliplatin (or oxaliplatin salt or acid) molar ratio in the complex is in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/platinum-based chemotherapeutic (or a salt or acid or analog thereof) molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the complex has a cyclodextrin/platinum-based chemotherapeutic (or a salt or acid or analog thereof) molar ratio of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a molar ratio of cyclodextrin/platinum-based chemotherapeutic agent (or salt or acid or analog thereof) of: 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In additional embodiments, the cyclodextrin/platinum-based chemotherapeutic agent (or salt or acid or analog thereof) complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In some embodiments, the present disclosure provides a composition comprising a cyclodextrin/taxane-based chemotherapeutic agent complex. In some embodiments, the taxane-based chemotherapeutic agent is selected from: paclitaxel (PTX), Docetaxel (DTX), Larotaxel (LTX) and Cabazitaxel (CTX), or salts or acids thereof. In some embodiments, the complex has a cyclodextrin/taxane-based agent molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/taxane-based agent molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the complex has a cyclodextrin/taxane-based agent molar ratio of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a cyclodextrin/taxane-based agent molar ratio of: 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In further embodiments, the cyclodextrin// taxane-based agent complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In additional embodiments, the present disclosure provides a complex comprising a cyclodextrin and Paclitaxel (PTX) or a salt or acid thereof. In other embodiments, the cyclodextrin/taxane-based chemotherapeutic complex comprises an analog of Paclitaxel (PTX) or a salt or acid thereof. In some embodiments, the complex has a cyclodextrin/paclitaxel (or paclitaxel salt or acid) molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/paclitaxel (or paclitaxel salt or acid) molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the cyclodextrin/paclitaxel (or paclitaxel salt or acid) molar ratio in the complex is 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a cyclodextrin/paclitaxel (or paclitaxel salt or acid) molar ratio of: 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In additional embodiments, the cyclodextrin/paclitaxel (or paclitaxel salt or acid) complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In additional embodiments, the present disclosure provides a complex comprising cyclodextrin and Docetaxel (DTX) or a salt or acid thereof. In other embodiments, the cyclodextrin/taxane-based chemotherapeutic complex comprises an analog of Docetaxel (DTX) or a salt or acid thereof. In some embodiments, the complex has a cyclodextrin/docetaxel (or docetaxel salt or acid) molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/docetaxel (or docetaxel salt or acid) molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the complex has a cyclodextrin/docetaxel (or docetaxel salt or acid) molar ratio of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the complex has a cyclodextrin/docetaxel (or docetaxel salt or acid) molar ratio of: 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In additional embodiments, the cyclodextrin/docetaxel (or docetaxel salt or acid) complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In additional embodiments, the present disclosure provides a complex comprising a cyclodextrin and ralfataxel (LTX) or a salt or acid thereof. In other embodiments, the cyclodextrin/taxane-based chemotherapeutic complex comprises an analog of ralfataxel (LTX) or a salt or acid thereof. In some embodiments, the complex has a cyclodextrin/raloxitol (or raloxitol salt or acid) molar ratio in the range of 1-10: 1. In some embodiments, the complex has a molar ratio of cyclodextrin/raloxitol (or raloxitol salt or acid) of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the molar ratio of cyclodextrin/raloxitol (or raloxitol salt or acid) in the complex is 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the molar ratio of cyclodextrin/raloxitol (or raloxitol salt or acid) in the complex is: 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In additional embodiments, the cyclodextrin/raloxitol (or raloxitol salt or acid) complex is encapsulated in a liposome (e.g., as described herein or otherwise known in the art).
In additional embodiments, the present disclosure provides a complex comprising cyclodextrin and Cabazitaxel (CTX) or salts or acids thereof. In other embodiments, the cyclodextrin/taxane-based chemotherapeutic complex comprises an analog of Cabazitaxel (CTX) or a salt or acid thereof. In some embodiments, the complex has a cyclodextrin/cabazitaxel (or cabazitaxel salt or acid) molar ratio in the range of 1-10: 1. In some embodiments, the complex has a cyclodextrin/cabazitaxel (or cabazitaxel salt or acid) molar ratio of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, or 10: 1. In some embodiments, the complex has a molar ratio of cyclodextrin/cabazitaxel (or cabazitaxel salt or acid) of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1 (21-50), or 1: > 50. In some embodiments, the molar ratio of alpha cyclodextrin/cabazitaxel (or cabazitaxel salt or acid) in the complex is: 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, (21-50):1 or >50: 1. In additional embodiments, the cyclodextrin/cabazitaxel (or cabazitaxel salt or acid) complexes are encapsulated in liposomes (e.g., as described herein or otherwise known in the art).
The cyclodextrin of the cyclodextrin/therapeutic agent complex may be derivatized or underivatized. In some embodiments, the cyclodextrin is derivatized. In other embodiments, the cyclodextrin is a derivatized β -cyclodextrin (e.g., hydroxypropyl β -cyclodextrin (HP- β -CD) and sulfobutyl ether β -CD (sbe) - β -cyclodextrin)). In some embodiments, the cyclodextrin of the cyclodextrin/therapeutic agent complex is a derivatized β -cyclodextrin comprising: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more 2-hydroxypropyl-3-group substitutions of hydroxyl; or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more sulfoalkyl ether groups of a hydroxyl group. In other embodiments, the cyclodextrin of the cyclodextrin/therapeutic agent complex is a derivatized β -cyclodextrin comprising: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more sulfobutyl ether groups of hydroxyl groups.
In some embodiments, the cyclodextrin of the cyclodextrin/therapeutic agent complex contained in the γ pantol liposome composition is a derivatized cyclodextrin of formula I:
Figure BDA0002715408560000811
wherein: n is 4, 5 or 6; and wherein R1, R2, R3, R4, R5, R6, R7, R8 and R9 are each independently-H, linear or branched C1-C8-alkylene, 2-hydroxypropyl-3-group; or an optionally substituted linear or branched C1-C6 group, wherein at least one of R1, R2, R3, R4, R5, R6, R7, R8 and R9 is a linear or branched C1-C8-alkylene or 2-hydroxypropyl-3-group.
In some embodiments, the cyclodextrin of the cyclodextrin/therapeutic agent complex contained in the γ pantol liposome composition is a derivatized cyclodextrin of formula II:
Figure BDA0002715408560000812
wherein: n is 4, 5 or 6; and wherein R1, R2, R3, R4, R5, R6, R7, R8, and R9 are each independently an-O-or-O- (C2-C6 alkylene) -SO 3-group; wherein at least one of R1 and R2 is independently an-O- (C2-C6 alkylene) -SO 3-group; and S1, S2, S3, S4, S5, S6, S7, S8, and S9 are each independently-H or a pharmaceutically acceptable cation. In other embodiments, wherein the pharmaceutically acceptable cation is selected from the group consisting of: alkali metals, such as Li +, Na +, or K +; alkaline earth metals, such as Ca +2 or Mg +2, and ammonium ions and amine cations, such as the cations of (C1-C6) -alkylamines, piperidines, pyrazines, (C1-C6) -alkanolamines and (C4-C8) -cycloalkanolamines.
In some embodiments, the γ pantol liposome comprises between 100 and 100,000 cyclodextrin/therapeutic agent complexes.
In some embodiments, the cyclodextrin derivative of the γ pantol/cyclodextrin complex and/or cyclodextrin/therapeutic agent complex is any one of the cyclodextrin derivatives of U.S. patent nos. 6,133,248, 5,874,418, 6,046,177, 5,376,645, 5,134,127, 7,034,013, 6,869,939; and the cyclodextrins disclosed in International application publication No. WO02005/117911, the contents of each of which are incorporated herein by reference in their entirety for all purposes.
In some embodiments, the cyclodextrin derivative of the cyclodextrin/therapeutic agent complex is a sulfoalkyl ether cyclodextrin. In some embodiments, the cyclodextrin derivative of the complex is sulfobutyl ether-3-cyclodextrin, e.g.
Figure BDA0002715408560000821
(CyDex Pharma.Inc., Lenexa, Kansas. methods for preparing sulfobutyl ether-3-cyclodextrins and other sulfoalkyl ether cyclodextrins are known in the art.
In some embodiments, the cyclodextrin derivative of the cyclodextrin/therapeutic agent complex is a compound of formula III:
Figure BDA0002715408560000822
wherein R is equal to:
(a)(H)21-Xor (- (CH)2)4-SO3Na) x, and x ═ 1.0 to 10.0, 1.0 to 5.0, 6.0 to 7.0, or 8.0 to 10.0;
(b)(H)21-Xor (- (CH)2CH(OH)CH3) x, and x ═ 1.0 to 10.0, 1.0 to 5.0, 6.0 to 7.0, or 8.0 to 10.0;
(c)(H)21-Xor (sulfoalkyl ether) x, and x ═ 1.0 to 10.0, 1.0 to 5.0, 6.0 to 7.0, or 8.0 to 10.0; or
(d)(H)21-XOr (- (CH)2)4-SO3Na) x, and x ═ 1.0 to 10.0, 1.0 to 5.0, 6.0 to 7.0, or 8.0 to 10.0。
Additional cyclodextrins and cyclodextrin/platinum-based therapeutic agent complexes that can be included in the gamma pantol liposomes and used in accordance with the disclosed methods are disclosed in U.S. application No. 62/583,432, the contents of which are incorporated herein by reference in their entirety.
In some embodiments, the γ pantol liposome comprises a complex of cyclodextrin and a platinum-based chemotherapeutic agent or a salt thereof. In some embodiments, the platinum-based chemotherapeutic agent is cisplatin or a cisplatin analog. In some embodiments, the platinum-based chemotherapeutic agent is carboplatin. In additional embodiments, the liposome composition comprises a platinum-based chemotherapeutic agent selected from the group consisting of: carboplatin, cisplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin, tetraplatin, lipoplatin (lipoplatin), lobaplatin, ormaplatin, zeniplatin, platinum-triamine, traplatin, enloplatin, JM-216, 254-S, NK 121, CI-973, DWA 2114R, NDDP, and dedaplatin. In some embodiments, the γ pantol liposomes comprise between 100 and 100,000 platinum-based chemotherapeutic agent/CD complex. In further embodiments, the liposome composition comprises liposomes having a diameter in the range of 20nm to 500nm or 20nm to 200nm or any range therebetween. In some embodiments, the liposomes in the composition comprise between 100 and 100,000 platinum.
(3) Targeted liposomes
In some embodiments, the present disclosure provides a liposomal gamma-polyglutamated antifolate composition, wherein the liposome comprises a gamma-polyglutamated antifolate and a targeting moiety attached to one or both of PEG and the exterior of the liposome, and wherein the targeting moiety has specific affinity for a surface antigen on a target cell of interest. Such liposomes may be generally referred to herein as "targeted liposomes," e.g., liposomes that include one or more targeting moieties or biodistribution modulators on the surface of the liposome or otherwise attached to the liposome. The targeting moiety of the targeted liposome can be any moiety or agent capable of specifically binding to the desired target (e.g., an antigen target expressed on the surface of a target cell of interest). In one embodiment, the targeted liposome specifically and preferentially binds to a target on the surface of a target cell of interest that internalizes the targeted liposome, and a liposome-encapsulated gamma polyglutamated antifolate (e.g., a gamma pentaglutamated antifolate or a gamma hexaglutamated antifolate) exerts its cytotoxic effect in the target cell of interest. In other embodiments, the target cell is a cancer cell, a tumor cell, or a metastatic cell. In some embodiments, the targeted liposome is pegylated.
The term "attachment" or "linked" refers, for example, to any type of linkage, such as covalent linkage, ionic linkage through hydrophobic interactions (e.g., avidin-biotin), and linkage through a functional group such as maleimide or a linker such as PEG. For example, detectable labels, steric stabilizers, liposomes, liposome components, immunostimulants can be directly linked to each other through maleimide functional groups or PEG-maleimide groups.
The composition and source of the targeting moiety is not limiting to the scope of the present disclosure. In some embodiments, the targeting moiety attached to the liposome is a polypeptide or peptidomimetic ligand. Peptide and peptidomimetic targeting moieties include those having naturally occurring or modified peptides, such as D or L peptides; gamma, beta or gamma peptides; an N-methyl peptide; an azapeptide; peptides having one or more amides, i.e., peptides in which the linkage is replaced with one or more urea, thiourea, carbamate, or sulfonylurea linkages; or a cyclic peptide. Peptidomimetics are molecules that can fold into a defined three-dimensional structure similar to a natural peptide. In some embodiments, the peptide or peptidomimetic targeting moiety is 2-50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
In some embodiments, the targeting moiety polypeptide is at least 40 amino acid residues in length. In other embodiments, the targeting moiety polypeptide is at least 50, 60, 75, 100, 125, 150, 175, 200, 250, or 300 amino acid residues in length.
In further embodiments, a targeting moiety polypeptide, such as an antibody or antigen-binding antibody fragment, binds a target antigen with an equilibrium dissociation constant (Kd) in the range of 0.5x 10 "10 to 10x 10" 6, such as using
Figure BDA0002715408560000831
The assay is analyzed.
In some embodiments, the targeting moiety is an antibody or antibody derivative. In other embodiments, the binding domain of the targeting moiety polypeptide is not derived from the antigen binding domain of an antibody. In some embodiments, the targeting moiety is a polypeptide derived from a binding scaffold selected from DARPin, affilin and armadillo repeats, D domains (see, e.g., WO 2016/164308), Z domains (Affibody), adnectins, lipocalins (lipocalins), affilins, anticalins, knottins, fynomers, atrimers, kunitz domains (see, e.g., WO 2004/063337), CTLA4 or avimer (see, e.g., U.S. publication nos. 2004/0175756, 2005/0053973, 2005/0048512 and 2006/0008844).
In further embodiments, the targeting moiety is an antibody or a derivative of the antigen binding domain of an antibody having specific affinity for an epitope on a target cell surface antigen expressed on the surface of a target cell. In some embodiments, the targeting moiety is a full length antibody. In some embodiments, the targeting moiety is an antigen binding portion of an antibody. In some embodiments, the targeting moiety is a scFv. In other embodiments, the targeting moiety is a Fab. In some embodiments, the targeting moiety comprises a binding domain derived from an antigen binding domain of an antibody (e.g., scFv, Fab ', F (ab')2, Fv fragments, disulfide linked Fv (sdfv), Fd fragments consisting of VH and CH1 domains, scFv, minibody, BiTE, Tandab, diabody ((VL-VH)2 or (VH-VL)2), single domain antibodies (e.g., sdabs, such as nanobodies (VL or VH)) and camelid VHH domains). In some embodiments, the targeting moiety comprises one or more Complementarity Determining Regions (CDRs) derived from an antibody. Examples of suitable antibody-based targeting moieties for the disclosed targeted liposomes include fully human antibodies, humanized antibodies, chimeric antibodies, antigen-binding fragments of antibodies, single chain antibodies, single domain antibodies, bispecific antibodies, synthetic antibodies, pegylated antibodies, and multimeric antibodies. The provided liposome-targeting antibodies can have a combination of the above features. For example, humanized antibodies may be antigen-binding fragments, and may also be pegylated and multimerized.
The term "humanized antibody" refers to a form of a non-human (e.g., murine) antibody that is a specific immunoglobulin chain, chimeric immunoglobulin, or fragment thereof that contains minimal non-human (e.g., murine) sequences. Typically, humanized antibodies are human immunoglobulins in which residues from the Complementarity Determining Regions (CDRs) are replaced by residues from CDRs of non-human species (e.g., mouse, rat, rabbit and hamster) having the desired specificity, affinity and capacity (Jones et al, Nature 321:522-525 (1986); Riechmann et al, Nature 332:323-327 (1988); Verhoeyen et al, Science 239:1534-1536 (1988)). In some cases, Fv Framework Region (FR) residues of the human immunoglobulin are replaced by corresponding residues in antibodies from non-human species having the desired specificity, affinity, and capacity. Humanized antibodies can be further modified by substitution of additional residues in the Fv framework regions and/or within the substituted non-human residues to improve and optimize antibody specificity, affinity, and/or capacity. In general, a humanized antibody will comprise substantially all of at least one and typically two or three variable domains comprising all or substantially all of the CDR regions corresponding to a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody may further comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically at least a portion of a human immunoglobulin. Examples of methods for generating humanized antibodies are described in U.S. Pat. nos. 5,225,539 and 5,639,641.
In other embodiments, the targeting moiety has specific affinity for an epitope on a surface antigen of the target cell of interest. In some embodiments, the target cell is a cancer cell. In some embodiments, the target cell is a tumor cell. In other embodiments, the target cell is an immune cell.
In some embodiments, the targeting moiety has specific affinity for an epitope expressed on a tumor cell surface antigen. The term "tumor cell surface antigen" refers to an antigen common to particular hyperproliferative disorders, such as cancer. In some embodiments, the targeting moiety has specific affinity for an epitope of a tumor cell surface antigen that is a Tumor Associated Antigen (TAA). TAAs are antigens found on both tumors and some normal cells. When the immune system is immature and unable to respond, TAAs may be expressed on normal cells during fetal development, or may be present at very low levels on normal cells in general, and at much higher levels on tumor cells. Due to the dynamic nature of tumors, in some cases, tumor cells may express distinct antigens at certain stages, and also express antigens that are also expressed on non-tumor cells at other stages. Thus, the inclusion of a marker as a TAA does not exclude that it is considered a tumor specific antigen. In some embodiments, the targeting moiety has specific affinity for an epitope of a tumor cell surface antigen that is a Tumor Specific Antigen (TSA). TSA is an antigen specific to tumor cells and is not present on other cells in the body. In some embodiments, the targeting moiety has specific affinity for an epitope of a tumor cell surface antigen expressed on the surface of a cancer, including, but not limited to, primary or metastatic melanoma, thymoma, lymphoma, sarcoma, lung cancer (e.g., NSCLC or SCLC), liver cancer, non-hodgkin's lymphoma, leukemia, multiple myeloma, glioblastoma, neuroblastoma, uterine cancer, cervical cancer, renal cancer, thyroid cancer, bladder cancer, renal cancer, mesothelioma, and adenocarcinomas such as breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, colon cancer, and others known in the art. In some embodiments, the targeting moiety has specific affinity for an epitope of a cell surface antigen expressed on the cell surface in the tumor microenvironment (e.g., and antigens such as VEGFR and TIE1 or TIE2 expressed on endothelial cells and macrophages, respectively, or antigens expressed on tumor stromal cells such as cancer-associated fibroblasts (CAF), tumor infiltrating T cells and other leukocytes, and myeloid cells including mast cells, eosinophils, and tumor-associated macrophages (TAMs)).
In some embodiments, a targeted liposomal γ pantol composition (e.g., TLp- γ pantol or TPLp- γ pantol) comprises a targeting moiety that has specific affinity for an epitope of a cancer or tumor cell surface antigen that is preferentially/differentially expressed on a target cell (e.g., a cancer cell or tumor cell) as compared to a normal or non-tumor cell, the epitope being present in the tumor cell but absent or inaccessible on the non-tumor cell. For example, in some cases, tumor antigens are located on the surface of normal and malignant cancer cells, but tumor epitopes are only exposed in cancer cells. As another example, a tumor cell surface antigen may undergo a confirmed change in cancerous state that results in the presence of a cancer cell specific epitope. Targeting moieties having specific affinity for an epitope on a tumor cell surface antigen described herein or otherwise known in the art are useful and are encompassed by the disclosed compositions and methods. In some embodiments, the tumor cell having a tumor cell surface antigen is a cancer cell. Examples of such tumor cell surface antigens include, but are not limited to, folate receptor alpha, folate receptor beta, and folate receptor.
In other embodiments, the targeting moiety comprises a polypeptide targeting moiety, such as an antibody or antigen binding antibody fragment, and the targeting moiety has binding specificity for the folate receptor. In some embodiments, e.g., using
Figure BDA0002715408560000841
The assay determines that the targeting moiety binds to folate receptor with an equilibrium dissociation constant (Kd) in the range of 0.5x 10-10 to 10x 10-6. In some embodiments, the folate receptor bound by the targeting moiety is one or more folate receptors selected from the group consisting of: folate receptor alpha (FR-. alpha.), folate receptor beta (FR-. beta.), and folate receptor (FR-). In another embodiment, the targeting moiety is directed against at least two antigens selected from the group consisting of folate receptor alpha, folate receptor beta and folate receptorHas specific affinity. In another embodiment, the targeting moiety is directed to folate receptor alpha; folate receptor beta; and folate receptors have specific affinities.
In some embodiments, the targeting moiety has specific affinity for an epitope of a cell surface antigen that, when bound, internalizes the targeting moiety. Many cell surface antigens that internalize a binding partner, such as an antibody, upon binding are known in the art and are considered to be binding targets for the targeting moieties disclosed herein that are expressed on targeted liposomal gamma pantol compositions (e.g., TLp-gamma pantol or TPLp-gamma pantol).
In some embodiments, the targeting moiety has specific affinity for an epitope of a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA.
In some embodiments, the targeting moiety has specific affinity for an epitope of a cell surface antigen selected from the group consisting of mannose-6-phosphate receptor, transferrin receptor, and Cell Adhesion Molecule (CAM). In other embodiments, the targeting moiety has specific affinity for an epitope of a CAM selected from the group consisting of: intercellular adhesion molecule (ICAM), platelet-endothelial adhesion molecule (PECAM), activated leukocyte adhesion molecule (ALCAM), B-lymphocyte cell adhesion molecule (BL-CAM), Vascular Cell Adhesion Molecule (VCAM), mucosal vascular addressen cell adhesion molecule (MAdCAM), CD44, LFA-2, LFA-3, and basal immunoglobulin.
The Folate Receptor (FR) discussed herein is distinct from the Reducing Folate Carrier (RFC) and utilizes a different pathway to bring folate and antifolates into the cell. In some embodiments, the targeting moiety specifically binds to a folate receptor. In other embodiments, the targeting moiety specifically binds to a folate receptor selected from the group consisting of folate receptor alpha, folate receptor beta, and folate receptor. Antibodies to folate receptor alpha can be routinely generated using techniques known in the art. In addition, the sequences of many anti-folate receptor antibodies are of the public domain and/or are commercially available and readily available.
Murine antibodies directed against the folate receptor are examples of antibodies that can be used as targeting moieties for the disclosed targeted liposomes, are murine antibodies directed against the folate receptor. The sequences of these antibodies are known and described, for example, in U.S. patent nos. 5,646,253; 8,388,972, respectively; 8,871,206, respectively; and 9,133,275, and international application numbers PCT/US2011/056966 and PCT/US 2012/046672. For example, based on sequences already disclosed in the public domain, genes for antibodies can be synthesized and placed in transient expression vectors and antibodies produced in the HEK-293 transient expression system. The antibody can be a whole antibody, a Fab, or any of the various antibody variants discussed herein or known in the art.
In some embodiments, the targeting liposome (e.g., TL- γ pantol or TPL- γ pantol) contains 1 to 1,000 or more than 1,000 targeting moieties on its surface. In some embodiments, the targeted liposomes contain 30 to 1,000, 30 to 500, 30 to 250, or 30-200 targeting moieties or any range therebetween. In some embodiments, the targeting liposome (e.g., TL- γ pantol or TPL- γ pantol) contains 30 to 1,000 or more than 1,000 targeting moieties on its surface. In some embodiments, the targeted liposome contains 30 to 500, 30 to 250, or 30-200 targeting moieties. In some embodiments, the targeted liposome contains less than 220 targeting moieties, less than 200 targeting moieties, or less than 175 targeting moieties. In some embodiments, the targeting moiety is non-covalently bonded to the exterior of the liposome (e.g., via ionic interaction or GPI anchor). In some embodiments, the targeted liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the targeted liposome comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the targeted liposome is a liposome according to any one of [48] - [67] of the detailed description section.
In some embodiments, the molecules on the exterior of the targeted liposome (e.g., TL- γ pantol or TPL- γ pantol) include lipids, targeting moieties, steric stabilizers (e.g., PEG), maleimides, and cholesterol. In some embodiments, the targeting moiety is covalently bound via a maleimide functional group. In some embodiments, the targeting moiety is covalently attached to a liposome component or a steric stabilizer, such as a PEG molecule. In some embodiments, all targeting moieties of the liposome are bound to one component of the liposome, such as PEG. In other embodiments, the targeting moiety of the targeted liposome is bound to a different component of the liposome. For example, some targeting moieties may be conjugated to a lipid component or cholesterol, some targeting moieties may be conjugated to a steric stabilizer (e.g., PEG), and other targeting moieties may be conjugated to a detectable label or another targeting moiety. In some embodiments, the exterior of the targeted liposome (e.g., TL- γ pantol or TPL- γ pantol) further comprises one or more of an immunostimulatory agent, a detectable label, and a maleimide disposed on at least one of the PEG and the exterior of the liposome. In some embodiments, the targeted liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the targeted liposome comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the targeted liposome is a liposome according to any one of [48] - [67] of the detailed description section.
In some embodiments, the targeting liposome (e.g., TL- γ pantol or TPL- γ pantol) is anionic or neutral. In some embodiments, the targeted anionic or neutral liposomes have a diameter in the range of 20nm to 500nm or 20nm to 200nm or any range therebetween. In other embodiments, the targeted anionic or neutral liposomes have a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the targeted liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the targeted liposome comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the targeted liposome is a liposome according to any one of [48] - [67] of the detailed description section.
In other embodiments, the targeting liposome (e.g., TL-gamma PANTIFOL or TPL-gamma PANTIFOL) is cationic. In some embodiments, the targeted anionic or neutral liposomes have a diameter in the range of 20nm to 500nm or 20nm to 200nm or any range therebetween. In other embodiments, the targeted anionic or neutral liposomes have a diameter in the range of 80nm to 120nm or any range therebetween. In some embodiments, the targeted liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the targeted liposome comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the targeted liposome is a liposome according to any one of [48] - [67] of the detailed description section.
In further embodiments, the liposome composition comprising the targeted liposomes (e.g., TL- γ pantol or TPL- γ pantol) comprises 30% -70%, 30% -60%, or 30% -50% liposome-embedded γ polyglutamated antifolate agent, or any range therebetween. In some embodiments, the liposome composition comprising the targeted liposomes comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75% or more than 75% of the starting material of the gamma polyglutamated antifolate encapsulated (embedded) in the targeted liposomes.
In some embodiments, the targeted liposome composition comprises 30% -70%, 30% -60%, or 30% -50% (w/w) of a gamma-tetraglutamated antifolate, or any range therebetween. In some embodiments, the targeted liposome comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or more than 75% (w/w) of a gamma-tetraglutamated antifolate. In some embodiments, at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or more than 75% of the starting material of the gamma-tetraglutamylated antifolate is encapsulated (embedded) in the targeted liposomes during the process of making the targeted liposomes.
In some embodiments, the targeted liposome composition comprises 30% -70%, 30% -60%, or 30% -50% (w/w) of a gamma-pentaglutamated antifolate, or any range therebetween. In some embodiments, the targeted liposome comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or more than 75% (w/w) of a gamma-pentaglutamated antifolate. In some embodiments, at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or more than 75% of the starting material of the gamma-pentaglutaminated antifolate is encapsulated (embedded) in the targeted liposome during the process of making the targeted liposome.
In some embodiments, the targeted liposome composition comprises 30% -70%, 30% -60%, or 30% -50% (w/w) of a gamma hexaglutaminated antifolate, or any range therebetween. In some embodiments, the targeted liposome comprises at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or more than 75% (w/w) of a gamma hexaglutamylated antifolate. In some embodiments, at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or more than 75% of the starting material of the gamma hexaglutamylated antifolate is encapsulated (embedded) in the targeting liposome during the process of preparing the targeting liposome.
Methods and techniques for covalently associating a polypeptide targeting moiety with a liposome surface molecule are known in the art and can be readily used to prepare TL- γ pantol or TPL- γ pantol liposome compositions.
Chemical binding of non-protein targeting moieties and other compositions to the surface of liposomes can be employed. Thus, the non-protein moiety may be covalently or non-covalently attached to, entrapped in, or adsorbed onto the liposome using any attachment or conjugation method known in the art and/or any suitable chemical linker. The exact type and chemical nature of such cross-linking agents and cross-linking methods are preferably adapted to the type of affinity groups used and the nature of the liposomes. Methods for binding or adsorbing or linking targeting moieties are also well known in the art. For example, in some embodiments, the targeting moiety may be attached to the group at the interface via, but not limited to, a polar group such as amino, SH, hydroxyl, aldehyde, formyl, carboxyl, His-tag, or other polypeptide. In addition, the targeting moiety can be attached via, but is not limited to, an active group such as succinimidyl succinate, cyanuric chloride, tosyl activating group, imidazole group, CNBr, NHS, activated CH, ECH, EAH, epoxy, thiopropyl, activated thiol, and the like. Furthermore, the targeting moiety may be attached via, but is not limited to, hydrophobic bonds (van der waals forces) or electrostatic interactions (e.g., dianions, polyanions, poly-cations, etc.) that may or may not include a cross-linking agent.
Production of liposomes
In some embodiments, the present disclosure provides a method of making a liposome composition disclosed herein. In one embodiment, the method comprises forming a mixture comprising: (1) a liposome component; and (2) gamma polyglutamated (e.g., penta-or hexa-glutamated) antifolates in aqueous solution. In other embodiments, the mixture comprises a pegylated liposome component. The mixture is then homogenized to form liposomes in aqueous solution. In addition, the mixture can be extruded through a membrane to form liposomes that encapsulate the gamma polyglutamated antifolate in an aqueous solution. It is to be understood that the liposomal composition of the present disclosure may comprise any lipid (including cholesterol), including functionalized lipids and lipids linked to a targeting moiety, a detectable label, and a steric stabilizer, or any subset of all of these. It should also be noted that the bioactive gamma polyglutamated antifolate agent in aqueous solution may comprise any of the reagents and chemicals discussed herein or otherwise known in the art, either internal or external to the liposome, including, for example, buffers, salts, and cryoprotectants.
In some embodiments, the present disclosure provides a method of making a targeted pegylated liposomal gamma polyglutamated antifolate (targeted PLp-gamma pantol) or non-targeted PLp-gamma pantol disclosed herein. In one embodiment, the method comprises forming a mixture comprising: (1) a liposome component; (2) gamma polyglutamated (e.g., pentaglutamated or hexaglutamated) antifolates in aqueous solution; and (3) a targeting moiety. The mixture is then homogenized to form liposomes in aqueous solution. In addition, the mixture can be extruded through a membrane to form liposomes that encapsulate the targeted gamma polyglutamated antifolate in an aqueous solution. It is to be understood that the targeted pegylated liposome component may comprise any lipid (including cholesterol), including functionalized lipids and lipids linked to a targeting moiety, a detectable label, and a steric stabilizer, or any subset of all of these. It should also be noted that targeted pegylated liposomes can comprise any of the reagents and chemicals discussed herein or known in the art either internal or external to the liposome, including, for example, buffers, salts, and cryoprotectants.
The above method optionally further comprises the step of lyophilizing the composition after the removing step to form a lyophilized composition. As described above, the targeted or non-targeted PTPLA in aqueous solution may comprise a cryoprotectant as described herein or otherwise known in the art. Cryoprotectants may be preferred if the composition is to be lyophilized.
In addition, after the lyophilizing step, the method optionally further comprises the step of reconstituting the lyophilized composition by dissolving the composition in a solvent after the lyophilizing step. Methods of reconstruction are known in the art. One preferred solvent is water. Other preferred solvents include saline solutions and buffered solutions.
While certain exemplary embodiments are discussed herein, it is to be understood that liposomes can be prepared by any method known in the art. See, e.g., g. gregoriadis (ed.), Liposome Technology, volumes 1-3, 1 st edition, 1983; 2 nd edition, 1993, CRC Press,45Boca Raton, Fla. Examples of methods suitable for preparing liposome compositions include extrusion, reverse phase evaporation, sonication, solvent (e.g., ethanol) infusion, microfluidization, detergent dialysis, ether infusion, and dehydration/rehydration. The size of the liposomes can generally be controlled by controlling the pore size of the membrane used for low pressure extrusion or the pressure used in the microfluidics and the number of passes or any other suitable method known in the art.
Typically, the gamma polyglutamated antifolate agent is contained within the interior of the liposome, i.e., in the interior (internal) space. In one embodiment, the substituted ammonium is partially or substantially completely removed from the external medium surrounding the liposome. Such removal can be accomplished by any suitable means known in the art (e.g., dilution, ion exchange chromatography, size exclusion chromatography, dialysis, ultrafiltration, and precipitation). Thus, the method of preparing the liposome composition described above or otherwise known in the art may optionally further comprise the step of removing the gamma polyglutamated antifolate agent from the aqueous solution outside the liposome after formation of the liposomes, e.g. by homogenization or by an extrusion step.
In other embodiments, the present disclosure provides a targeted pegylated liposomal gamma-polyglutamated antifolate (TPLp-gamma pantol) that selectively targets a folate receptor, comprising: a liposome comprising an interior space; a gamma polyglutamated antifolate agent disposed within the interior space; a steric stabilizer molecule attached to the exterior of the liposome; and a targeting moiety comprising a protein having specific affinity for at least one folate receptor, said targeting moiety being linked to at least one of a steric stabilizer and the exterior of the liposome. The components of this embodiment may be the same as described for other embodiments of the disclosure. For example, targeted pegylated liposomal gamma polyglutamated antifolates and steric stabilizers which may be PEG are as described in other sections of this disclosure.
In some embodiments, the present disclosure provides a method of making a targeted composition comprising pegylated liposomes comprising entrapped and/or encapsulated gamma polyglutamated antifolate agents; a targeting moiety is an amino acid chain comprising a plurality of amino acids, the targeting moiety having a specific affinity for at least one type of folate receptor, the specific affinity being defined as an equilibrium dissociation constant (Kd) that includes in the range of 0.5x 10 "10 to 10x 10" 6 moles [0.05nM to 10 μ Μ ] for at least one type of folate receptor, the targeting moiety being attached to one or both of PEG and the exterior of the liposome, the method comprising: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; processing the mixture to form liposomes encapsulating and/or encapsulating the gamma polyglutamated antifolate agent; and providing a targeting moiety on the surface of the liposome embedding and/or encapsulating the gamma polyglutamated antifolate agent, said targeting moiety having specific affinity for at least one of folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and folate receptor (FR-). In some embodiments, the method comprises: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; forming liposomes that entrap and/or encapsulate the gamma polyglutamated antifolate agent, e.g., by homogenizing or otherwise processing the mixture to form liposomes; and providing a targeting moiety on the surface of the liposome embedding and/or encapsulating the gamma polyglutamated antifolate agent, said targeting moiety having specific affinity for at least one of folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and folate receptor (FR-). In some embodiments, the processing comprises one or more of: film hydration, extrusion, in-line mixing, ethanol injection techniques, freeze-thaw techniques, reverse phase evaporation, dynamic high pressure micro-jets, micro-jet mixing, multiple emulsion methods, freeze-dried multiple emulsion methods, 3D printing, membrane contactor methods, and agitation, and once the particles are formed, the size of the particles can be further altered by one or more of extrusion and ultrasonic treatment. In some embodiments, at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or more than 75% of the starting material of the gamma polyglutamated antifolate is encapsulated (embedded) in the targeted liposome during the process of preparing the liposome. In some embodiments, the liposome is anionic or neutral. In some embodiments, the targeting moiety has a specific affinity for one or more of: folate receptor alpha (FR-. alpha.), folate receptor beta (FR-. beta.), and folate receptor (FR-). In other embodiments, the targeting moiety has specific affinity for folate receptor alpha (FR-alpha) and folate receptor beta (FR-beta). In further embodiments, the targeting moiety has specific affinity for an epitope on a tumor cell surface antigen that is present on a tumor cell but absent or inaccessible on a non-tumor cell.
Liposomes can also be prepared to target specific cells, organs or organelles by altering phospholipid composition or by inserting receptors or counter-receptors into the liposome. For example, liposomes prepared with high levels of nonionic surfactants have been used to target the liver. (see, e.g., Japanese patent 04-244,018 to Hayakawa et al; Kato et al, biol. pharm. Bull.16:960,1993.) Liposomal formulations of Dipalmitoylphosphatidylcholine (DPPC) with a mixture of soy-derived Sterol Glycosides (SG) and cholesterol (Ch) have also been shown to target the liver. (see, e.g., Shimizu et al, biol. pharm. Bull.20:881 (1997)).
Antibody delivery vehicle
In further embodiments, the present disclosure provides an antibody delivery vehicle (e.g., an ADC). In some embodiments, the present disclosure provides an immunoconjugate having the formula (a) - (L) - (γ pantol), wherein: (A) is an antibody or antigen-binding fragment of an antibody; (L) is a linker; and "(γ pantol)" is a γ pantol composition as described herein; and wherein the linker (L) connects (a) to (γ pantol). In some embodiments, the γ pantol is γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the polyglutamated antifolate is an antifolate described in section I herein.
In some embodiments, the antibody or antigen-binding antibody fragment has specific affinity for an epitope of a cell surface antigen on a target cell (e.g., an epitope and/or antigen described herein). In certain embodiments, the antibody binds to an antigen target expressed in or on the cell membrane (e.g., on the cell surface) of the cancer/tumor, and upon binding to the (antigen) target, the antibody is internalized by the cell, after which the γ pantol is released intracellularly. In some embodiments, the antibody is a full length antibody.
(A) The antibody or antigen-binding antibody fragment of the- (L) - (γ pantol) immunoconjugate may be an IgA, IgD, IgE, IgG or IgM antibody. The different classes of immunoglobulins have different and well-known subunit structures and three-dimensional configurations. In certain embodiments, the antibody is an IgG antibody. In some embodiments, the antibody is an IgG1, IgG2, IgG3, or IgG4 antibody. In certain embodiments, the antibody is an IgG1 antibody.
In some embodiments, (a) is an antigen-binding fragment of an antibody. In some embodiments, (a) is an antigen-binding fragment of an antibody.
A "linker" is any chemical moiety capable of linking a compound (typically a drug, such as γ pantol) to an antibody or antigen-binding fragment of an antibody in a stable covalent manner. The linker may be susceptible or substantially resistant to acid-induced cleavage, light-induced cleavage, peptidase-induced cleavage, esterase-induced cleavage, and disulfide bond cleavage under conditions in which the compound or antibody retains activity. Suitable linkers are well known in the art and include, for example, disulfide groups, thioether groups, acid labile groups, photolabile groups, peptidase labile groups, and esterase labile groups. Linkers also include charged linkers and hydrophilic forms thereof.
In some embodiments, the linker is selected from the group consisting of a cleavable linker, a non-cleavable linker, a hydrophilic linker, and a dicarboxylic acid-based linker. In another embodiment, the linker is a non-cleavable linker. In another embodiment, the linker is selected from the group consisting of: n-succinimidyl 4- (2-pyridyldithio) valerate (SPP); 4- (2-pyridyldithio) butanoic acid N-succinimide ester (SPDB) or 4- (2-pyridyldithio) -2-sulfobutanoic acid N-succinimide ester (sulfo-SPDB); 4- (maleimidomethyl) cyclohexane-carboxylic acid N-succinimidyl ester (SMCC); 4- (maleimidomethyl) cyclohexanecarboxylic acid N-sulfosuccinimidyl ester (sulfo SMCC); n-succinimidyl 4- (iodoacetyl) -aminobenzoate (SIAB); and N-succinimidyl- [ (N-maleimidopropionamido) -tetraethylene glycol ] ester (NHS-PEG 4-maleimide). In another embodiment, the linker is N-succinimidyl- [ (N-maleimido-propionamido) -tetraethylene glycol ] ester (NHS-PEG 4-maleimide).
In some embodiments, the gamma polyglutamated antifolate is linked (coupled) to the antibody or antigen-binding antibody fragment of the immunoconjugate, either directly or through a linker, using techniques known in the art. Such attachment of one or more gamma pantol can involve a number of chemical mechanisms, such as covalent binding, affinity binding, insertion, coordination binding and complexation. Covalent binding of gamma pantol to an antibody or antigen-binding antibody fragment can be achieved by direct condensation of existing side chains or by incorporation of external bridging molecules. Many di-or polyvalent agents can be used to associate the polypeptide with other proteins using coupling agents such as carbodiimides, diisocyanates, glutaraldehyde, diazobenzenes, and hexamethylenediamine. This list is not intended to be exhaustive of the various coupling agents known in the art, but rather is exemplary of the more common coupling agents. In some embodiments, the antibody or antigen-binding antibody fragment is derivatized and then linked to a gamma polyglutamated antifolate. Alternatively, γ pantol can be derivatized and linked to an antibody or antigen-binding antibody fragment using techniques known in the art.
In some embodiments, the immunoconjugate comprises an antibody or antigen-binding fragment of an antibody and γ pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups (including glutamyl groups of antifolates). In some embodiments, the immunoconjugate comprises the γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the immunoconjugate comprises a polyglutamate of an antifolate described in part I herein. In some embodiments, the immunoconjugate comprises a gamma polyglutamated antifolate comprising two or more glutamyl groups in L form. In other embodiments, the immunoconjugate comprises a gamma polyglutamated antifolate agent comprising a glutamyl group in D form. In other embodiments, the immunoconjugate comprises a gamma polyglutamated antifolate comprising a glutamyl group in the D form and two or more glutamyl groups in the L form. In further embodiments, the immunoconjugate comprises a gamma polyglutamated antifolate comprising two or more glutamyl groups having a gamma carboxy linkage. In some embodiments, the immunoconjugate comprises a gamma-pentaglutaminated antifolate. In other embodiments, the immunoconjugate comprises an L-gamma-pentaglutamated antifolate, a D-gamma-pentaglutamated antifolate, or an L-and D-gamma-pentaglutamated antifolate. In some embodiments, the immunoconjugate comprises a gamma hexaglutaminated antifolate (Lp-gamma pantol). In other embodiments, the immunoconjugate comprises an L-gamma hexaglutaminated antifolate, a D-gamma hexaglutaminated antifolate, or an L-and D-gamma hexaglutaminated antifolate.
In some embodiments, the antibody delivery vehicle composition comprises a gamma polyglutamated antifolate and an antibody or antigen-binding antibody fragment having specific affinity for an epitope on a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, bindin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, Pcadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin receptor, HER2, HER 72, EGFR, EGFRvIII, FGFR 72, 3, FGFR3, CD 36ZD 3, CD 36ZD 3, CD 36, CD19, CD20, CD22, CD26, CD27L, CD28, CD30, CD33, CD34, CD37, CD38, CD40, CD40L, CD44, CD56, CD70, CD74, CD79, CD79 79, CD105, CD133, CD138, cripto, IGF-1 79-2 79, EphA receptors, EphB receptors, EphA 79, EphB 79, integrin (for example, integrin α v β 3, α v β 5 or α v β 6), TrkC, VEGFR 72, PDGF, VEGF-PGR, VEGF-P79, VEGF-P11, VEGF-P11, VEGF-P-. In some embodiments, the delivery vehicle comprises a targeting moiety with specific affinity for an epitope on a cell surface antigen (such as a neoantigen) derived from or determined to be expressed on a cancer (tumor) in a particular subject. In some embodiments, the antibody delivery vehicle composition comprises a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section.
In some embodiments, the antibody delivery vehicle composition comprises a gamma polyglutamated antifolate and an antibody or antigen-binding antibody fragment having specific affinity for an epitope on an antigen selected from the group consisting of mannose 6-phosphate receptor, transferrin receptor, and Cell Adhesion Molecule (CAM). In other embodiments, the targeting moiety has specific affinity for an epitope of a CAM selected from the group consisting of: intercellular adhesion molecule (ICAM), platelet-endothelial adhesion molecule (PECAM), activated leukocyte adhesion molecule (ALCAM), B-lymphocyte cell adhesion molecule (BL-CAM), Vascular Cell Adhesion Molecule (VCAM), mucosal vascular addressen cell adhesion molecule (MAdCAM), CD44, LFA-2, LFA-3, and basal immunoglobulin. In some embodiments, the antibody delivery vehicle composition comprises a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section.
In some embodiments, the antibody delivery vehicle composition comprises 1, 2, 3, 4, 5-10, or greater than 10 gamma polyglutamated antifolates. In some embodiments, the antibody delivery vehicle composition comprises 1, 2, 3, 4, 5-10, or greater than 10 gamma-pentaglutaminated antifolates. In some embodiments, the antibody delivery vehicle composition comprises 1, 2, 3, 4, 5-10, or greater than 10 gamma hexaglutaminated antifolates. In some embodiments, the antibody delivery vehicle composition comprises a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section.
Pharmaceutical compositions and administration
In some embodiments, the liposome composition is provided as a pharmaceutical composition comprising a liposome and a carrier (e.g., a pharmaceutically acceptable carrier). In some embodiments, the liposome composition is a liposome according to any one of [12] to [67] of the detailed description section. Examples of pharmaceutically acceptable carriers included in the provided pharmaceutical compositions include physiological saline, isotonic dextrose, isotonic sucrose, ringer's solution, and hanks solution. In some embodiments, a buffering substance is added to maintain the optimal pH for storage stability of the pharmaceutical composition. In some embodiments, the pH of the pharmaceutical composition is between 6.0 and 7.5. In some embodiments, the pH is between 6.3 and 7.0. In other embodiments, the pH is 6.5. Ideally, the pH of the pharmaceutical composition allows for the stability of the liposome membrane lipids and retention of the embedded entities. Histidine, hydroxyethylpiperazine-ethylsulfonate (HEPES), Morpholinoethylsulfonate (MES), succinate, tartrate and citrate, typically at concentrations of 2-20mM, are exemplary buffer substances. Other suitable carriers include, for example, water, aqueous buffer, 0.4% NaCl, and 0.3% amino acid. Protein, carbohydrate or polymer stabilizers and tonicity adjusting agents, such as gelatin, albumin, dextran or polyvinylpyrrolidone, may be added. The tonicity of the composition may be adjusted to a physiological level of 0.25-0.35mol/kg with glucose or a more inert compound such as lactose, sucrose, mannitol or dextrin. These compositions may be conventionally sterilized using conventional sterilization techniques known in the art (e.g., by filtration). The resulting aqueous solution may be packaged for use or filtered under sterile conditions and lyophilized, the lyophilized formulation being combined with a sterile aqueous medium prior to administration.
The provided pharmaceutical liposome compositions may also contain other pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, and tonicity adjusting agents, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, and calcium chloride. Additionally, the liposome suspension may include lipid protectors that protect the lipids from free radicals and lipid peroxidation upon storage. Lipophilic free radical quenchers such as gamma tocopherol and water soluble iron specific chelators such as ferrioxamine are suitable.
The concentration of liposomes in the provided fluid pharmaceutical formulations can vary widely as desired, for example, typically less than about 0.05% by weight or at least about 2% -10% by weight up to 30% to 50% by weight and will be selected primarily by fluid volume and viscosity depending on the particular mode of administration selected. For example, the concentration may be increased to reduce the fluid load associated with the treatment. This may be particularly desirable in patients with atherosclerosis-associated congestive heart failure or severe hypertension. Alternatively, the liposomal pharmaceutical composition consisting essentially of the irritating lipid may be diluted to a low concentration to reduce inflammation at the site of administration.
Some embodiments relate to a method of delivering targeted pegylated liposomal formulations of gamma polyglutamated antifolates to tumors expressing folate receptors on the surface. An exemplary method comprises the step of administering a liposomal pharmaceutical composition provided herein in an amount to deliver a therapeutically effective dose of a targeted pegylated liposomal gamma-polyglutamated antifolate agent to a tumor.
The amount of liposomal pharmaceutical composition administered will depend on the particular gamma polyglutamated antifolate agent entrapped within the liposomes, the disease state being treated, the type of liposomes used, and the judgment of the clinician. Typically, the amount of liposomal pharmaceutical composition administered will be sufficient to deliver a therapeutically effective dose of the particular therapeutic entity.
The amount of liposomal pharmaceutical composition necessary to deliver a therapeutically effective dose can be determined by conventional in vitro and in vivo methods common in the art of pharmaceutical testing. See, e.g., d.b. budman, a.h.calvert, e.k.rowinsky (editors.) Handbook of Anticancer Drug Development, LWW, 2003. Therapeutically effective dosages of various therapeutic compositions are known to those skilled in the art. In some embodiments, the therapeutic entity is delivered via a pharmaceutical liposome composition and provides at least the same or higher activity as that obtained by administering the same amount of therapeutic entity in its conventional non-liposomal formulation. Typically, the dosage of the liposomal pharmaceutical composition most typically ranges between about 0.005 and about 5000 grams of therapeutic entity per square meter of body surface area, between about 0.1 and about 1000 grams of therapeutic entity per square meter of body surface area.
For example, if the subject has a tumor, the effective amount can be the amount of agent (e.g., a gamma polyglutamated antifolate composition) that reduces tumor volume or burden (e.g., as determined by imaging the tumor). The effective amount may also be routinely assessed by the presence and/or frequency of cancer cells in blood or other body fluids or tissues (e.g., biopsies). If the tumor is affecting the normal function of a tissue or organ, the effective amount can be routinely assessed by measuring the normal function of the tissue or organ. In some instances, an effective amount is that amount necessary to alleviate or eliminate one or more, and preferably all, of the symptoms.
Also provided are pharmaceutical compositions comprising gamma polyglutamated antifolate compositions (e.g., liposomes containing pentaglutamated or hexaglutamated antifolates). The pharmaceutical composition is a sterile composition comprising the sample liposomes, and preferably comprising the gamma polyglutamated antifolate agent, preferably in a pharmaceutically acceptable carrier.
Unless otherwise indicated herein, a variety of routes of administration may be used. The particular mode selected will depend on the particular active agent selected, the particular condition being treated and the dosage required for therapeutic efficacy. The provided methods may be practiced using any known mode of administration that is medically acceptable and in accordance with good medical practice. In some embodiments, the route of administration is injection. In other embodiments, the injection is by a parenteral route selected from intramuscular, subcutaneous, intravenous, intraarterial, intraperitoneal, intraarticular, intradural, intrathecal, intravenous, intramuscular, or intrasternal injection. In some embodiments, the route of administration is infusion. In additional embodiments, the route of administration is oral, nasal, mucosal, sublingual, intratracheal, ocular, rectal, vaginal, ocular, topical, transdermal, pulmonary, or inhalation.
Therapeutic compositions containing a gamma pantol composition, such as a liposomal gamma pantol composition as described herein, can be routinely administered intravenously, e.g., by injection of unit doses. The term "unit dose" when used in relation to the therapeutic compositions provided herein, refers to physically discrete units suitable as unitary dosages for subjects, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a desired diluent (e.g., carrier or vehicle). In a specific embodiment, the therapeutic composition containing the adapter is administered subcutaneously.
In some embodiments, the γ -pantol composition is administered in a manner compatible with dosage formulation and in a therapeutically effective amount. The amount to be administered depends on the subject to be treated, the ability of the subject's system to utilize the active ingredient, and the degree of therapeutic effect desired. The precise amount of active ingredient required for administration depends on the judgment of the practitioner and is specific to each individual. However, suitable dosage ranges for systemic administration are disclosed herein and depend on the route of administration. Suitable regimens for administration are also variable, but are represented by repeated doses at one or more hourly intervals, by initial administration, followed by subsequent injection or other administration. Alternatively, continuous intravenous infusion sufficient to maintain the concentration in the blood within the range prescribed by in vivo therapy is contemplated.
The gamma pantol compositions are formulated, dosed and administered in a manner consistent with good medical practice. Considerations in this context include the particular condition being treated, the particular patient being treated, the clinical condition of the individual patient, the etiology of the condition, the site of delivery of the agent, the method of administration, the time course of administration, and other factors known to medical practitioners. The dosage range for administration of the gamma pantol composition is that which is sufficiently large to produce the desired effect wherein the symptoms of the disease mediated by the target molecule are ameliorated. The dose should not be so large as to cause adverse side effects such as high viscosity syndrome, pulmonary edema, congestive heart failure, and other adverse side effects known in the art. In general, the dosage will vary with the age, weight, height, body surface area, health (e.g., kidney and liver function), condition, sex, and extent of the disease of the patient, and can be routinely determined by one of ordinary skill in the art. The dosage may be adjusted by the individual physician if there are any complications.
The dosage schedule and amount effective for therapeutic and prophylactic use (i.e., the "dosing regimen") will depend upon a variety of factors, including the cause, stage and severity of the disease or disorder, the health, physical state, age of the subject being treated, and the site and manner of delivery of the gamma pantol composition. The therapeutic efficacy and toxicity of the gamma pantol compositions can be determined by standard pharmaceutical, pharmacological and toxicological procedures in cell culture or experimental animals. The data obtained from these procedures can also be used to formulate a range of human dosages. In addition, the therapeutic index (i.e., the dose therapeutically effective in 50% of the population divided by the dose lethal to 50% of the population (ED50/LD50)) can be readily determined using known procedures. The dosage is preferably within a concentration range that includes the ED50 with little or no toxicity, and may vary within this range depending on the dosage form employed, the sensitivity of the patient, and the route of administration.
The dosage regimen also takes into account pharmacokinetic parameters known in the art such as drug absorption, bioavailability, metabolism and clearance (see, e.g., Hidalgo-Aragons, J.Steroid biochem. mol. biol.58:611-617 (1996); Groning et al, Pharmazie 51:337-341 (1996); Fotherby, content 54:59-69 (1996); and Johnson et al, J.Pharm. Sci.84:1144-1146 (1995)). For the clinician, determining the dosage regimen for each subject treated is well within the purview of the prior art. In addition, single or multiple doses of the γ pantol composition may be administered depending on the dosage and frequency required and tolerated by the subject. The duration of prophylactic and therapeutic treatment will vary depending on the particular disease or condition being treated. Some diseases are amenable to acute treatment, while others require long-term, chronic treatment. The gamma pantol composition may be administered sequentially or simultaneously with an additional therapeutic agent.
In some embodiments, the γ pantol composition is administered in the form of a liposome composition at a dose of between 0.005 and 5000mg of γ pantol per square meter of body surface area, or any range therebetween. In other embodiments, the γ pantol composition is administered in the form of a liposome composition at a dose of between 0.1 and 1000mg of γ pantol per square meter of body surface area, or any range therebetween.
In some embodiments, the γ PANTIFOL composition is administered in the form of an immunoconjugate composition at a dose of 1mg/kg to 500mg/kg, 1mg/kg to 250mg/kg, 1mg/kg to 200mg/kg, 1mg/kg to 150mg/kg, 1mg/kg to 100mg/kg, 1mg/kg to 50mg/kg, 1mg/kg to 25mg/kg, 1mg/kg to 20mg/kg, 1mg/kg to 15mg/kg, 1mg/kg to 10mg/kg, or 1mg/kg to 5mg/kg, or any range therebetween.
In another embodiment, the gamma pantol composition is administered in combination with one or more additional therapeutic agents.
In some embodiments, the PLp- γ pantol and/or the targeting PLp- γ pantol are prepared as an infusion composition, an injection composition, a parenteral composition, or a topical composition. In other embodiments, the injection comprises one or more of the following: intraperitoneal injection, direct intratumoral injection, intra-arterial and intravenous injection, subcutaneous injection, intramuscular injection, empirical transdermal and intranasal route delivery. In another embodiment, the PLp- γ pantol and/or the targeting PLp- γ pantol is a liquid solution or suspension. However, also provided herein are solid forms suitable for dissolution or suspension in a liquid vehicle prior to injection. In some embodiments, the targeted pegylated liposomal gamma polyglutamated antifolate composition is formulated into an enterically coated tablet or gelcap according to methods known in the art.
In some embodiments, targeted pegylated liposomal gamma polyglutamated antifolate formulations are administered to central nervous system tumors using slow, continuous intracranial infusion of liposomes directly into the tumor (e.g., Convection Enhanced Delivery (CED)). See Saito et al, Cancer Research 64: 2572-; mamot et al, J.neuro-Oncology 68:1-9 (2004). In other embodiments, the formulation is applied directly to the tissue surface. Sustained release, pH dependent release, and other specific chemical or environmental condition mediated release delivery of pegylated liposomal gamma polyglutamated antifolate formulations (e.g., depot injections and erodible implants) are also provided. Examples of such release-mediated compositions are further described herein or otherwise known in the art.
For administration by inhalation, the compositions may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas). In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount.
Where systemic delivery of the compound is desired, the compound may be formulated for parenteral administration by injection (e.g., by bolus injection or continuous infusion). Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers. Parenteral pharmaceutical formulations comprise aqueous solutions of the ingredients. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Alternatively, suspensions of liposomes can be prepared as oil-based suspensions. Suitable lipophilic solvents or vehicles include fatty oils (e.g. sesame oil) or synthetic fatty acid esters (e.g. ethyl oleate or triglycerides).
Alternatively, the non-targeted or targeted pegylated liposomal gamma polyglutamated antifolate agent may be in powder form or lyophilized form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to use.
The provided compositions (e.g., gamma polyglutamated antifolate and liposomes containing gamma polyglutamated antifolate) can also be formulated into rectal or vaginal compositions, such as, for example, suppositories or retention enemas containing conventional suppository bases such as cocoa butter or other glycerides.
Methods of use and treatment
In additional embodiments, the present disclosure provides methods of using gamma polyglutamated antifolate (gamma pantol) compositions. In some embodiments, the γ PANTIFOL composition is used to treat a disease or disorder.
In some embodiments, the present disclosure provides a method of killing a cell, the method comprising contacting the cell with a composition comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the gamma polyglutamated antifolate is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the polyglutamated antifolate agent described in section I. In some embodiments, the cell is contacted with a liposome composition comprising a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the contacted cell is a mammalian cell. In other embodiments, the contacted cell is a human cell. In some embodiments, the contacted cell is a hyperproliferative cell. In other embodiments, the hyperproliferative cell is a cancer cell. In other embodiments, the cancer cell is a primary cell or a cell from a cell line obtained/derived from a cancer selected from: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dysplasias or dyscrasias. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from colorectal cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from ovarian cancer. In some embodiments, the contacted cancer cells are primary cells or cells from a cell line obtained/derived from endometrial cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from pancreatic cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from liver cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a head and neck cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from osteosarcoma. In some embodiments, the method is performed in vivo. In other embodiments, the method is performed in vitro. In some embodiments, the gamma pantol composition contains 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the gamma pantol composition comprises a gamma pentaglutaminated antifolate agent. In some embodiments, the gamma pantol composition comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the γ pantol composition comprises a L γ polyglutamated antifolate. In some embodiments, the γ pantol composition comprises a D γ polyglutamated antifolate. In some embodiments, the γ pantol composition comprises L and D γ polyglutamated antifolate agents.
In additional embodiments, the present disclosure provides a method of killing a cell comprising contacting the cell with a liposome containing a gamma-polyglutamated antifolate (e.g., an Lp-gamma pantol, such as PLp-gamma pantol, NTLp-gamma pantol, ntplpp-gamma pantol, NTPLp-gamma pantol, or TPLp-gamma pantol disclosed herein). In some embodiments, the liposome composition comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome compositions comprise the polyglutamated antifolate agent described in section I. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the liposome is pegylated (e.g., PLp- γ pantol and NTPLp- γ pantol). In some embodiments, the liposomes comprise on their surface a targeting moiety (e.g., TLp- γ PANTIFLOL and TPLp- γ PANTIFLOL) that specifically binds to an antigen on the cell surface. In other embodiments, the liposomes are pegylated and comprise on their surface a targeting moiety that specifically binds to an antigen on the cell surface (e.g., TPLp- γ pantol). In some embodiments, the contacted cell is a mammalian cell. In other embodiments, the contacted cell is a human cell. In further embodiments, the contacted cell is a hyperproliferative cell. In other embodiments, the hyperproliferative cell is a cancer cell. In other embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a cancer selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, leukemia, and lymphoma. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from colorectal cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from ovarian cancer. In some embodiments, the contacted cancer cells are primary cells or cells from a cell line obtained/derived from endometrial cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from pancreatic cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from liver cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a head and neck cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from osteosarcoma. In some embodiments, the method is performed in vivo. In other embodiments, the method is performed in vitro. In some embodiments, the liposome contains gamma pantol, which contains 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposome comprises a L γ polyglutamated antifolate agent. In some embodiments, the liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises a D γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered liposomes comprise 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the present disclosure provides a method of killing a hyperproliferative cell, the method comprising contacting the hyperproliferative cell with a delivery vehicle (e.g., a liposome or an antibody) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the delivery vehicle comprises a γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the delivery vehicle comprises a polyglutamated antifolate agent described in section I. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the delivery vehicle is non-targeted. In other embodiments, the delivery vehicle is targeted and comprises on its surface a targeting moiety that has a specific affinity for an epitope on an antigen on the surface of a hyperproliferative cell. In other embodiments, the delivery vehicle comprises a targeting moiety having specific affinity for an epitope on an antigen on the surface of a hyperproliferative cell selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the method is performed in vivo. In some embodiments, the method is performed in vitro. In some embodiments, the delivery vehicle comprises gamma pantol consisting of 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In particular embodiments, the method of killing hyperproliferative cells is performed using a liposomal delivery vehicle comprising a gamma polyglutamated antifolate (e.g., an Lp-gamma pantol, such as PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, tlpp-gamma pantol, or TPLp-gamma pantol disclosed herein). In some embodiments, the delivery vehicle comprises a γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the delivery vehicle comprises a polyglutamated antifolate agent described in section I. In some embodiments, the delivery vehicle is a non-targeted liposome. In some embodiments, the delivery vehicle comprises on its surface a targeting moiety (e.g., TLp- γ PANTIFLOL and TPLp- γ PANTIFLOL) having specific affinity for an epitope on an antigen on the surface of a hyperproliferative cell. In some embodiments, the delivery vehicle is a liposome comprising on its surface a targeting moiety with specific affinity for an epitope on an antigen on the surface of a hyperproliferative cell. In other embodiments, the targeting moiety has specific affinity for an epitope on an antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the liposome is pegylated (e.g., PLp- γ pantol and NTPLp- γ pantol). In other embodiments, the liposomes are pegylated and comprise on their surface a targeting moiety (e.g., TPLp- γ pantol) having specific affinity for an epitope on an antigen on the surface of a hyperproliferative cell. In other embodiments, the liposome is non-pegylated. In some embodiments, the liposomes are non-pegylated and the liposomes comprise on their surface a targeting moiety (e.g., TPLp- γ pantol) having specific affinity for an epitope on an antigen on the surface of a hyperproliferative cell. In some embodiments, the liposome comprises γ pantol consisting of 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposome comprises a gamma-tetraglutamated antifolate. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the liposome comprises a L γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises a D γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In additional embodiments, the present disclosure provides a method of inhibiting proliferation of a cancer cell, the method comprising contacting the cancer cell with a delivery vehicle (e.g., a liposome or an antibody) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the delivery vehicle comprises a γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the delivery vehicle comprises a polyglutamated antifolate agent described in section I. In some embodiments, the delivery vehicle is a liposome according to any one of [12] - [67] of the detailed description section. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the delivery vehicle is non-targeted. In some embodiments, the delivery vehicle is targeted and comprises on its surface a targeting moiety having specific affinity for an epitope on an antigen on the surface of a cancer cell. In other embodiments, the delivery vehicle comprises a targeting moiety having specific affinity for an epitope on a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, Pcadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin receptor, HER2, HER 72, EGFR, IGFR-1, CD RvIII, CD3, CD, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and cala. In some embodiments, the delivery vehicle is an antibody having specific affinity for an epitope on an antigen on the surface of a cancer cell. In some embodiments, the contacted cancer cell is a mammalian cell. In other embodiments, the contacted cancer cell is a human cell. In further embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a cancer selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, leukemia, and lymphoma. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from colorectal cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from ovarian cancer. In some embodiments, the contacted cancer cells are primary cells or cells from a cell line obtained/derived from endometrial cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from pancreatic cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from liver cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a head and neck cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from osteosarcoma. In some embodiments, the method is performed in vivo. In some embodiments, the method is performed in vitro. In some embodiments, the delivery vehicle is an antibody having specific affinity for an epitope on one of the cell surface antigens listed above. In other embodiments, the targeting vehicle is a liposome comprising a targeting moiety having specific affinity for an epitope on the surface of a cancer cell. In other embodiments, the targeting vehicle is a liposome comprising a targeting moiety having specific affinity for an epitope on one of the cell surface antigens listed above. In some embodiments, the delivery vehicle is a pegylated liposome. In other embodiments, the delivery vehicle is a non-pegylated liposome. In some embodiments, the delivery vehicle comprises a gamma pantol composition containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In other embodiments, the present disclosure provides a method of inhibiting proliferation of a cancer cell, the method comprising contacting the cancer cell with a liposome comprising a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein). In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome comprises a polyglutamated antifolate described in section I. In some embodiments, the liposome is any one of [12] to [67] according to the detailed description. In some embodiments, the liposome is non-targeted. In some embodiments, the liposome is targeted and comprises on its surface a targeting moiety having specific affinity for an epitope on an antigen on the surface of a cancer cell. In other embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope on a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the contacted cancer cell is a mammalian cell. In other embodiments, the contacted cancer cell is a human cell. In further embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a cancer selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, leukemia, and lymphoma. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from colorectal cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from ovarian cancer. In some embodiments, the contacted cancer cells are primary cells or cells from a cell line obtained/derived from endometrial cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from pancreatic cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from liver cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from a head and neck cancer. In some embodiments, the contacted cancer cell is a primary cell or a cell from a cell line obtained/derived from osteosarcoma. In some embodiments, the method is performed in vivo. In some embodiments, the method is performed in vitro. In other embodiments, the targeting vehicle is a liposome comprising a targeting moiety having specific affinity for an epitope on one of the cell surface antigens listed above. In some embodiments, the liposome is pegylated. In other embodiments, the liposome is non-pegylated. In some embodiments, the liposome comprises γ pantol consisting of 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposome comprises a gamma-tetraglutamated antifolate. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the liposome comprises a L γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises a D γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In additional embodiments, the present disclosure provides a method for treating a hyperproliferative disorder, comprising administering to a subject having or at risk of having a hyperproliferative disorder an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the administered delivery vehicle comprises a targeting moiety with specific affinity for an epitope of an antigen on the surface of the hyperproliferative cell. In further embodiments, the delivery vehicle comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the administered delivery vehicle is free of a targeting moiety having specific affinity for an epitope on a cell surface antigen of the hyperproliferative cell. In some embodiments, the delivery vehicle comprises gamma pantol consisting of 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the delivery vehicle comprises a γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the delivery vehicle comprises a polyglutamated antifolate agent described in section I. In some embodiments, the delivery vehicle is a liposome according to any one of [12] - [67] of the detailed description section. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the hyperproliferative disorder is cancer. In some embodiments, the hyperproliferative disorder is an autoimmune disease (e.g., rheumatoid arthritis). In some embodiments, the hyperproliferative disorder is a benign or malignant tumor; leukemia, malignant diseases of the blood system or lymphatic system. In other embodiments, the hyperproliferative disorder is selected from the group consisting of neuronal disorders, glial disorders, astrocytic disorders, hypothalamic disorders, glandular disorders, macrophage disorders, epithelial disorders, interstitial disorders, blastocoel disorders, inflammatory disorders, angiogenesis, and immune disorders, including autoimmune diseases.
In additional embodiments, the present disclosure provides a method for treating a hyperproliferative disorder, comprising administering to a subject having or at risk of having a hyperproliferative disorder an effective amount of liposomes comprising a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, such as PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol or TPLp-gamma pantol). In some embodiments, the liposome is pegylated. In some embodiments, the liposome is non-pegylated. In additional embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of a hyperproliferative cell. In further embodiments, the liposome comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the liposome is free of a targeting moiety having specific affinity for an epitope on a cell surface antigen of a hyperproliferative cell. In some embodiments, the liposome comprises γ pantol consisting of 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposome comprises a gamma-tetraglutamated antifolate. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the delivery vehicle comprises a polyglutamated antifolate agent described in section I. In some embodiments, the delivery vehicle is a liposome according to any one of [12] - [67] of the detailed description section. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises a D γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the hyperproliferative disorder is cancer. In some embodiments, the hyperproliferative disorder is an autoimmune disease (e.g., rheumatoid arthritis). In some embodiments, the hyperproliferative disorder is a benign or malignant tumor; leukemia, malignant diseases of the blood system or lymphatic system. In other embodiments, the hyperproliferative disorder is selected from the group consisting of neuronal disorders, glial disorders, astrocytic disorders, hypothalamic disorders, glandular disorders, macrophage disorders, epithelial disorders, interstitial disorders, blastocoel disorders, inflammatory disorders, angiogenesis, and immune disorders, including autoimmune diseases.
Exemplary hyperproliferative disorders that can be treated according to the disclosed methods include, but are not limited to, disorders associated with benign, premalignant, and malignant cell proliferation, including, but not limited to, neoplasms and tumors (e.g., histiocytoma, glioma, astrocytoma, osteoma), cancer (e.g., lung cancer, small cell lung cancer, gastrointestinal cancer, intestinal cancer, colorectal cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, liver cancer, kidney cancer, bladder cancer, pancreatic cancer, brain cancer, sarcomas (e.g., osteosarcoma, kaposi's sarcoma), and melanoma), leukemia, psoriasis, bone disease, fibroproliferative disorders (e.g., fibroproliferative disorders of connective tissue), and atherosclerosis.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the administered delivery vehicle comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of the cancer cell. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the cancer is selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies (e.g., leukemia or lymphoma). In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma.
In additional embodiments, the present disclosure provides a method for treating, reducing, or inhibiting metastasis, the method comprising administering to a subject having or at risk of having cancer an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the disclosed methods provide, inter alia: (1) reducing or inhibiting the growth, proliferation, survival, migration or invasion of a primary tumor, cancer or neoplasia; (2) reducing or inhibiting the growth, proliferation, survival, migration, or invasion of a primary tumor, cancer, or neoplasia that is likely to or does develop metastasis; (3) reducing or inhibiting the formation or establishment of metastasis, caused by the primary tumor, cancer or neoplasia, to one or more other sites, locations, regions or systems distinct from the primary tumor, cancer or neoplasia; (4) reducing or inhibiting the growth or proliferation of metastases at one or more other sites, locations, regions or systems distinct from the primary tumor, cancer or neoplasia after the metastases have formed or established; and/or (5) reduce or inhibit the formation or establishment of additional metastases after the metastases have been formed or established. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the administered delivery vehicle comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of the cancer cell. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the cancer is selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies (e.g., leukemia or lymphoma). In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the administered delivery vehicle comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of the cancer cell. In further embodiments, the delivery vehicle comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the cancer is selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies (e.g., leukemia or lymphoma). In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of liposomes comprising a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, such as PLp-gamma pantol, NTLp-gamma pantol, NTPLp-gamma pantol, TLp-gamma pantol or TPLp-gamma pantol). In some embodiments, the liposome is pegylated. In some embodiments, the liposome is non-pegylated. In additional embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of a cancer cell. In further embodiments, the liposome comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the cancer is selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies (e.g., leukemia or lymphoma). In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the liposome comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposome comprises a gamma-tetraglutamated antifolate. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the liposome comprises a L γ polyglutamated antifolate agent. In some embodiments, the liposome comprises γ pantol containing 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 γ -glutamyl groups in the form of L. In some embodiments, the liposome comprises a D γ polyglutamated antifolate agent. In some embodiments, the liposome comprises γ pantol containing 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 γ -glutamyl groups in D form. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the liposome comprises γ pantol containing 2, 3, 4, 5, or more than 5 γ -glutamyl groups in L form and 1, 2, 3, 4, 5, or more than 5 γ -glutamyl groups in D form. In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposome composition comprising liposomes comprising a gamma polyglutamated antifolate and a targeting moiety having specific affinity for an epitope of an antigen on the surface of the cancer. In some embodiments, the liposome comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, Pcadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin receptor, HER2, HER 72, EGFR, IGFR-1, CD RvIII, CD3, CD, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and cala. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., TPLp- γ pantol). In some embodiments, the administered liposome composition comprises non-pegylated liposomes. In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposomes of the liposome composition comprise gamma pantol containing 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 gamma-glutamyl groups in the form of L. In some embodiments, the liposomes of the liposome composition comprise a D γ polyglutamated antifolate. In some embodiments, the liposomes of the liposome composition comprise gamma pantol containing 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 gamma-glutamyl groups in D form. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the liposomes of the liposome composition comprise γ pantol containing 2, 3, 4, 5, or more than 5 γ -glutamyl groups in L form and 1, 2, 3, 4, 5, or more than 5 γ -glutamyl groups in D form.
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of a Tumor Specific Antigen (TSA) or Tumor Associated Antigen (TAA). In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen selected from the group consisting of: tumor differentiation antigens (e.g., MART1/Melana, GP100(Pmel 17), tyrosinase, TRP1 and TRP2), tumor specific multiple lineage antigens (e.g., MAGE1, MAGE3, BAGE, GAGE1, GAGE2 and p15), overexpressed embryonic antigens (e.g., carcinoembryonic antigen (CEA)), overexpressed oncogenes or mutated tumor suppressor gene products (e.g., p53, Ras and HER2/neu), unique tumor antigens resulting from chromosomal translocations (e.g., BCR-ABL, E2A-PRL, H4-RET, IGH-IGK and MYL-RAR), viral antigens (e.g., Epstein Barr virus antigen EBVA, Human Papilloma Virus (HPV) antigen E6 or E7), GP 100), Prostatic Acid Phosphatase (PAP), Prostate Specific Antigen (PSA), PTGER4, CD 4, EBVA 4662, CD52, EVGA 69-CD 4624, CD 465, CXCR 24, CD 39361, CD 3946, CD 24, CD 2I, SLC39A8, MICB, LRRC70, CLELC2B, HMHA1, LST1, and CMTM6 CKLFSF 6). In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome is Lp- γ pantol according to any one of [48] - [67] of the specific embodiment.
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of a hematologic tumor antigen. In other embodiments, the targeting moiety has specific affinity for an epitope of a hematologic tumor antigen selected from the group consisting of: CD19, CD20, CD22, CD30, CD138, CD33, CD34, CD38, CD123, CS1, ROR1, lewis y, Ig kappa light chain, TCR, BCMA, TACI, BAFFR (CD268), cala, and NKG2DL ligands. In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of a B cell lymphoma specific idiotype immunoglobulin or B cell differentiation antigen (e.g., CD19, CD20, and CD 37). In some embodiments, the liposome comprises a targeting moiety with specific affinity for an epitope of an antigen on multiple myeloma cells (e.g., CS-1, CD38, CD138, MUC1, HM1.24, CYP1B1, SP17, PRAME, wilms tumor 1WT1), and heat shock protein gp96) or on myeloid cells (e.g., TSLPR and IL-7R). In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome is Lp- γ pantol according to any one of [48] - [67] of the specific embodiment.
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of a solid tumor antigen. In other embodiments, the targeting moiety has specific affinity for an epitope of a hematologic tumor antigen selected from the group consisting of: disialoganglioside (GD2), o-acetyl GD2, EGFRvIII, ErbB2, VEGFR2, FAP, mesothelin, IL13Ra2 (glioma), cMET, PSMA, L1CAM, CEA, and EGFR. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome is Lp- γ pantol according to any one of [48] - [67] of the specific embodiment.
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-gamma, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin receptor, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD2, CD3, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin γ v β 3, γ v β 5, or γ v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and cala. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome is Lp- γ pantol according to any one of [48] - [67] of the specific embodiment.
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen selected from the group consisting of: CD137, PDL, CTLA, CD, KIR, TNFRSF10 (DR), TIM, PD, cMet, glycolipid F, EGFRvIII, HLAA (NY-ESO-1), LAG, CD134 (OX), HVEM, BTLA, TNFRSF (DR), CD133, MAGE A, PSCA, MUC, CD44 v/7, CD44 v/8, IL11, ephA, CAIX, MNCAIX, CSPG, MUC, EPCAM (EGP), TAG, EGP, ErbB receptor family, ErbB (HER), ErbB/4, RAGE, GD, FAR, LewisY, NCAM, HLAA/MAGE, MAGEA, MAGE-A, B7H, ErbB, Melana (MART), HPV E, thyroglobulin, tyrosinase, PSA, CLL1GD, Ag, PRT, Tn, FLEA, CD 7H, PDG, PGF, PGM, PDG, PGM, PGF, PGM, PGF, PGM, GPRC5D, CXORF61, CD97, CD7a, HLE, CD179a, ALK, polysialic acid, PLAC1, Globoh, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E K, TARP, E1K, legumain, E K, ETV K-AML, sperm protein 17, XAGE K, Tie 2, MAD-CT K, Fos-related antigen 1, p K mutant, prostate specific protein, survivin, telomerase, PCTA K (galectin 8), Ras mutant, hTERT, sarcoma translocation, ML-IAP, ERG (TMPRSS fusion gene), NA K, PAX K, PAFCB receptor, cyclin B K, BOLRCN 72, BOMUC 7-IRU K, SARCE K, SLEC K, CD K, TRPC K, TROP K, SLEC K, TROP 72, SLEC K, TRPC K, SLEC K, TROP 72, SLEC K, TROP K, SLEC K, TROP 72, SL, EMR2, LY75, GPC3, FCRLS, IGLL1, TSP-180, MAGE1, VEGFR1, IGF 11, hepatocyte growth factor receptor, p185ErbB 1, p180ErbB-3, nm-23H1, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, β -catenin, CDK 1, Mum1, p1, 43-9F, 5T 1, 791Tgp 1, β -human chorionic gonadotropin, SDCA 225, BTA, CA125, CA 1-3, CA 27.29(BCAA), CA195, CA242, CA-50, CA1, CD1, CO-369, BCA 250, HTMA gp 175, M344, MG 72, MOVAV 1, MOVAP-72, AFP 1, TAPS 1, and related lectin 1. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome is Lp- γ pantol according to any one of [48] - [67] of the specific embodiment.
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen selected from the group consisting of: PDGF, VEGFR, neuropilin 1 (NRP), neuropilin 2 (NRP), betacellulin, PLGF, RET (transfection rearrangement), TIE (TEK), CA125, CD CD, CD32, CD (e.g., CD44 v), CD49 (integrin gamma 5), CD (ICAM), CD200, CD147, CD166, CD200, ESA, SHH, DHH, WNH, patched 1 (PTCH), Smoothened (SMO), WNT, IHT 2, WNT3, KRT, FZD, ZD, JaWNT 5, WNT7, WNT8, WNT10, EAT 16, LKP, Dc, TNFR, TNFRSF (TNFRSF) TNFRSF11, TNFRSF (TNFRSF) TNFRSF, TNFRSF (TNFRSF) and TNFRSF11, TNFRSF (TROY), TNFRSF (DR), ILIRI, 1L1R, IL2, IL5, IL6, 1L8, IL10, IL12, IL13, IL15, IL18, IL19, IL21, IL23, REGIV, FGFR, ALK, ALCAM, Axl, TGFb, TGFBR, IGFIR, BMPRI, N-cadherin, E-cadherin, VE-cadherin, ganglioside GM, ganglioside GD, PSGR, DCC, CDCP, CXCR, CCR, SEALAN 1, SEALAN 2, SEALAN 3, SEALAN 4, TMF, neuregulin, MCSF, CSF, CSFR (fms), GCSF, SFAM, BCBCCA, HLA-BRDR, SMCC, VLCC, VLCA, LYCA, CAGR, CANV, CAGR, and CAGR, and CAGR, Leukotriene B4 receptor (LTB4R), neurotensin NT receptor (NTR), 5T4 carcinoembryonic antigen, tenascin C, MMP2, MMP7, MMP9, MMP12, MMP14, MMP26, cathepsin G, SULF1, SULF2, MET, CA9, TM4SF1, syndecan (SDCl), ephrin B4, TEM1, TGF β 1 and TGFBRII. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome is Lp- γ pantol according to any one of [48] - [67] of the specific embodiment.
In some embodiments, the liposomes comprise a targeting moiety that has specific affinity for an epitope of an antigen associated with an immune system disorder (e.g., autoimmune and inflammatory disorders), or is associated with modulating an immune response. In some embodiments, the targeting moiety has specific affinity for an epitope of a cell surface antigen expressed on the surface of a macrophage (expressing CD 44).
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an immunosuppressive target. In another embodiment, AD is an epitope of an immunosuppressive target selected from: IL1Ra, IL6R, CD26L, CD28, CD80, Fc γ RIIB. In another embodiment, the AD in the adaptor is an epitope of an immunostimulatory target selected from the group consisting of: CD25, CD28, CTLA4, PD1, B7H1(PDL1), B7H4 TGF β, TNFRSF4(OX40), TNFRSF5(CD40), TNFRSF9(41BB, CD137), TNFRSF14(HVEM), TNFRSF25(DR3) and TNFRSF18 (GITR).
In some embodiments, the liposome comprises a targeting moiety having specific affinity for an epitope of an antigen selected from the group consisting of: IL1Rb, C3AR, C5AR, CXCR1, CXCR2, CCR1, CCR3, CCR7, CCR8, CCR9, CCR10, ChemR23, MPL, GP130, TLR2, TLR3, TLR4, TLR5, TLR7, TREM 7, CD49 7 (integrin gamma 1), integrin a5B 7, gamma 4 protein subunit, A4B7 integrin, cathepsin 7 (LTBR), TNFRSF7 (Fas, CD 7), TNFRSF6 7 (DcCSF), TNFRSF7(CD 7), TNFRSF11 7 (RANK), TNFRSF7 (NGFR 7), TNFRSF19 (RELT), TNFRSF7 (TNFRSF 7), TNFRSF7, TNFRFSF 7, TNFRSF 363672, TNFRSF7, TNFRSF 3636363636363672, TNFRSF7, TNFRSF 36363636363672, TNFRSF 3636363636363672, TNFRSF7, TNFRSF 36363672, TNFRSF 363636363636363672, TNFRSF7, TNFSK 363672, TNFRSF7, TNFRSF 3636363672, TNFRSF7, TNFRSF 363636363636363672, TNFRSF7, TNFRSF 3636363672, TNFRSF7, TNFRSF 36363672, TNFRSF 36363636363636363636363636363636363636363672, TNFRSF 36, CD2, CD4, CD11a, CD18, CD30, CD40, CD86, CXCR3, CCR2, CCR4, CCR5, CCR8, RhD, IgE and Rh.
In a further embodiment, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having a cancer that expresses a folate receptor on the cell surface thereof an effective amount of a liposomal composition, wherein the liposomal composition comprises liposomes comprising (a) a gamma polyglutamated antifolate (γ pantol) and (b) a targeting moiety having specific binding affinity for a folate receptor. In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and/or folate receptor (FR-). In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and/or folate receptor (FR-). In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha) and folate receptor beta (FR-beta). In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., TPLp- γ pantol). In some embodiments, the administered liposome composition comprises non-pegylated liposomes. In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposomes of the liposome composition comprise gamma pantol containing 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 gamma-glutamyl groups in D form. In some embodiments, the liposomes of the liposome composition comprise L and D γ polyglutamated antifolate agents. In some embodiments, the liposomes of the liposome composition comprise γ pantol containing 2, 3, 4, 5, or more than 5 γ -glutamyl groups in L form and 1, 2, 3, 4, 5, or more than 5 γ -glutamyl groups in D form. In some embodiments, the liposomes of the liposome composition comprise a tetraglutamated antifolate. In some embodiments, the liposomes of the liposome composition comprise a pentaglutamated antifolate agent. In some embodiments, the liposomes of the liposome composition comprise a hexaglutaminated antifolate.
In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the liposome composition is administered to treat epithelial tissue malignancies. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the liposome composition is administered to treat colorectal cancer. In some embodiments, the liposome composition is administered to treat ovarian cancer. In some embodiments, the liposome composition is administered to treat endometrial cancer. In some embodiments, the liposome composition is administered to treat pancreatic cancer. In some embodiments, the liposome composition is administered to treat liver cancer. In some embodiments, the liposome composition is administered to treat head and neck cancer. In some embodiments, the liposome composition is administered to treat osteosarcoma.
In some embodiments, the present disclosure provides a method for treating lung cancer (e.g., non-small cell lung cancer), the method comprising administering to a subject having or at risk of having lung cancer an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In certain embodiments, the cancer is non-small cell lung cancer. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the delivery vehicle comprises on its surface a targeting moiety having specific affinity for an epitope on an antigen on the surface of a lung cancer (e.g., non-small cell lung cancer) cell. In other embodiments, the delivery vehicle comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: mucin 1, bindin 4, NaPi2b, CD56, EGFR and SC-16. In some embodiments, the targeting moiety is an antibody or a fragment of an antibody. In further embodiments, the delivery vehicle is a liposome and the liposome comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: mucin 1, bindin 4, NaPi2b, CD56, EGFR and SC-16. In other embodiments, the delivery vehicle is a pegylated liposome comprising a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: mucin 1, bindin 4, NaPi2b, CD56, EGFR and SC-16. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent.
In some embodiments, the present disclosure provides a method for treating pancreatic cancer, the method comprising administering to a subject having or at risk of having pancreatic cancer an effective amount of a delivery vehicle (e.g., an Antibody (ADC) or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein). In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the delivery vehicle comprises on its surface a targeting moiety having specific affinity for an epitope on an antigen on the surface of a pancreatic cancer cell. In other embodiments, the delivery vehicle comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: tactd 2(TROP2), mucin 1, mesothelin, guanylate cyclase c (gcc), SLC44a4, and bindin 4. In other embodiments, the delivery vehicle is a liposome and the liposome comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: tactd 2(TROP2), mucin 1, mesothelin, guanylate cyclase c (gcc), SLC44a4, and bindin 4. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In additional embodiments, the present disclosure provides a method for treating breast cancer (e.g., triple negative breast cancer (estrogen receptor-, progestin receptor-and HER2)) comprising administering to a subject having or at risk of having breast cancer an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the administered delivery vehicle is a liposome comprising a gamma polyglutamated antifolate agent. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In a further embodiment, the delivery vehicle comprises on its surface a targeting moiety having specific affinity for an epitope on an antigen on the surface of a breast cancer cell. In other embodiments, the delivery vehicle comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: LIV-1(ZIP6), EGFR, HER2, HER3, mucin 1, gon mb, and bindin 4. In some embodiments, the targeting moiety is an antibody or a fragment of an antibody. In further embodiments, the delivery vehicle is a liposome and the liposome comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: LIV-1(ZIP6), EGFR, HER2, HER3, mucin 1, gon mb, and bindin 4. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the present disclosure provides a method for treating a hematologic cancer, the method comprising administering to a subject having or at risk of having a hematologic cancer an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein). In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the delivery vehicle comprises on its surface a targeting moiety having specific affinity for an epitope on an antigen on the surface of a hematologic cancer cell. In other embodiments, the delivery vehicle comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: CD30, CD79b, CD19, CD138, CD74, CD37, CD19, CD22, CD33, CD34, and CD 98. In other embodiments, the delivery vehicle is a liposome and the liposome comprises a targeting moiety having specific affinity for an epitope on an antigen selected from the group consisting of: CD30, CD79b, CD19, CD138, CD74, CD37, CD19, CD22, CD33, CD34, and CD 98. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the present disclosure provides a method for treating a subject having or at risk of having a cancer that is distinguishable by the expression of an antigen on the cell surface thereof. Thus, in some embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a targeting moiety having specific affinity for an epitope on a surface antigen of the cancer and a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the administered delivery vehicle comprises a γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered delivery vehicle comprises a polyglutamated antifolate agent described in section I. In some embodiments, the delivery vehicle administered is a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the targeting moiety is an antibody or a fragment of an antibody. In further embodiments, the delivery vehicle is a liposome. In some embodiments, the administered delivery vehicle comprises gamma pantol consisting of 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the disclosed compositions (e.g., liposomes containing gamma polyglutamated antifolates) are administered to a subject having, or at risk of having, a cancer, a solid tumor and/or a metastasis which can be distinguished by the expression of a tumor-specific antigen or a tumor-associated antigen on the cell surface thereof. Thus, in some embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer, a solid tumor and/or metastasis, which cancer, solid tumor and/or metastasis is distinguishable by expression of a tumor-specific antigen or a tumor-associated antigen on its cell surface, and wherein the targeting moiety has specific binding affinity for an epitope on the tumor-specific antigen or tumor-associated antigen, an effective amount of a delivery vehicle (e.g., a liposome) comprising a targeting moiety and a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein). In some embodiments, the delivery vehicle administered is a liposome. In other embodiments, the liposome is pegylated. In further embodiments, the delivery vehicle comprises a targeting moiety having specific affinity for an epitope on a cell surface antigen expressed on the surface of a cancer, solid tumor, and/or metastatic cell. For example, folate receptor- α, folate receptor- β or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, bindin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA 7 (carbonic anhydrase), 5T 7 (TPBG), SLTRK 7, SC-16, tissue factor, LIV-1(ZIP 7), CGEN-15027, Pcadherin, fibronectin exodomain B (ED-B), VEGFR 7 (CD309), tenascin, collagen IV, periostin, endothelin receptor, HER 7, EGFR, EGFRvIII, FGFR 7, CD 36ZD 7, CD ZD7, CD 36ZD 7, CD 36ZD 72, CD7, CD 36ZD 7, CD 36ZD 7, CD7, CD34, CD37, CD38, CD40, CD79 40, CD105, CD133, CD138, cripto, IGF-1 40-2 40, EphA receptor, EphB receptor, EphA 40, EphB 40, integrin (e.g., integrin α v β 3, α v β 5 or UFα v β 6), ApoC 242 antigen, Apo 40, PSSK, NGEP, PSCA, EFTMF 40, endoglin, endothelial glycoprotein, CANA, CAAg, TrkC-VEGFR, CALb-1, VEGFRK-1, PDGFRK, TkD-2, PDGFRK, PGFR, PDGFR, PGFR, and PGFR. In some embodiments, the delivery vehicle comprises a targeting moiety with specific affinity for an epitope on one or more cell surface antigens (such as neoantigens) derived from or determined to be expressed on a cancer (tumor) in a particular subject. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the delivery vehicle comprises γ pantol containing 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 γ -glutamyl groups in the L form. In some embodiments, the delivery vehicle comprises a γ pantol containing 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 γ -glutamyl groups in L form. In some embodiments, the delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the delivery vehicle comprises a γ pantol containing 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 γ -glutamyl groups in D form. In some embodiments, the delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the delivery vehicle comprises γ pantol containing 2, 3, 4, 5, or more than 5 γ -glutamyl groups in L form, and 1, 2, 3, 4, 5, or more than 5 γ -glutamyl groups in D form. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent.
In further embodiments, the targeting moiety has specific affinity for an epitope on an antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin, HER2, HER3, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA.
In other embodiments, the delivery vehicle is a liposome and the liposome comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin receptor, HER2, HER 72, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposome comprises a gamma-pentaglutaminated antifolate. In other embodiments, the liposome comprises a gamma-hexaglutaminated antifolate. In some embodiments, the liposome comprises γ pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome is Lp- γ pantol according to any one of [48] - [67] of the specific embodiment. In some embodiments, the liposome comprises a L γ polyglutamated antifolate agent. In some embodiments, the liposome comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the liposome comprises a D γ polyglutamated antifolate agent. In some embodiments, the liposome comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In other embodiments, the present disclosure provides a method for treating cancer comprising administering to a subject having or at risk of having cancer containing cells expressing a folate receptor on their cell surface an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising on its surface a targeting moiety that specifically binds to a folate receptor and a gamma polyglutamated antifolate (e.g., gamma pantol as disclosed herein). In some embodiments, the targeting moiety is an antibody or an antigen-binding fragment of an antibody. In other embodiments, the targeting moiety has specific affinity for folate receptor alpha, folate receptor beta, or folate receptor. As disclosed herein, folate receptor-targeted pegylated liposomes containing gamma polyglutamated antifolates are capable of delivering large amounts of gamma polyglutamated antifolates to cancer cells, and in particular cancer cells expressing folate receptors, as compared to normal cells (i.e., cells distinct from cancer cells do not actively uptake the liposomes, and/or do not express folate receptors). Any cancer that expresses a folate receptor can be treated according to the disclosed methods. It should be noted that some cancers may express the folate receptor at an early stage, while most cancers may express the folate receptor at a late stage. In some embodiments, the delivery vehicle administered is a liposome. In other embodiments, the liposome is pegylated. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In additional embodiments, the present disclosure provides a method for cancer maintenance therapy, the method comprising administering to a subject undergoing or having undergone cancer therapy an effective amount of a liposome composition comprising liposomes containing a gamma polyglutamated antifolate (e.g., gamma pantol, disclosed herein). In some embodiments, the liposome composition administered is PLp- γ pantol, NTLp- γ pantol, NTPLp- γ pantol, TLp- γ pantol or TPLp- γ pantol. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the administered liposome composition comprises a targeting moiety (e.g., TLp- γ PANTIFLOL or TPLp- γ PANTIFLOL) having specific affinity for an epitope on a surface antigen of the cancer cell. In some embodiments, the administered liposome composition comprises a liposome (e.g., TPLp- γ pantol) that is pegylated and comprises a targeting moiety. In some embodiments, the administered liposome composition comprises targeted liposomes and liposomes that do not comprise a targeting moiety (e.g., non-targeted). In some embodiments, the administered liposome composition comprises pegylated and non-pegylated liposomes. In some embodiments, the liposomes of the administered liposome composition comprise a gamma polyglutamated antifolate containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the administered liposome composition comprises a gamma hexaglutaminated antifolate. In some embodiments, the administered liposome composition comprises a L γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises a L γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises a D γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the cancer treated by one or more of the methods disclosed herein is a solid tumor lymphoma. Examples of solid tumor lymphomas include hodgkin lymphoma, non-hodgkin lymphoma, and B-cell lymphoma.
In some embodiments, the cancer treated by one or more methods disclosed herein is bone cancer, brain cancer, breast cancer, colorectal cancer, connective tissue cancer, cancer of the digestive system, endometrial cancer, esophageal cancer, eye cancer, head and neck cancer, gastric cancer, intraepithelial tumors, melanoma neuroblastoma, non-hodgkin's lymphoma, non-small cell lung cancer, prostate cancer, retinoblastoma, or rhabdomyosarcoma.
In some embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a composition comprising a delivery vehicle and a gamma polyglutamated antifolate. In some embodiments, the administered composition comprises a pegylated delivery vehicle. In some embodiments, the administered composition comprises a targeting moiety that has a specific affinity for an epitope of an antigen on the surface of a target cell of interest (e.g., a cancer cell). In some embodiments, the delivery vehicle comprises an antibody or antigen-binding antibody fragment. In some embodiments, the composition is administered to treat a cancer selected from: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, renal cancer, cholangiocarcinoma, gallbladder cancer, bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, leukemia, and lymphoma. In some embodiments, the composition is administered to treat a cancer selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma.
In some embodiments, the administered composition contains 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle is an immunoconjugate. In some embodiments, the administered delivery vehicle comprises a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered delivery vehicle comprises a polyglutamate of the antifolate agent disclosed in section I herein. In some embodiments, the delivery vehicle administered is a liposome. In some embodiments, the delivery vehicle administered is a liposome according to any one of [12] to [67] of the detailed description section.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposome composition comprising liposomes containing a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, TLp-gamma pantol or TPLp-gamma pantol). In some embodiments, the liposome compositions comprise a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposomal composition comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, renal cancer, cholangiocarcinoma, gallbladder cancer, bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, leukemia, and lymphoma. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the liposome composition is administered to treat colorectal cancer. In some embodiments, the liposome composition is administered to treat ovarian cancer. In some embodiments, the liposome composition is administered to treat endometrial cancer. In some embodiments, the liposome composition is administered to treat pancreatic cancer. In some embodiments, the liposome composition is administered to treat liver cancer. In some embodiments, the liposome composition is administered to treat head and neck cancer. In some embodiments, the liposome composition is administered to treat osteosarcoma. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposome composition comprising targeted liposomes (e.g., TLp- γ pantol or TPLp- γ pantol), wherein the liposome composition comprises liposomes comprising a gamma polyglutamated antifolate (Lp- γ pantol) and further comprising a targeting moiety having specific affinity for a surface antigen (epitope) on the cancer. In some embodiments, the liposome compositions comprise a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposomal composition comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the liposome composition comprises a liposome according to any one of [12] to [67] of the detailed description section. In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposome composition comprising liposomes containing a gamma polyglutamated antifolate (e.g., Lp-gamma pantol, PLp-gamma pantol, NTLp-gamma pantol, TLp-gamma pantol or TPLp-gamma pantol). In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, renal cancer, cholangiocarcinoma, gallbladder cancer, bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, leukemia, and lymphoma. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the liposome composition is administered to treat colorectal cancer. In some embodiments, the liposome composition is administered to treat ovarian cancer. In some embodiments, the liposome composition is administered to treat endometrial cancer. In some embodiments, the liposome composition is administered to treat pancreatic cancer. In some embodiments, the liposome composition is administered to treat liver cancer. In some embodiments, the liposome composition is administered to treat head and neck cancer. In some embodiments, the liposome composition is administered to treat osteosarcoma.
In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the administered liposome composition comprises a L γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises a D γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In additional embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of a liposome composition comprising targeted liposomes (e.g., TLp- γ pantol or TPLp- γ pantol), wherein the liposome composition comprises liposomes comprising a gamma polyglutamated antifolate (Lp- γ pantol) and further comprising a targeting moiety having specific affinity for a surface antigen (epitope) on the cancer. In some embodiments, the liposome compositions comprise a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the γ pantol is a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the liposome composition comprises a liposome according to any one of [48] - [67] of the detailed description section. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, renal cancer, cholangiocarcinoma, gallbladder cancer, bladder cancer, sarcoma (e.g., osteosarcoma), brain cancer, central nervous system cancer, melanoma, myeloma, leukemia, and lymphoma. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the liposome composition is administered to treat colorectal cancer. In some embodiments, the liposome composition is administered to treat ovarian cancer. In some embodiments, the liposome composition is administered to treat endometrial cancer. In some embodiments, the liposome composition is administered to treat pancreatic cancer. In some embodiments, the liposome composition is administered to treat liver cancer. In some embodiments, the liposome composition is administered to treat head and neck cancer. In some embodiments, the liposome composition is administered to treat osteosarcoma. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., PLp- γ pantol, NTPLp- γ pantol, or TPLp- γ pantol). In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the administered liposome composition comprises a L γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises a D γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In other embodiments, the present disclosure provides a method for treating cancer, the method comprising administering to a subject having or at risk of having a cancer that expresses a folate receptor on the cell surface thereof an effective amount of a liposome composition containing targeted liposomes (e.g., TLp- γ pantol or TPLp- γ pantol), wherein the liposome composition comprises liposomes comprising (a) a γ -polyglutamated antifolate (γ pantol) and (b) a targeting moiety having specific binding affinity for a folate receptor. In some embodiments, the administered liposome composition comprises pegylated liposomes (e.g., TPLp- γ pantol). In some embodiments, the liposome compositions comprise a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the polyglutamated antifolate is a polyglutamate of an antifolate described in section I herein. In some embodiments, the liposome composition comprises a liposome according to any one of [48] - [67] of the detailed description section. In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and/or folate receptor (FR-). In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and/or folate receptor (FR-). In some embodiments, the targeting moiety has specific binding affinity for folate receptor alpha (FR-alpha) and folate receptor beta (FR-beta). In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies. In some embodiments, the liposome composition is administered to treat a cancer selected from the group consisting of: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) cancer. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the composition is administered to treat colorectal cancer. In some embodiments, the composition is administered to treat ovarian cancer. In some embodiments, the composition is administered to treat endometrial cancer. In some embodiments, the composition is administered to treat pancreatic cancer. In some embodiments, the composition is administered to treat liver cancer. In some embodiments, the composition is administered to treat head and neck cancer. In some embodiments, the composition is administered to treat osteosarcoma. In some embodiments, the liposomes of the administered liposome composition comprise gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-tetraglutamated antifolate. In some embodiments, the liposomes of the administered liposome composition comprise a gamma-pentaglutaminated antifolate. In other embodiments, the liposomes of the administered liposome composition comprise a gamma hexaglutaminated antifolate. In some embodiments, the administered liposome composition comprises a L γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises a D γ polyglutamated antifolate. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered liposome composition comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered liposome composition comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the present disclosure provides a method for treating an immune system disorder (e.g., autoimmune diseases, such as inflammation and rheumatoid arthritis), the method comprising administering to a subject having or at risk of having an immune system disorder an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma pantol disclosed herein). In some embodiments, the autoimmune disease is rheumatoid arthritis. In some embodiments, the delivery vehicle comprises a gamma polyglutamated antifolate agent according to any one of [1] to [11] of the detailed description section. In some embodiments, the polyglutamated antifolate is a polyglutamate of an antifolate described in section I herein. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the delivery vehicle is a liposome according to any one of [12] - [67] of the detailed description section. In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the administered delivery vehicle comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of an immune cell associated with an immune system disorder. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the present disclosure provides a method for treating an infectious disease (e.g., HIV, malaria, and schistosomiasis), the method comprising administering to a subject having or at risk of having the infectious disease an effective amount of a delivery vehicle (e.g., an antibody or liposome) comprising a gamma polyglutamated antifolate (e.g., gamma panto dol as disclosed herein). In some embodiments, the delivery vehicle comprises a gamma polyglutamated antifolate agent according to any one of [1] to [11] of the detailed description section. In some embodiments, the polyglutamated antifolate is a polyglutamate of an antifolate described in section I herein. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In some embodiments, the delivery vehicle is a liposome according to any one of [12] - [67] of the detailed description section. In some embodiments, the administered delivery vehicle is pegylated. In some embodiments, the administered delivery vehicle is non-pegylated. In further embodiments, the administered delivery vehicle comprises a targeting moiety having specific affinity for an epitope of an antigen on the surface of a pathogen associated with an infectious disease. In some embodiments, the targeting moiety is an antibody or antigen binding antibody fragment. In some embodiments, the administered delivery vehicle comprises gamma pantol containing 4, 5, 2-10, 4-6, or more than 5 gamma-glutamyl groups. In some embodiments, the administered delivery vehicle comprises a gamma-tetraglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises a gamma-pentaglutaminated antifolate agent. In other embodiments, the administered delivery vehicle comprises a gamma hexaglutaminated antifolate agent. In some embodiments, the administered delivery vehicle comprises a L γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises a D γ polyglutamated antifolate agent. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the administered delivery vehicle comprises L and D γ polyglutamated antifolate agents. In some embodiments, the administered delivery vehicle comprises 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
In some embodiments, the delivery vehicle administered is a liposome or an antibody. In some embodiments, the delivery vehicle comprises a gamma polyglutamated antifolate agent according to any one of [1] to [11] of the detailed description section. In some embodiments, the delivery vehicle comprises a polyglutamate of the antifolate agent disclosed in section I herein. In some embodiments, the delivery vehicle is a liposome according to any one of [12] - [67] of the detailed description section. In some embodiments, the delivery vehicle is an antibody (e.g., a full-length IgG antibody, a bispecific antibody, or an scFv). In some embodiments, the delivery vehicle is a liposome (e.g., Lp- γ PANTIFLOL, such as PLp- γ PANTIFLOL, NTLp- γ PANTIFLOL, NTPLp- γ PANTIFLOL, TLp- γ PANTIFLOL, or TPLp- γ PANTIFLOL). In other embodiments, the liposome is pegylated. In further embodiments, the delivery vehicle comprises on its surface a targeting moiety that specifically binds to an antigen on the surface of a target cell of interest. In other embodiments, the delivery vehicle comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin receptor, HER2, HER 72, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA.
In other embodiments, the delivery vehicle is a liposome and the liposome comprises a targeting moiety that specifically binds to a cell surface antigen selected from the group consisting of: GONMB, TACTD 2(TROP2), CEACAM5, EPCAM, folate receptor (e.g., folate receptor-alpha, folate receptor-beta, or folate receptor-), mucin 1(MUC-1), MUC-6, STEAP1, mesothelin, desmin 4, ENPP3, Guanylate Cyclase C (GCC), SLC44A4, NaPi2B, CD70(TNFSF7), CA9 (carbonic anhydrase), 5T4(TPBG), SLTRK6, SC-16, tissue factor, LIV-1(ZIP6), CGEN-15027, P-cadherin, fibronectin ectodomain B (ED-B), VEGFR2(CD309), tenascin, collagen IV, periostin, endothelin receptor, HER2, HER 72, EGFR, IGFR-1, CD RvIII, CD3, CD 36, CD105, CD133, CD138, cripto, CD38, EphA receptor, EphB receptor, EphA2, integrin (e.g., integrin α v β 3, α v β 5, or α v β 6), C242 antigen, Apo2, PSGR, NGEP, PSCA, TMEFF2, endoglin, PSMA, CD98, CD56, CanAg, and CALLA. In some embodiments, the liposomes comprise a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the liposome comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the delivery vehicle is a liposome according to any one of [48] - [67] of the detailed description section.
In some embodiments, the present disclosure provides the use of a composition comprising a gamma polyglutamated antifolate for the manufacture of a medicament for the treatment of a hyperproliferative disease. In some embodiments, the compositions comprise a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the composition comprises a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the composition comprises a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the composition comprises a liposome according to any one of [48] - [67] of the detailed description section, the liposome comprising a targeting moiety. In some embodiments, the gamma polyglutamated antifolate comprises 5 or more glutamyl groups. In some embodiments, the gamma polyglutamated antifolate agent is pentaglutamated or hexaglutamated. In some embodiments, the gamma polyglutamated antifolate is in a liposome. In some embodiments, the hyperproliferative disease is cancer. In some embodiments, the cancer is selected from: lung cancer (e.g., non-small cell lung cancer), pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies. In some embodiments, the cancer is selected from: breast cancer, advanced head and neck cancer, lung cancer, gastric cancer, osteosarcoma, non-hodgkin's lymphoma (NHL), Acute Lymphoblastic Leukemia (ALL), mycosis fungoides (cutaneous T-cell lymphoma), choriocarcinoma, chorioadenoma, non-leukemic meningeal cancer, soft tissue sarcoma (desmoid, invasive fibromatosis), bladder cancer, and Central Nervous System (CNS) lymphoma. In some embodiments, the liposome composition is administered to treat lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the liposome composition is administered to treat breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the composition is administered to treat colorectal cancer. In some embodiments, the composition is administered to treat ovarian cancer. In some embodiments, the composition is administered to treat endometrial cancer. In some embodiments, the composition is administered to treat pancreatic cancer. In some embodiments, the composition is administered to treat liver cancer. In some embodiments, the composition is administered to treat head and neck cancer. In some embodiments, the composition is administered to treat osteosarcoma. In some embodiments, the cancer is leukemia or lymphoma. In some embodiments, the hyperproliferative disease is an autoimmune disease. In some embodiments, the hyperproliferative disease is inflammation and rheumatoid arthritis.
The disclosed methods may be practiced in any subject that may benefit from the delivery of compositions contemplated herein (e.g., gamma polyglutamated antifolate compositions, such as liposomes containing gamma pentaglutamated or gamma hexaglutamated antifolates). Mammalian subjects, and particularly human subjects, are preferred. In some embodiments, subjects also include animals, such as domestic pets (e.g., dogs, cats, rabbits, and ferrets), livestock or domestic animals (e.g., cattle, pigs, sheep, chickens, and other poultry), horses (e.g., thoroughbred horses), laboratory animals (e.g., mice, rats, and rabbits), and other mammals. In other embodiments, the subject includes fish and other aquatic species.
The subject to which the agent is delivered may be a normal subject. Alternatively, a subject may have or be at risk of developing a condition that may be diagnosed or may benefit from delivery of one or more provided compositions. In some embodiments, such disorders include cancer (e.g., solid tumor cancer or non-solid cancer, such as leukemia). In some embodiments, these conditions (e.g., cancer) involve cells expressing an antigen that can be specifically bound by the targeted pegylated liposomal gamma polyglutamated antifolate agents disclosed herein. In other embodiments, these antigens specifically bind to a targeted pegylated liposomal gamma-polyglutamated antifolate and internalize the pegylated liposomal gamma-polyglutamated antifolate into a cell. In some embodiments, the targeted pegylated liposomal gamma polyglutamated antifolate specifically binds to a folate receptor expressed on the surface of a cancer cell (e.g., folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and folate receptor (FR-)).
Tests for diagnosing conditions treatable with the provided compositions are known in the art and familiar to medical practitioners. Commercially available antibodies can be used to determine whether a cell type expresses a folate receptor. These laboratory tests include, but are not limited to, microscopic analysis, culture-dependent tests (e.g., culture), and nucleic acid detection tests. These include wet scaffolds, dye enhanced microscopy, immuno microscopy (e.g., FISH), hybridization microscopy, particle agglutination, enzyme-linked immunosorbent assay, urine screening tests, DNA probe hybridization, and serological tests. In addition to performing the above laboratory tests, medical practitioners typically record a complete medical history and perform a comprehensive physical examination.
A subject having cancer can be, for example, a subject having detectable cancer cells. For example, a subject at risk for cancer may be a subject with a higher than normal probability of developing cancer. Such subjects include, for example, subjects with genetic abnormalities that have been shown to be more likely to be associated with developing cancer, subjects with familial predisposition to cancer, subjects exposed to carcinogens (i.e., carcinogens) such as tobacco, asbestos, or other chemical toxins, and subjects who have previously been treated for cancer and significantly remitted.
In some embodiments, the present disclosure provides methods for selectively delivering a folate receptor targeted pegylated liposomal gamma polyglutamated antifolate to tumor cells expressing folate receptors on their surface at a higher rate (e.g., at least two times higher, at least three times higher, at least four times higher, or at least five times higher) than cells that do not express folate receptors on their cell surface. In some embodiments, the pegylated liposomes comprise a L γ polyglutamated antifolate. In some embodiments, the pegylated liposomes comprise an alpha polyglutamated antifolate according to any one of [1] to [11] of the detailed description section. In some embodiments, the pegylated liposomes comprise a polyglutamate of the antifolate disclosed in section I herein. In some embodiments, the liposome is any one of [48] - [67] according to the detailed description section. In some embodiments, the pegylated liposomes comprise 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the pegylated liposomes comprise a D γ polyglutamated antifolate. In some embodiments, the pegylated liposomes comprise 2, 3, 4, 5, or more than 5D-gamma glutamyl groups. In some embodiments, the pegylated liposomes comprise L and D γ polyglutamated antifolates. In some embodiments, the pegylated liposomes comprise 2, 3, 4, 5, or more than 5L-gamma glutamyl groups. In some embodiments, the pegylated liposomes comprise 2, 3, 4, 5, or more than 5L-gamma glutamyl groups and 2, 3, 4, 5, or more than 5D-gamma glutamyl groups.
Combination therapy
In certain embodiments, the method or treatment comprises administering at least one additional therapeutic agent in addition to administering the gamma polyglutamated antifolate composition described herein. The additional therapeutic agent may be administered prior to, concurrently with, and/or after administration of the gamma polyglutamated antifolate composition. The additional therapeutic agent may be associated with the gamma polyglutamated antifolate delivery vehicle present in a solution containing the gamma polyglutamated antifolate delivery vehicle (e.g., co-encapsulated with the gamma polyglutamated antifolate in liposomes), or in a separate formulation from the composition containing the gamma polyglutamated antifolate composition. Also provided are pharmaceutical compositions comprising the polypeptide or agent and one or more additional therapeutic agents. In some embodiments, the at least one additional therapeutic agent comprises 1, 2, 3, or more additional therapeutic agents.
Combination therapies using two or more therapeutic agents often use agents that act through different mechanisms of action, although this is not necessary. Combination therapy with agents having different mechanisms of action may produce additive or synergistic effects. Combination therapy may allow the use of lower doses of each dose than those used in monotherapy, thereby reducing toxic side effects and/or increasing the therapeutic index of one or more polypeptides or agents. Combination therapy can reduce the likelihood that resistant cancer cells will develop. In some embodiments, the combination therapy includes a therapeutic agent that affects an immune response (e.g., enhances or activates a response) and a therapeutic agent that affects (e.g., inhibits or kills) a tumor/cancer cell.
In some embodiments, the present disclosure provides a method for treating cancer comprising administering an effective amount of a gamma polyglutamated antifolate composition and biologic disclosed herein. In some embodiments, the gamma polyglutamated antifolate agent administered is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered gamma polyglutamated antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the administered gamma polyglutamated antifolate agent is in a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the gamma polyglutamated antifolate is administered in combination with a therapeutic antibody. In other embodiments, the gamma polyglutamated antifolate is administered in combination with an anti-CD antibody (e.g., rituximab) or an antibody that binds an immune checkpoint protein (e.g., CTLA4, PD1, PDL1, and TIM 3). In other embodiments, the gamma polyglutamated antifolate is administered in combination with an fc fusion protein (e.g., etanercept).
In some embodiments, the present disclosure provides a method for treating an immune system disorder comprising administering an effective amount of a gamma polyglutamated antifolate composition and a biologic formulation disclosed herein. In some embodiments, the gamma polyglutamated antifolate agent administered is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered gamma polyglutamated antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the administered gamma polyglutamated antifolate agent is in a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the gamma polyglutamated antifolate is administered in combination with a therapeutic antibody. In other embodiments, the gamma polyglutamated antifolate is administered in combination with an anti-TNF antibody (e.g., adalimumab). In some embodiments, the gamma polyglutamated antifolate is administered in combination with an fc fusion protein (e.g., etanercept).
In some embodiments of the methods described herein, the combination of the gamma pantol composition described herein and at least one additional therapeutic agent produces an additive or synergistic result. In some embodiments, the combination therapy results in an increase in the therapeutic index of the γ pantol or agent. In some embodiments, the combination therapy results in an increase in the therapeutic index of the one or more additional therapeutic agents. In some embodiments, the combination therapy results in reduced toxicity and/or side effects of the γ pantol or agent. In some embodiments, the combination therapy results in a reduction in toxicity and/or side effects of one or more additional therapeutic agents.
In some embodiments, in addition to administering the gamma polyglutamated antifolate composition described herein, the methods or treatments described herein further comprise administering at least one additional therapeutic agent selected from the group consisting of: anti-tubulin agents, auristatins, DNA minor groove binding agents, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cisplatin, mono (platinum), bis (platinum), and trinuclear platinum complexes and carboplatin), anthracyclines, antibiotics, antifolates (e.g., polyglutamated or non-polyglutamated antifolates), antimitotics (e.g., vinca alkaloids such as vincristine, vinblastine, vinorelbine, or vindesine), radiosensitizers, steroids, taxanes, topoisomerase inhibitors (e.g., doxorubicin HCl, daunorubicin citrate, mitoxantrone HCl, actinomycin D, etoposide, topotecan HCl, teniposide (VM-26), and irinotecan), antimetabolites, chemotherapeutic sensitizers, duocarmycin, etoposide, fluorinated pyrimidines, ionophores, pharmaceutical compositions, and methods of the use thereof, Levofloxacin, nitrosoureas, cisplatin, purine antimetabolites, PARP inhibitors, and puromycin. In certain embodiments, the second therapeutic agent is an alkylating agent, an antimetabolite, an antimitotic agent, a topoisomerase inhibitor, or an angiogenesis inhibitor. In some embodiments, the gamma polyglutamated antifolate agent administered is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered gamma polyglutamated antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the administered gamma polyglutamated antifolate agent is in a liposome according to any one of [12] to [67] of the detailed description section.
Therapeutic agents that can be administered in combination with the gamma pantol compositions described herein include chemotherapeutic agents. Thus, in some embodiments, the methods or treatments described herein further comprise administering the γ pantol compositions described herein in combination with a chemotherapeutic agent or in combination with a mixture of chemotherapeutic agents. In some embodiments, the gamma polyglutamated antifolate agent administered is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered gamma polyglutamated antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the administered gamma polyglutamated antifolate agent is in a liposome according to any one of [12] to [67] of the detailed description section. Treatment with the gamma pantol composition may occur prior to, concurrently with, or after administration of chemotherapy. Combined administration may include co-administration (whether in a single pharmaceutical formulation or using separate formulations), or sequential administration in any order, but is typically used over a period of time such that all active agents are capable of exerting their biological activities simultaneously. The preparation and schedule of administration of such chemotherapeutic agents may be used according to manufacturer's instructions or determined empirically by a skilled practitioner. Preparation and dosing schedules for this Chemotherapy are also described in The Chemotherapy Source Book, 4 th edition, 2008, m.c. perry, editions, Lippincott, Williams & Wilkins, philiadelphia, PA.
Chemotherapeutic agents useful in the present invention include, but are not limited to, alkylating agents, such as thiotepa and cyclophosphamide
Figure BDA0002715408560001211
Alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridine hospitals such as benzodopa (benzodopa), carboquone (carboquone), midodopa (meteredopa), and ulidopa (uredopa); ethyleneimines and methylmelamines, including hexaminesMelamines (altretamine), triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, and trimethylolmelamine; nitrogen mustards, such as chlorambucil, chlorambucil (chlorenaphazine), chlorophosphamide, estramustine (estramustine), isocyclophosphamine, dichloromethyldiethanamine (mechlorethamine), mechlorethamine hydrochloride, melphalan, neoenticine (novembichin), benzene mustard cholesterol (phereneine), prednimustine (prednimustine), trofosfamide (trofosfamide), uracil mustard; nitrosoureas such as carmustine, chlorozotocin (chlorozotocin), fotemustine (fotemustine), lomustine, nimustine (nimustine), ramustine (ranimustine); antibiotics, such as aclacinomycins (aclacinomycins), actinomycins, antromycin (aurramycin), azaserine, bleomycin (bleomycin), actinomycin (cactinomycin), calicheamicin (calicheamicin), karabinin (carahemin), carminomycin (caminomycin), carcinomycin (carzinophilin), chromomycin (chromomycins), dactinomycin, daunomycin (detritucin), ditorelbirubicin (detubicin), 6-diazo-5-oxo-L-norleucine, adriamycin, epirubicin, esorubicin (esorubicin), idarubicin (idarubicin), marijudamycin (marijubamycin), mitomycin, mycophenolic acid, nogaxomycin (nogalamoutamycin), olivomycin (olivomycin), plemycin (polypicins), puromycin (poromycin), pleomycin), streptomycin (streptomycin), streptomycin (streptomycin, streptomyc, Neat stastatin (zinostatin), zorubicin (zorubicin); antimetabolites such as antifolates and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, antifolates, pteropterin, trimetrexate; purine analogues, such as fludarabine (fludarabine), 6-mercaptopurine, thioprimine (thiaapine), thioguanine (thioguanine); pyrimidine analogs such as ancitabine (ancitabine), azacitidine, 6-azauridine, carmofur (carmofur), cytarabine, dideoxyuridine (dideoxyuridine), doxifluridine, enocitabine (enocitabine), floxuridine, 5-FU; androgens, e.g. carroterone (calusterone), propanil Dromostanolone acid, epithioandrostanol, mepiquitaran, and testolactone; anti-adrenergic agents, such as aminoglutethimide, mitotane, trilostane; folic acid supplements, such as folinic acid (frilic acid); acetoglucurolactone (acegultone); an aldehydic phosphoramide glycoside; (ii) aminolevulinic acid; ambridine (amsacrine); betributil (betlabucil); bisantrene; edatrexate (edatraxate); defluoromine (defofamine); colchicine (de mecolcine); diazaquinone (diaziqutone); eflornithine (elformithine); ammonium etitanium acetate; etoglut (etoglucid); gallium nitrate; a hydroxyurea; lentinan; lonidamine (lonidamine); mitoguazone (mitoguzone); mitoxantrone (mitoxantrone); mopidamol (mopidamol); diamine nitracridine (nitrarine); pentostatin (pentostatin); methionine mustard (phenamett); pirarubicin (pirarubicin); podophyllinic acid; 2-ethyl linear hydrazine; procarbazine; PSK; razoxane (rizoxane); azofurans (sizofurans); germanium spiroamines (spirogyranium); tenuazonic acid (tenuazonic acid); triimine quinone (triaziquone); 2,2' -trichlorotriethylamine; urethane (urethan); vindesine (vindesine); dacarbazine (dacarbazine); mannomustine (mannomustine); dibromomannitol (mitobronitol); dibromodulcitol (mitolactol); pipobromane (pipobroman); gatifloxacin (gacytosine); cytarabine (Ara-C); taxanes, e.g. paclitaxel
Figure BDA0002715408560001221
And docetaxel
Figure BDA0002715408560001222
Chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; platinum analogs, such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; novier; norfloxacin (novantrone); (ii) teniposide; daunomycin; aminopterin; ibandronate (ibandronate); CPT 11; topoisomerase inhibitor RFS 2000; difluoromethyl ornithine (DMFO)(ii) a Retinoic acid; esperamicins (esperamicins); capecitabine
Figure BDA0002715408560001223
Anti-hormonal agents, such as tamoxifen, raloxifene, aromatase inhibiting 4(5) -imidazole, 4-hydroxyttamoxifen, trioxifene, raloxifene (keoxifene), LY117018, onapristone and toremifene
Figure BDA0002715408560001224
Antiandrogens, such as flutamide (flutamide), nilutamide (nilutamide), bicalutamide (bicalutamide), leuprolide acetate and goserelin (goserelin); and a pharmaceutically acceptable salt, acid or derivative according to any of the above. In certain embodiments, the additional therapeutic agent is cisplatin. In certain embodiments, the additional therapeutic agent is carboplatin. In other embodiments, the additional therapeutic agent is oxaliplatin (oxaloplatin).
Additional therapeutic agents that may be administered in combination with the gamma pantol compositions described herein include one or more immunotherapeutic agents.
In some embodiments, the γ pantol compositions described herein are administered in combination with an immunotherapeutic agent that inhibits one or more T cell associated inhibitory molecules (e.g., CTLA4, PD1, lymphocyte activating gene-3 (LAG-3, CD223), T cell immunoglobulin-3 (TIM-3), T cell immunoglobulin and ITIM domains (TIGIT), T cell activated V domain Ig suppressor (VISTA), B7 homolog 3(B7-H3, CD276), B and T cell lymphocyte attenuator (BTLA, CD272) or adenosine A2a receptor (A2aR) or CD 73). In some embodiments, the γ pantol composition is administered separately from the immunotherapeutic agent. In some embodiments, the γ pantol composition is administered concurrently (e.g., simultaneously or sequentially) with the immunotherapeutic agent. In some embodiments, the gamma pantol composition and the immunotherapeutic agent are encapsulated within or otherwise associated with the same liposome.
In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a PD1 inhibitor. In some embodiments, the γ pantol composition is administered in combination with pembrolizumab. In some embodiments, the γ pantol composition is administered in combination with nivolumab. In some embodiments, the gamma pantol composition is administered separately from the PD1 inhibitor. In some embodiments, the gamma pantol composition is administered simultaneously (e.g., simultaneously or sequentially) with the PD1 inhibitor. In some embodiments, the gamma pantol composition and PD1 inhibitor are encapsulated within or otherwise associated with the same liposome.
In other embodiments, the gamma pantol composition is administered in combination with a PDL1 inhibitor. In some embodiments, the γ pantol composition is administered in combination with atuzumab. In some embodiments, the γ pantol composition is administered in combination with avizumab. In some embodiments, the γ pantol composition is administered in combination with de novo mab. In some embodiments, the gamma pantol composition is administered in combination with PDR 001. In some embodiments, the gamma pantol composition is administered separately from the PDL-1 inhibitor. In some embodiments, the gamma pantol composition is administered simultaneously (e.g., simultaneously or sequentially) with the PDL-1 inhibitor. In some embodiments, the gamma pantol composition and PDL-1 inhibitor are encapsulated within or otherwise associated with the same liposome.
In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition in combination with a therapeutic agent that inhibits the activity of CTLA4 LAG3, TIM-3, TIGIT, VISTA, B7-H3, BTLA, A2aR, or CD 73. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a CTLA4 inhibitor. In other embodiments, the γ PANTIFOL composition is administered in combination with ipilimumab. In some embodiments, the methods of treatment provided herein comprise administering a gamma pantol composition in combination with a LAG3 inhibitor. In other embodiments, the gamma PANTIFOL composition is administered in combination with TSR-033, MK-4280, LAG525, BMS-986106, or MGD 013. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition in combination with a TIM-3 inhibitor. In other embodiments, the gamma pantol composition is administered in combination with MBG453 or MEDI 9447. In some embodiments, the methods of treatment provided herein comprise administering a gamma pantol composition in combination with a TIGIT inhibitor. In other embodiments, the gamma PANTIFOL composition is administered in combination with BMS-986207 or OMP-31M 32. In some embodiments, the methods of treatment provided herein comprise administering a gamma pantol composition in combination with a VISTA inhibitor. In other embodiments, the gamma PANTIFOL composition is administered in combination with JNJ-61610588 or CA-170. In some embodiments, the methods of treatment provided herein comprise administering a gamma pantol composition in combination with a B7-H3 inhibitor. In other embodiments, the γ pantol composition is administered in combination with neobilituzumab, enrlizumab (enobilituzumab), MGD009, or 8H 9. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition in combination with a BTLA inhibitor. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition in combination with an A2aR or CD73 inhibitor. In other embodiments, the gamma pantol composition is administered in combination with CPI 444. In some embodiments, the γ pantol composition is administered separately from the immunotherapeutic agent. In some embodiments, the γ pantol composition is administered concurrently (e.g., simultaneously or sequentially) with the immunotherapeutic agent. In some embodiments, the gamma pantol composition and the immunotherapeutic agent are encapsulated within or otherwise associated with the same liposome.
In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition in combination with a therapeutic agent that inhibits Transforming Growth Factor (TGF) - β, phosphoinositide 3-kinase γ (PI3K γ), killer immunoglobulin-like receptor (KIR, CD158), CD47, or indoleamine 2, 3-dioxygenase (IDO) activity. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a TGF antagonist. In other embodiments, the gamma pantol composition is administered in combination with M7824 or galencidine (galussertinib) (LY 2157299). In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a PI3K γ antagonist. In other embodiments, the gamma pantol composition is administered in combination with IPI-549. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a KIR antagonist. In other embodiments, the gamma pantol composition is administered in combination with IPH4102 or riluzumab (lirilumab). In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a CD47 antagonist. In other embodiments, the gamma PANTIFOL composition is administered in combination with Hu5F9-G4 or TTI-621. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with an IDO antagonist. In other embodiments, the γ pantol composition is administered in combination with BMS-986205, indoimod (indoximod), or indomethastat (epacadostat). In some embodiments, the γ pantol composition is administered separately from the therapeutic agent. In some embodiments, the γ pantol composition is administered simultaneously (e.g., simultaneously or sequentially) with the therapeutic agent. In some embodiments, the gamma pantol composition and the therapeutic agent are encapsulated within or otherwise associated with the same liposome.
In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition in combination with a therapeutic agent that is an agonist of OX40(CD134), an Inducible Costimulator (ICOS), a glucocorticoid-induced TNF receptor family-related protein (GITR), 4-1BB (CD137), CD40, CD27-CD70, or a Toll-like receptor (TLR). In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with an OX40 agonist. In other embodiments, the gamma PANTIFOL composition is administered in combination with GSK3174998, MOXR0916, 9B12, PF-04518600(PF-8600), MEDI6383, MEDI0562, INCACGN 01949, or GSK 3174998. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with an ICOS agonist. In other embodiments, the γ PANTIFOL composition is administered in combination with JTX-2011, GSK3359609, or MEDI-570. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a GITR agonist. In other embodiments, the gamma PANTIFOL composition is administered in combination with TRX-518, AMG 228, BMS-986156, MEDI1873, MK-4166, INCACGN 01876, or GWN 32. In some embodiments, the methods of treatment provided herein comprise administering a gamma pantol composition described herein in combination with a 4-1BB agonist. In other embodiments, the γ PANTIFOL composition is administered in combination with urotuzumab (utomobilab) or urlumab (ureluab) (PF-05082566). In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a CD40 agonist. In other embodiments, the gamma PANTIFOL composition is administered in combination with CP-870893, APX005M, ADC-1013, lucatumumab (lucatumumab), Chi Lob 7/4, daclizumab (dacetuzumab), SEA-CD40, or RO 7009789. In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a CD27-CD70 agonist. In other embodiments, the gamma PANTIFOL composition is administered in combination with ARGX-110 or BMS-936561 (MDX-1203). In some embodiments, the methods of treatment provided herein comprise administering a γ pantol composition described herein in combination with a TLR agonist. In other embodiments, the γ PANTIFOL composition is administered in combination with MEDI9197, PG545 (piratimod, pINN), or poly-ICLC. In some embodiments, the γ pantol composition is administered separately from the therapeutic agent. In some embodiments, the γ pantol composition is administered simultaneously (e.g., simultaneously or sequentially) with the therapeutic agent. In some embodiments, the gamma pantol composition and the therapeutic agent are encapsulated within or otherwise associated with the same liposome.
In some embodiments, the present disclosure provides combination therapies wherein a gamma polyglutamated antifolate composition described herein is administered in combination with another DMARD. In some embodiments, the gamma polyglutamated antifolate agent administered is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered gamma polyglutamated antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the administered gamma polyglutamated antifolate agent is in a liposome according to any one of [12] to [67] of the detailed description section. In other embodiments, the gamma polyglutamated antifolate composition is administered in combination with sulfasalazine or hydroxychloroquine. In some embodiments, the present disclosure provides combination therapies wherein the gamma polyglutamated antifolate compositions described herein are administered in combination with chloroquine.
In some embodiments, the present disclosure provides combination therapies wherein the gamma polyglutamated antifolate compositions described herein are administered in combination with a steroid. In some embodiments, the gamma polyglutamated antifolate agent administered is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered gamma polyglutamated antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the administered gamma polyglutamated antifolate agent is in a liposome according to any one of [12] to [67] of the detailed description section. In other embodiments, the gamma polyglutamated antifolate composition is administered in combination with prednisolone.
In some embodiments, the present disclosure provides combination therapies wherein the gamma polyglutamated antifolate compositions described herein are administered in combination with a biologic. In some embodiments, the gamma polyglutamated antifolate agent administered is gamma pantol according to any one of [1] to [11] of the detailed description section. In some embodiments, the administered gamma polyglutamated antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the administered gamma polyglutamated antifolate agent is in a liposome according to any one of [12] to [67] of the detailed description section. In some embodiments, the biological agent is a therapeutic antibody. In other embodiments, the therapeutic agent binds TNF- α or CD-20.
Kits comprising gamma PANTIFOL compositions
The present disclosure also provides kits comprising the gamma PANTIFOL compositions described herein and useful for performing the methods described herein. In certain embodiments, the kit comprises at least one purified gamma pantol composition in one or more containers. In some embodiments, the kit comprises a gamma polyglutamated antifolate according to any one of [1] to [11] of the detailed description. In some embodiments, the kit comprises a gamma polyglutamated antifolate, wherein the antifolate is a polyglutamate of an antifolate disclosed in section I herein. In some embodiments, the kit comprises a gamma polyglutamated antifolate according to any one of [12] to [67] of the detailed description section.
In some embodiments, the kit contains all of the components necessary and/or sufficient to perform the detection assay, including all controls, guidance for performing the assay, and any necessary software for analysis and presentation of results. One skilled in the art will readily recognize that the disclosed agents can be readily incorporated into one of the established kit forms well known in the art.
In some embodiments, the kit comprises at least one gamma pantol composition (e.g., gamma pantol liposome) as disclosed herein or a dose of a pharmaceutical formulation thereof (e.g., for treatment or diagnosis). The kit may also include suitable packaging and/or instructions for using the composition. The kit may further comprise a device for delivering the composition or pharmaceutical formulation thereof, such as an injection syringe or other device as described herein and known to those skilled in the art. One skilled in the art will readily recognize that the disclosed gamma PANTIFOL compositions can be readily incorporated into one of the established kit forms well known in the art.
Kits comprising the gamma PANTIFOL compositions and at least one additional therapeutic agent are also provided. In certain embodiments, the second (or more) therapeutic agent is an antimetabolite. In certain embodiments, the second (or more) therapeutic agent is a chemotherapeutic agent.
The following examples are intended to illustrate, but not to limit the disclosure in any way, state, or form, either explicitly or implicitly. While they are representative of embodiments that may be used, other procedures, methods, or techniques known to those skilled in the art may be used without departing from the scope of the present disclosure.
Fig. 1B-1N show the chemical formulas of exemplary gamma polyglutamates contemplated by the present disclosure.
Examples
Example 1: liposome gamma polyglutamated antifolate composition
Method of producing a composite material
Production of gamma-hexaglutaminated pemetrexed (gamma HgPMX) liposomes
Briefly, gamma-hexaglutaminated pemetrexed (gGR6) and D gamma-hexaglutaminated pemetrexed (gDGR6) were encapsulated in liposomes by the following procedure. First, the lipid components of the liposome membrane were weighed out and combined as a concentrated ethanol solution at a temperature of about 65 ℃. In this example, the lipids used were hydrogenated soy phosphatidylcholine, cholesterol and DSPE-PEG-2000(1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000 ]). The molar ratio of HSPC to cholesterol to PEG-DSPE is about 3:2: 0.15. Next, gGR6 or gDGR6 was dissolved at a concentration of 150mg/ml in 5% aqueous buffer at a pH of 6.5-6.9. The drug solution was heated to 65 ℃. The ethanolic lipid solution was injected into gGR6 or gDGR6 solution using a small bore needle. During this step, the drug solution was thoroughly stirred using a magnetic stirrer. Mixing is performed at elevated temperatures (63-72 ℃) to ensure that the lipids are in a liquid crystalline state (as opposed to a gel state achieved at temperatures below the lipid transition temperature Tm-51-54 ℃). As a result, the lipids hydrate and form multiple bilayer (multilamellar) vesicles (MLVs) containing gGR6 or gDGR6 in the aqueous core.
Shrinking MLV using filter extrusion
MLVs are fragmented into monolayer (single bilayer) vesicles of the desired size by high pressure extrusion using three passes through a stack of (track etched polycarbonate) membranes. The first pass was through a stacked membrane consisting of two layers with a pore size of 200 nm. The other two passes were made through a stacked membrane consisting of three layers with a pore size of 100 nm. During extrusion, the temperature is kept above Tm to ensure plasticity of the lipid film. Due to extrusion, large MLVs that are not uniform in size and layer become small, uniform (90-125nm) unilamellar vesicles (ULVs), which sequester the drug inside. Hydrodynamic dimensions (diameter) at 25 ℃ were measured in a quartz microcuvette using a Malvern Zetasizer Nano ZS instrument (Southborough, MA) with a backscatter detector (90 °). Samples were diluted 50-fold in the formulation matrix prior to analysis.
Purification of liposomes
After generating ULV containing gGR6 or gDGR6 pemetrexed, the liposome-external free drug was removed using tangential flow diafiltration against a suitable buffer for small volumes or for large volumes. Although any buffer solution may be used, in this example, the buffer used is 5mM HEPES, 145mM sodium chloride (pH 6.7). After completion of purification, filter sterilization was performed using a 0.22 micron filter.
Typical characteristics of the liposome derivatives are shown in the table below.
Figure BDA0002715408560001261
Dose response study of gamma HGP (Pemetrexed Hexametaxel) and liposomes
Passage on day 3 (48 hours) and day 4 (72 hours)
Figure BDA0002715408560001262
(CTG) luminescence cell viability assay to determine cell viability. This assay determines the number of viable cells in culture based on quantifying the ATP present therein, which in turn indicates the presence of metabolically active cells. CTG assays use luciferase as readout. To assess cell viability, use was made of
Figure BDA0002715408560001263
Luminocyte viability assay study dose-response inhibition of pemetrexed, HGP and liposomes on growth of different cancer cells. Human cancer cells were harvested, counted and seeded at the same cell density on day 0. On day 1, a series of 8 dilutions of each test article was added to the cells. Make itDose response curves were generated and fitted with GraphPad Prism and the IC50 for each test article was calculated. The lower the IC50, the more effective the test article is in inhibiting cancer cell growth.
On day 0, cells were seeded into 96-well plates in 100 μ l fresh medium at a cell density of 5x 104 cells per well. 8 serial 2-fold dilutions of each test article in culture medium were generated and added to the cells in triplicate on day 1. In addition, cells from three wells were treated with vehicle only (HBS for free drug or empty liposomes for liposomal HGP) as control.
On day 3 and 4, 100. mu.l of each well was added
Figure BDA0002715408560001264
Reagents and incubated for 15 minutes at room temperature. Luciferase luminescence was recorded for each well. In addition, 8 serial 2-fold dilutions of vehicle (HBS or empty liposomes) in culture medium were added to the empty wells and included in the assay to generate background luminescence signal. Luciferase signals were normalized by subtracting the background luminescence signal from the readings, respectively.
Human normal primary bone marrow CD34+ cells were obtained from ATCC (ATCC accession No. PCS-800-012). Cells were thawed at 37 ℃ for 1 minute and then placed on ice. The cells were then resuspended in StemBan SFEM (Stem cell technology catalog number 9650) plus 10% heat-inactivated fetal bovine serum (Corning 35-015-CV). Cells were seeded at a density of 2.5x104 cells/well in 96-well culture plates. The next day, viable cells were collected via centrifugation and resuspended at a density of 2.5 × 104 cells/well in neutrophil growth medium (StemBan SFEM plus 10% heat-inactivated fetal bovine serum plus 100ng/ml human stem cell factor (Sigma Cat. No. H8416), 20ng/ml human granulocyte colony stimulating factor (Sigma Cat. No. H5541), and 10ng/ml human recombinant IL3(Sigma SRP 3090). The cells were incubated at 37 ℃ for 10 days.fresh medium was added every two days.mature neutrophils were then collected and seeded into 96-well plates at a density of 1 × 104 cells/well, and incubated overnight at 37 deg.C the next day, the test article or vehicle was resuspended in neutrophil growth medium and added to the plate, then the cells were incubated at 37 deg.C for 48 or 72 hours, measurements were then performed at each time point using the Cell Titer Glo assay (Promega catalog No. G7572).
The methods used for the cell lines AML12 (non-cancerous hepatocytes) and CCD841 (non-cancerous colonic epithelial cells) are similar to those used for cancer cells.
Results
In a set of dose-response experiments, 6 cell lines representing different types of cancer were studied, namely HT-29 (colon cancer), H2342(NSCLC, adenocarcinoma subtype), H292(NSCLC, adenocarcinoma subtype), SW620(CRC), H1806 (triple negative breast cancer) and OAW28 (ovarian cancer) (fig. 5). The treatment consisted of 48 hours exposure using 2 different encapsulated derivatives of liposomal gamma pemetrexed hexaglutamate, namely liposomal gamma L hexaglutamate (liposomal gG6) and its mirror image liposomal gamma D hexaglutamate (liposomal gDG6), also known as its corresponding enantiomer.
The relative potency of the above derivatives compared to pemetrexed within 48 hours after exposure is shown in fig. 5. For each cell line, as shown in this figure, the relative potency of treatment with the various derivatives was calculated by dividing IC50 for pemetrexed by IC50 for liposomal γ pemetrexed hexaglutamate. As shown in this figure, the potency of liposomal gamma pemetrexed hexaglutamate far exceeded that of pemetrexed in all cell lines. For example, consider NSCLC cell line H292. As shown, the potency of liposomal gamma pemetrexed hexaglutamate was greater than or equal to 50 times that of pemetrexed. This indicates that 2% or lower doses of liposomal gamma pemetrexed hexaglutamate may have the same therapeutic effect as 100% doses of pemetrexed.
Cancer cell viability studies comparing the cytotoxic activity of liposomal gamma pemetrexed hexaglutamate derivatives (liposomal L γ G6/Lps Hexa gG6 and liposomal D γ G6/Lps Hexa gDG6) and pemetrexed on representative cell lines in breast, colon, lung and ovarian cancers are shown in figures 2, 3, 4, 6, 7 and 8. These data indicate that both liposomal γ L pemetrexed hexaglutamate and liposomal γ D pemetrexed hexaglutamate are more effective than pemetrexed. Furthermore, as an indicator of efficacy, experimental results for the same cell line at different dose levels in the range of 16 to 128nM are depicted in fig. 9-11. As shown in these figures, liposomal γ L pemetrexed hexaglutamate and liposomal γ D pemetrexed hexaglutamate are superior to pemetrexed in inhibiting cancer cells for lung and breast cancer cell lines in each of these dose ranges. In ovarian cancer cell lines, pemetrexed at 128nM dose appears to be as effective as liposomal gamma pemetrexed hexaglutamate, while liposomal gamma pemetrexed hexaglutamate at 32nM and 64nM doses had better therapeutic efficacy than pemetrexed; at 16nM, the therapeutic effect was lower and similar in magnitude for liposomal gamma pemetrexed hexaglutamate and pemetrexed.
The major toxicity observed in patients treated with pemetrexed is myelosuppression, which is manifested as a decrease in blood counts, including neutrophil counts (a type of white blood cell). There are also some adverse effects on the intima of the mouth and intestines, which are manifested as diarrhea and mucositis, and in some cases, adverse effects on the liver. To assess the above toxicity, the treatment of liposomal gamma pemetrexed hexa-glutamate derivatives (L and D) and pemetrexed was measured at 48 hours on CD34+ cells differentiated into neutrophils, CCD841 colonic epithelial cells, and AML12 hepatocytes. As shown in fig. 12, liposomal gamma pemetrexed hexaglutamate is significantly less toxic to differentiated human neutrophils compared to pemetrexed. This is also supported by the better retained neutrophil counts after treatment with liposomal γ L pemetrexed hexaglutamate or liposomal γ D pemetrexed hexaglutamate at doses ranging from 16nM to 128nM compared to pemetrexed (fig. 13). Strikingly, there did not appear to be any toxicity to hepatocytes after treatment with liposomal L γ pemetrexed hexaglutamate or liposomal γ D pemetrexed hexaglutamate at the dose levels studied (fig. 14). In contrast, pemetrexed at all doses studied resulted in approximately 40% reduction in hepatocyte counts. Finally, the same trend was observed after treatment of epithelial colon cells (fig. 15). As shown in this figure, all doses of pemetrexed studied resulted in a reduction in cell number of approximately ≧ 50% as compared to a reduction of approximately 20% or less after treatment with liposomal γ L pemetrexed hexaglutamate and liposomal γ D pemetrexed hexaglutamate.
Example 2:targeted liposomal polyglutamated antifolate cell delivery
The method comprises the following steps:
production of targeted gamma-hexaglutaminated pemetrexed (HGP) liposomes
Gamma HGP (gG6) was encapsulated in liposomes and the liposomes were reduced and purified according to the procedure essentially described in example 1.
Antibody conjugation
Activated liposomes were prepared by adding DSPE-PEG-maleimide to the lipid composition. Liposomes contain four different lipids: hydrogenated Soy Phosphatidylcholine (HSPC), cholesterol, 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000] (DSPE-PEG-2000) and 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ maleimide (polyethylene glycol) -2000] (DSPE-PEG-maleimide) in a ratio of 3:2:0.1125: 0.0375.
Antibody thiolation is accomplished by linking a sulfhydryl group to a primary amine using Traut's reagent (2-iminothiolane). The antibody was suspended in PBS at a concentration of 0.9-1.6 mg/ml. Traut reagent (14mM) was added to the antibody solution at a final concentration of 1-5mM, and then removed by dialysis after incubation at room temperature for 1 hour. Thiolated antibody was added to the activated liposomes at a ratio of 60g/mol phospholipid, and the reaction mixture was incubated at room temperature for 1 hour and overnight with 4uL cysteine to terminate the reaction, and unconjugated antibody was removed by dialysis.
Exemplary direct and post-insertion antibody-liposome conjugation methods are provided below.
Exemplary antibody conjugation method 1: direct conjugation
The antibody or fragment thereof (e.g., Fab or scFv) can be conjugated directly to the thiol-reactive liposome. Thiol-reactive liposomes were prepared by adding DSPE-PEG-maleimide to the lipid composition. Liposomes contain four different lipids: hydrogenated Soy Phosphatidylcholine (HSPC), cholesterol, 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000] (DSPE-PEG-2000) and 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ maleimide (polyethylene glycol) -2000] (DSPE-PEG-maleimide) in a ratio of 3:2:0.1125: 0.0375.
Thiolation of an antibody (or fragment thereof, such as a Fab or scFv) is accomplished by linking a sulfhydryl group to a primary amine using Traut's reagent (2-iminothiolane). The antibody (or fragment thereof) was suspended in PBS at a concentration of 0.9-1.6 mg/ml. Traut reagent (14mM) was added to the antibody (or fragment thereof) solution at a final concentration of 1-5mM, and then removed by dialysis after incubation at room temperature for 1 hour. The thiolated antibody (or fragment thereof) is added to the thiol-reactive liposomes at a ratio of 60g/mol phospholipid, and the reaction mixture is incubated at room temperature for 1 hour and at 4 ℃ overnight. L-cysteine was used to stop the reaction and unconjugated antibody (or fragment thereof) was removed by dialysis.
Antibodies or fragments thereof containing a cysteine residue at the C-terminus (such as Fab or scFv) can be conjugated directly to liposomes by incubating the reduced antibody (or fragment thereof) with thiol-reactive liposomes. Antibodies (or fragments thereof) with a cysteine tail are solubilized and reduced by 10-20mM reducing agents (such as 2-mercaptoethylamine, cysteine or dithioerythritol) at pH < 7. Excess reducing agent is removed completely by size exclusion chromatography or dialysis. The purified and reduced antibody (or fragment thereof) may be directly conjugated to a thiol-reactive liposome.
Exemplary antibody conjugation method 2: rear insert
Antibodies or fragments thereof containing a cysteine residue at the C-terminus (e.g., Fab or scFv) can be conjugated and incorporated into liposomes by "post-insertion" methods. Micelles of thiol-reactive lipopolymers (such as DSPE-PEG-maleimide) are prepared by dissolving in an aqueous solution at 10 mg/ml. Antibodies (or fragments thereof) with a cysteine tail are solubilized and reduced by 10-20mM reducing agents (such as 2-mercaptoethylamine, cysteine or dithioerythritol) at pH < 7. Excess reducing agent is removed completely by size exclusion chromatography or dialysis. The purified and reduced antibody (or fragment thereof) is then incubated with the micelles of thiol-reactive lipid polymer at a molar ratio of 1: 4. At the end of the reaction, the excess maleimide groups were quenched by a small amount of cysteine (1mM) or mercaptoethanol. Unconjugated antibody (or fragments thereof) is removed by size exclusion chromatography. The purified conjugated micelles are then incubated with liposomes at 37 ℃ or elevated temperature.
Physical characteristics of the nanoparticles
Dose response studies of HGP (pentaglutaminated pemetrexed) and liposomes.
Passage on day 3 (48 hours) and day 4 (72 hours)
Figure BDA0002715408560001281
(CTG) luminescence cell viability assay to determine cell viability. This assay determines the number of viable cells in culture based on quantifying the ATP present therein, which in turn indicates the presence of metabolically active cells. CTG assays use luciferase as readout. To assess cell viability, use was made of
Figure BDA0002715408560001282
Luminocyte viability assay study dose-response inhibition of pemetrexed, HGP and liposomes on growth of different cancer cells. Human cancer cells were harvested, counted and seeded at the same cell density on day 0. On day 1, a series of 8 dilutions of each test article was added to the cells. Dose response curves were generated and fitted using GraphPad Prism and the IC50 for each test article was calculated. The lower the IC50, the more effective the test article is in inhibiting cancer cell growth.
On day 0, cells were seeded into 96-well plates in 100 μ l fresh medium at a cell density of 5x 104 cells per well. 8 serial 2-fold dilutions of each test article in culture medium were generated and added to the cells in triplicate on day 1. In addition, cells from three wells were treated with vehicle only (HBS for free drug or empty liposomes for liposomal HGP) as control.
On day 3 and 4, 100. mu.l of each well was added
Figure BDA0002715408560001291
Reagents and incubated for 15 minutes at room temperature. Luciferase luminescence was recorded for each well. In addition, 8 serial 2-fold dilutions of vehicle (HBS or empty liposomes) in culture medium were added to the empty wells and included in the assay to generate background luminescence signal. Luciferase signals were normalized by subtracting the background luminescence signal from the readings, respectively.
Human normal primary bone marrow CD34+ cells were obtained from ATCC (ATCC accession No. PCS-800-012). Cells were thawed at 37 ℃ for 1 minute and then placed on ice. The cells were then resuspended in StemBan SFEM (Stem cell technology catalog number 9650) plus 10% heat-inactivated fetal bovine serum (Corning 35-015-CV). Cells were seeded at a density of 2.5x104 cells/well in 96-well culture plates. The next day, viable cells were collected via centrifugation and resuspended at a density of 2.5 × 104 cells/well in neutrophil growth medium (StemBan SFEM plus 10% heat-inactivated fetal bovine serum plus 100ng/ml human stem cell factor (Sigma Cat. No. H8416), 20ng/ml human granulocyte colony stimulating factor (Sigma Cat. No. H5541), and 10ng/ml human recombinant IL3(Sigma SRP 3090). The cells were incubated at 37 ℃ for 10 days.fresh medium was added every two days.mature neutrophils were then collected and seeded into 96-well plates at a density of 1 × 104 cells/well, and incubated overnight at 37 deg.C the next day, the test article or vehicle was resuspended in neutrophil growth medium and added to the plate, then the cells were incubated at 37 deg.C for 48 or 72 hours, measurements were then performed at each time point using the Cell Titer Glo assay (Promega catalog No. G7572).
The methods used for the cell lines AML12 (non-cancerous hepatocytes) and CCD841 (non-cancerous colonic epithelial cells) are similar to those used for cancer cells.
Results
Dose response relationships of free pemetrexed gamma hexaglutamate (gG6), (non-targeted) liposomal gamma hexaglutamate (liposomal gG6), pemetrexed, and folate receptor alpha-targeting antibody (FR1Ab) liposomal pemetrexed gamma hexaglutamate (liposomal gG6-FR1Ab) in the NCI H2342 non-small cell lung cancer (NSCLC) adenocarcinoma subtype are shown in FIG. 2. The output is the percentage of viable cells as measured by luciferase luminescence after 48 hours of treatment. As shown in fig. 2, free pemetrexed gG6 appears to be least potent as measured by IC 50. Both liposomal pemetrexed gG6 and liposomal pemetrexed gG6-FR1Ab were 7-fold and 40-fold more potent than free pemetrexed, respectively.
Similar data for HT-29 colon cancer cell lines, which depict cell viability in percent, are shown in figure 3. As shown in this figure, free pemetrexed gG6 appears to be least potent. In this case, the liposomal pemetrexed gG6 was twice as potent as pemetrexed, and the liposomal pemetrexed gG6-FR1Ab was 5 times more potent than free pemetrexed.
Example 3:polyglutamated antifolate-cisplatin complex (PGPD)
Method of producing a composite material
Folic acid analogs (also known as antifolates) have been an important anticancer therapy for the past 70 years. Such anticancer drugs used in this case interfere with various enzymes in important folate metabolic pathways. This can lead to impaired pyrimidine and purine (DNA and RNA) synthesis, impaired glycine and serine metabolism, impaired redox reactions and impaired intracellular methylation processes.
In clinical practice, antifolates (such as pemetrexed) are often used in combination with platinum agents (such as cisplatin and carboplatin). The combination results in enhanced efficacy. In this case, we set out to co-encapsulate polyglutamate with platinum agents in a specific ratio to facilitate the controlled delivery of a predetermined ratio of two anticancer drugs, namely polyglutamated pemetrexed and a platinum analog. It has been surprisingly found that long forms of polyglutamated pemetrexed (e.g., pentaglutamated pemetrexed) form complexes with cisplatin that are stable at high pH, and that such complexes dissociate into polyglutamate and cisplatin at low pH. Low pH is thought to occur in many tumor cells and tumor cell environments, particularly in hypoxic environments. The use of this discovery provides the ability to facilitate delivery of a combination of γ PPMX and a therapeutic agent, such as cisplatin, to a target cell, such as a tumor cell, and release the drug from the complex under physiologically relevant low pH conditions.
Production of polyglutamated pemetrexed-DDAP (cisplatin) complex (PGPD):
to produce polyglutaminated pemetrexed complex (polyglutaminated pemetrexed-DDAP complex), gamma pemetrexed hexaglutamate (aG6) and diaminedicarboxylic acid platinum (DDAP) were used. The complexing process depends on the presence of the platinum chloride compound and the pH conditions. Complexation is achieved by nucleophilic attack of one or both carboxyl groups of glutamate by the platinate derivative. Briefly, the complex was formed by the following procedure. First, the active compound DDAP was weighed out and dissolved in 5% dextrose. After the DDAP solubilization step, aG6 was weighed out and added to the DDAP-Captisol solution and allowed to stir for 1 hour at 45 deg.C-55 deg.C. The pH of the solution was adjusted to 6.5-7.0 using 1N NaOH and the solution was stirred for 1-2 hours. The formation of the complex was visually confirmed. However, when the pH was adjusted to acidic pH 3 to 5, the color returned to the original color, indicating the decomplexation of polyglutaminated pemetrexed and cisplatin.
Complex formation was confirmed using HPLC, which showed two distinct peaks that were combined into 1 large peak at high pH of 6.5 to 7.5, and then reappeared at low pH of 3 to 5. Repeated experiments in the absence of Captisol showed that complex formation was independent of
Figure BDA0002715408560001301
FIG. 16 depicts the structures of polyglutamate pemetrexed, Cisplatin (CDDP), and two potential gG 6-cisplatin complexes. The pH-dependent formation of interchain and/or intrachain coordination between carboxyl groups of polyglutamated pemetrexed and cisplatin may break down into individual molecules of gG6 and cisplatin when encountering the acidic pH of lysosomes (pH 3-5) and the presence of chloride ions inside the cell.
Production of Pentaglutated Pemetrexed-DDAP/CDDP Complex (PGPD) liposomes
Briefly, PGPD was encapsulated in liposomes by the following procedure. First, the lipid components of the liposome membrane were weighed out and combined as a concentrated ethanol solution at a temperature of about 65 ℃. In this example, the lipids used were hydrogenated soy phosphatidylcholine, cholesterol and DSPE-PEG-2000(1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000 ]). The molar ratio of HSPC to cholesterol to PEG-DSPE is about 3:2: 0.15. Next, PGPD was prepared as described above. The PGPD drug solution was heated to 65 ℃. The ethanol lipid solution was injected into the PGPD solution using a small bore needle. During this step, the drug solution was thoroughly stirred using a magnetic stirrer. Mixing is performed at elevated temperatures (63-72 ℃) to ensure that the lipids are in a liquid crystalline state (as opposed to a gel state achieved at temperatures below the lipid transition temperature Tm-51-54 ℃). As a result, the lipids hydrate and form multiple bilayer (multilamellar) vesicles (MLVs) containing PGPD in the aqueous core.
Minification of MLV using filter extrusion:
MLVs are fragmented into monolayer (single bilayer) vesicles of the desired size by high pressure extrusion using two passes through a stack of (track etched polycarbonate) membranes. The stacked membrane had two layers with a pore size of 200nm and six layers with a pore size of 100 nm. During extrusion, the temperature is kept above Tm to ensure plasticity of the lipid film. Due to extrusion, large MLVs that are not uniform in size and lamellar become small, uniform (90-120nm) unilamellar vesicles (ULVs), which sequester the drug inside. Hydrodynamic dimensions (diameter) at 25 ℃ were measured in polystyrene micro cuvettes using a Malvern Zetasizer Nano ZS instrument (south borough, MA) with a backscatter detector (90 °). Samples were diluted 50-fold in the formulation matrix prior to analysis.
And (3) purifying the liposome:
after generating PGPD-containing ULVs, the liposome-external PGPD is removed using tangential flow diafiltration against a suitable buffer for small volumes of column or for large volumes. Although many different buffers known in the art can be used, in this example the buffer used is 5mM HEPES, 145mM sodium chloride (pH 6.7). After completion of purification, filter sterilization was performed using a 0.22 micron filter.
Example 4: in vivo studies
The following examples describe in vivo efficacy and toxicity data obtained following administration of α G6(Lp-aG6) (α polyglutamated pemetrexed) in an in vivo (murine) model. The skilled person will understand that under the same conditions (although possibly different levels), the efficacy and reduced toxicity observed for liposomal alpha polyglutamated pemetrexed compositions is also expected to be observed after administration of the corresponding liposomal gamma polyglutamated pemetrexed (gamma G6(Lp-gG 6)).
The method comprises the following steps:
safety studies in mice
Since some of the major toxicities associated with pemetrexed-based therapies are of the hematological and hepatic, it is important to evaluate the effect of liposome α G6(Lp-aG6) in an in vivo (murine) model and compare changes in the blood and hepatic serochemical groups after treatment. To obtain this data, an initial dose range study was performed using healthy female BALB/c mice (6-8 weeks old) purchased from The Jackson Laboratory (Bar Harbor, ME). Prior to the study, animals were weighed, randomly assigned by body weight, observed for clinical abnormalities, and divided into groups (5 mice per group). Doses from 10mg/kg up to 200mg/kg were studied to identify tolerable doses in mice. Treatment was administered intravenously once a week for four weeks. Body weight and detailed clinical observations were recorded daily. At the end of the study, mice were euthanized at day 28, and blood and tissue were harvested from untreated control mice as well as mice treated with 40mg/kg liposomal aG6(Lp-aG6) and 80mg/kg liposomal aG6(Lp-aG 6). Whole blood was collected into K2-EDTA anticoagulation tubes for comprehensive complete blood cell count (CBC), and serum was separated for comprehensive chemical analysis and sent to IDEXX (Westbrook, ME) on the day of collection.
As a result:
in general, treatment with liposomal aG6 at two dose levels of 40mg/kg and 80mg/kg once per week for 4 weeks was well tolerated and there was no significant difference in body weight compared to untreated controls. To assess some of the effects on hematological parameters, White Blood Cell (WBC) counts, neutrophil counts, and platelet counts were measured after 4 weeks of treatment with liposomal aG6 at two dose levels of 40mg/kg and 80mg/kg administered once a week. As can be seen in fig. 17, there was no significant decrease in mean neutrophil, mean leukocyte and mean platelet counts after four weeks of treatment with liposome aG6 in treated animals compared to untreated control animals. Hemoglobin and reticulocyte indices were measured to assess the effect on erythrocytes. As shown in fig. 18, at the higher dose level, the mean hemoglobin concentration decreased minimally. In parallel, the mean reticulocyte proliferation index increased slightly, indicating the bone marrow response to treatment by increasing red blood cell production. Overall, this effect appeared to be minor, as the hemoglobin level of the mice was maintained after 4 weeks of treatment. These data taken together indicate that at these dosage levels of 40mg/kg and 80mg/kg once a week, there is little effect on bone marrow and related hematological indicators.
Another problem with pemetrexed is that hepatotoxicity is observed in some patients treated with pemetrexed-based therapies. To assess liver health in mice, serum chemistry including serum aspartate Aminotransferase (AST) and serum alanine Aminotransferase (ALT) and serum albumin was measured. As shown in fig. 19, mean AST and mean ALT levels of hepatic transaminase did not increase significantly at 4 weeks compared to untreated controls after 4 weeks of treatment with liposome aG6 at both dose levels of 40mg/kg and 80mg/kg administered once a week. The mean albumin level was also unchanged. These data taken together indicate an advantageous safety profile of liposome aG 6.
Preliminary experimental efficacy studies in mouse xenografts
To assess whether there was any tumor control after treatment with liposome α G6(Lp-aG6), a preliminary study was conducted. In this study, immunodeficient female nude mice (Nu/J; 6-8 weeks old) were purchased from The Jackson Laboratory (Bar Harbor, ME). NCI-H292 (non-small cell lung cancer) cells were incubated at 37 ℃ with 5% CO2The incubators were cultured in RPMI medium supplemented with 10% fetal bovine serum. 1X 106One cell was inoculated subcutaneously into the flank of the back of each mouse. Tumor volume and body weight were monitored twice weekly. Tumor-bearing mice were randomly assigned by tumor volume on day 0 And divided into groups (5 mice per group): control, pemetrexed and liposome aG 6. Pemetrexed was administered intravenously every three weeks at a dose of 167 mg/kg. This murine dose of 167mg/kg every three weeks is equivalent to 500mg/m every three weeks approved by FDA/EMA2Human dosage and schedule. Liposomal aG6 was administered intravenously at 80mg/kg once a week for 4 weeks. Tumor size was measured with calipers and tumor burden was calculated using the following equation: tumor volume is 0.5x (tumor length) x (tumor width)2(ii) a Relative tumor volume (tumor volume/tumor volume at day 0) x 100%. This study was still in progress, but preliminary data are shown in fig. 20. In this figure, the relative tumor volumes after treatment with liposome aG6 and pemetrexed are shown. As can be seen from these preliminary data, the liposome aG6 provided better tumor control than pemetrexed.
Other embodiments are as follows:
in a non-limiting embodiment of the present disclosure, a composition is provided comprising a gamma polyglutamated antifolate.
In the composition of the preceding paragraph, the composition may comprise a pentaglutamated or a hexaglutamated antifolate.
In a composition according to any of the preceding two paragraphs, the composition may comprise a gamma polyglutamated antifolate agent, which may comprise a pentaglutamated or a hexaglutamated antifolate agent.
Non-limiting exemplary liposomal gamma polyglutamated antifolate (L-gamma pantol) compositions may comprise compositions according to any of the first three paragraphs, and the liposomes may optionally be pegylated (PL-gamma pantol).
In the L- γ PANTIFOL or PL- γ PANTIFOL compositions of the previous paragraph, the gamma polyglutamated antifolate agent may comprise a pentaglutamated or a hexaglutamated antifolate agent.
In the L- γ pantol or PL- γ pantol compositions according to any of the preceding two paragraphs, the liposomes may be anionic or neutral.
In the L- γ pantol or PL- γ pantol composition according to any of the three paragraphs, a targeting moiety may be attached to one or both of PEG and the exterior of the liposome, and the targeting moiety may have specific affinity for a surface antigen on a target cell of interest (TL- γ pantol or TPL- γ pantol).
In the L- γ pantol or PL- γ pantol composition according to any of the preceding four paragraphs, the targeting moiety may be attached to one or both of PEG and the exterior of the liposome and may be a polypeptide.
In the L- γ pantol or PL- γ pantol composition according to any of the preceding five paragraphs, the targeting moiety may be attached to one or both of PEG and the exterior of the liposome and may be an antibody or a fragment of an antibody.
In the L- γ pantIFol or PL- γ pantIFol composition according to any one of the first six paragraphs, one or more of an immunostimulant, a detectable label and a maleimide may be disposed on at least one of the PEG and the exterior of the liposome.
In the L-gamma PANTIFOL or PL-gamma PANTIFOL composition according to any of the preceding seven paragraphs, e.g. using
Figure BDA0002715408560001321
The polypeptide can bind to the antigen with an equilibrium dissociation constant (Kd) in the range of 0.5x 10-10 to 10x 10-6 as determined by the assay.
In the L- γ pantol or PL- γ pantol composition according to any of the preceding eight paragraphs, the polypeptide may specifically bind to one or more folate receptors selected from: folate receptor alpha (FR-. alpha.), folate receptor beta (FR-. beta.), and folate receptor (FR-).
A non-limiting exemplary method of killing hyperproliferative cells, comprising contacting hyperproliferative cells with a liposomal gamma polyglutamated antifolate composition according to any one of the preceding nine paragraphs.
In the method of the previous paragraph, the hyperproliferative cell is a cancer cell.
A non-limiting exemplary method for treating cancer comprises administering to a subject having or at risk of having cancer an effective amount of a gamma polyglutamated antifolate composition according to any one of the preceding paragraphs of the preceding eleventh through third paragraphs.
In the method of the previous paragraph, the cancer may be one or more selected from the group consisting of: lung cancer, pancreatic cancer, breast cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colon cancer, esophageal cancer, cervical cancer, renal cancer, bile duct cancer, gallbladder cancer, and hematologic malignancies. In some embodiments, the cancer is lung cancer (e.g., NSCLC or mesothelioma). In some embodiments, the cancer is breast cancer (e.g., HER2+ + or triple negative breast cancer). In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is a head and neck cancer. In some embodiments, the cancer is osteosarcoma.
A non-limiting exemplary maintenance therapy for a subject undergoing or having undergone cancer therapy comprises administering to a subject undergoing or having undergone cancer therapy an effective amount of a gamma polyglutamated antifolate composition according to any one of the preceding paragraphs of the first thirteenth through fifth paragraphs.
Non-limiting exemplary pharmaceutical compositions can comprise any gamma polyglutamated antifolate composition according to part II.
A non-limiting exemplary method for treating an immune system disorder can comprise administering to a subject having or at risk of having an immune system disorder an effective amount of a gamma polyglutamated antifolate composition according to any one of the preceding paragraphs of the first fourteenth through sixth paragraphs.
A non-limiting exemplary method for treating an infection can comprise administering to a subject having or at risk of having an infectious disease an effective amount of a gamma polyglutamated antifolate composition according to any one of the preceding paragraphs of the preceding fifteenth paragraph through the preceding seventh paragraph.
A non-limiting exemplary method of delivery of a glutamated antifolate to a tumor expressing a folate receptor on its surface can comprise administering to a subject having said tumor an amount of a polyglutamated antifolate composition according to any one of the preceding paragraphs of the preceding sixteenth through eighth paragraphs to deliver a therapeutically effective dose of said gamma polyglutamated antifolate to said tumor.
A non-limiting exemplary method of making a liposomal gamma-polyglutamated antifolate composition comprising a gamma-polyglutamated antifolate composition according to any one of the preceding paragraphs preceding the seventeenth paragraph to the ninth paragraph comprises forming a mixture comprising a liposomal component, a gamma-polyglutamated antifolate in solution; homogenizing the mixture to form liposomes in the solution; and processing the mixture to form liposomes containing the polyglutamated antifolate agent.
A non-limiting exemplary pharmaceutical composition comprises a gamma polyglutamated antifolate composition according to any one of the preceding paragraphs of the preceding eighteenth through tenth paragraphs.
While the disclosure has been described with reference to various embodiments, it will be understood that various modifications may be made without departing from the spirit of the disclosure. The scope of the disclosure should, therefore, be determined with reference to the appended claims, along with the full scope of equivalents to which such claims are entitled. Throughout this application, various publications are referred to by author name and date or by patent number or patent publication number. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art as known to those skilled in the art as of the date of the invention described and claimed herein. However, citation of a reference herein shall not be construed as an admission that such reference is prior art to the present invention.
Various novel chemical entities, methods and apparatus for preparing these chemical entities are set forth in the following appended claims. It should be understood that the detailed description section, and not the summary and abstract sections, is intended to be used to interpret the claims. The summary and abstract sections may set forth one or more, but not all exemplary embodiments of the invention as contemplated by the inventors, and are, therefore, not intended to limit the invention and the appended claims in any way.
Us application No. 62/627,732 filed on 7.2.2018; us application No. 62/627,733 filed on 7.2.2018; us application No. 62/630,613 filed on 14/2/2018; us application No. 62/630,620 filed on 14/2/2018; us application No. 62/630,625 filed on 14/2/2018; us application No. 62/630,652 filed on 14/2/2018; us application No. 62/630,751 filed on 14/2/2018; us application No. 62/630,824 filed on 14/2/2018; us application No. 62/636,289 filed on 28.2.2018; us application No. 62/662,372 filed on 25.4.2018; us application No. 62/702,774 filed 24.7.2018; us application No. 62/702,779 filed 24.7.2018; us application No. 62/764,945 filed on 8/17 in 2018; and us application No. 62/764,951 filed 2018, 8, 17, the disclosures of each are hereby incorporated by reference in their entirety.
Sequence listing
<110> L.E.A.F. HOLDINGS GROUP, LLC (L.E.A.F. HOLDING GS GROUP LLC)
C.Nixsa (NIYIKIZA, Clet)
V.M. Moxiabout (MOYO, Victor M.)
<120> gamma polyglutamated antifolate and use thereof
<130> 6155.0112
<140> to be distributed
<141> accompanying submission
<150> US 62/627,732
<151> 2018-02-07
<150> US 62/627,733
<151> 2018-02-07
<150> US 62/630,613
<151> 2018-02-14
<150> US 62/630,620
<151> 2018-02-14
<150> US 62/630,625
<151> 2018-02-14
<150> US 62/630,652
<151> 2018-02-14
<150> US 62/630,751
<151> 2018-02-14
<150> US 62/630,824
<151> 2018-02-14
<150> US 62/636,289
<151> 2018-02-28
<150> US 62/662,372
<151> 2018-04-25
<150> US 62/702,774
<151> 2018-07-24
<150> US 62/702,779
<151> 2018-07-24
<150> US 62/764,945
<151> 2018-08-17
<150> US 62/764,951
<151> 2018-08-17
<160> 44
<170> PatentIn version 3.5
<210> 1
<211> 25
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 1
Arg Gln Ile Lys Ile Trp Phe Gln Asn Arg Arg Met Lys Trp Lys Lys
1 5 10 15
Arg Lys Lys Arg Arg Gln Arg Arg Arg
20 25
<210> 2
<211> 11
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<220>
<221> MISC _ feature
<222> (6)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 2
Arg Lys Lys Arg Arg Xaa Arg Arg Arg Gly Cys
1 5 10
<210> 3
<211> 13
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 3
ccgccaagaa gcg 13
<210> 4
<211> 100
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 4
gcgtgcacac gcgcgtagac ttcccccgca agtcactcgt tagcccgcca agaagcgacc 60
cctccggggc gagctgagcg gcgtggcgcg ggggcgtcat 100
<210> 5
<211> 101
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 5
acgtgcatac gcacgtagac attccccgct tcccactcca aagtccgcca agaagcgtat 60
cccgctgagc ggcgtggcgc gggggcgtca tccgtcagct c 101
<210> 6
<211> 58
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 6
acttcccccg caagtcactc gttagcccgc caagaagcga cccctccggg gcgagctg 58
<210> 7
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 7
Arg Lys Lys Arg Arg Gln Arg Arg Arg
1 5
<210> 8
<211> 13
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 8
Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg Thr Pro Gln
1 5 10
<210> 9
<211> 11
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 9
Tyr Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg
1 5 10
<210> 10
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 10
Ala Ala Val Ala Leu Leu Pro Ala Val Leu Leu Ala Leu Leu Ala
1 5 10 15
<210> 11
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 11
Met Gly Leu Gly Leu His Leu Leu Val Leu Ala Ala Ala Leu Gln
1 5 10 15
<210> 12
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 12
Gly Ala Leu Phe Leu Gly Phe Leu Gly Ala Ala Gly Ser Thr Met
1 5 10 15
<210> 13
<211> 21
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 13
Ala Gly Tyr Leu Leu Gly Lys Ile Asn Leu Lys Ala Leu Ala Ala Leu
1 5 10 15
Ala Lys Lys Ile Leu
20
<210> 14
<211> 16
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 14
Arg Val Ile Arg Val Trp Phe Gln Asn Lys Arg Cys Lys Asp Lys Lys
1 5 10 15
<210> 15
<211> 16
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 15
Arg Gln Ile Lys Ile Trp Phe Gln Asn Arg Arg Met Lys Trp Lys Lys
1 5 10 15
<210> 16
<211> 20
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 16
Gly Leu Phe Glu Ala Ile Ala Gly Phe Ile Glu Asn Gly Trp Glu Gly
1 5 10 15
Met Ile Asp Gly
20
<210> 17
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 17
Gly Trp Thr Leu Asn Ser Ala Gly Tyr Leu Leu Gly Lys Ile Asn
1 5 10 15
<210> 18
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 18
Arg Ser Gln Ser Arg Ser Arg Tyr Tyr Arg Gln Arg Gln Arg Ser
1 5 10 15
<210> 19
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 19
Leu Ala Ile Pro Glu Gln Glu Tyr
1 5
<210> 20
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 20
Leu Gly Ile Ala Glu Gln Glu Tyr
1 5
<210> 21
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 21
Leu Gly Ile Pro Ala Gln Glu Tyr
1 5
<210> 22
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 22
Leu Gly Ile Pro Glu Ala Glu Tyr
1 5
<210> 23
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 23
Leu Gly Ile Pro Glu Gln Ala Tyr
1 5
<210> 24
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 24
Leu Gly Ile Ala Glu Ala Glu Tyr
1 5
<210> 25
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 25
Leu Gly Ile Pro Glu Ala Ala Tyr
1 5
<210> 26
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 26
Leu Gly Ile Ala Glu Gln Ala Tyr
1 5
<210> 27
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 27
Leu Gly Ile Ala Glu Ala Ala Tyr
1 5
<210> 28
<211> 18
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 28
Leu Leu Ile Ile Leu Arg Arg Arg Ile Arg Lys Gln Ala His Ala His
1 5 10 15
Ser Lys
<210> 29
<211> 12
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 29
Leu Lys Ala Leu Ala Ala Leu Ala Lys Lys Ile Leu
1 5 10
<210> 30
<211> 18
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 30
Lys Leu Ala Leu Lys Leu Ala Leu Lys Ala Leu Lys Ala Ala Leu Lys
1 5 10 15
Leu Ala
<210> 31
<211> 21
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 31
Lys Glu Thr Trp Trp Glu Thr Trp Trp Thr Glu Trp Ser Gln Pro Lys
1 5 10 15
Lys Lys Arg Lys Val
20
<210> 32
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 32
Asp His Gln Leu Asn Pro Ala Phe
1 5
<210> 33
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 33
Asp Pro Lys Gly Asp Pro Lys Gly
1 5
<210> 34
<211> 18
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 34
Val Thr Val Thr Val Thr Val Thr Val Thr Gly Lys Gly Asp Pro Lys
1 5 10 15
Pro Asp
<210> 35
<211> 16
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 35
Arg Gln Ile Lys Ile Trp Phe Gln Asn Arg Arg Met Lys Trp Lys Lys
1 5 10 15
<210> 36
<211> 13
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 36
Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg Pro Pro Gln
1 5 10
<210> 37
<211> 27
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 37
Gly Trp Thr Leu Asn Ser Ala Gly Tyr Leu Leu Gly Lys Ile Asn Leu
1 5 10 15
Lys Ala Leu Ala Ala Leu Ala Lys Lys Ile Leu
20 25
<210> 38
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 38
Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg
1 5 10
<210> 39
<211> 7
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 39
Arg Arg Arg Arg Arg Arg Arg
1 5
<210> 40
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 40
Arg Arg Arg Arg Arg Arg Arg Arg
1 5
<210> 41
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 41
Arg Arg Arg Arg Arg Arg Arg Arg Arg
1 5
<210> 42
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 42
Arg Arg Arg Arg Arg Arg Arg Arg Arg Arg
1 5 10
<210> 43
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<400> 43
Arg Arg Arg Arg Arg Arg Arg Arg Arg Arg
1 5 10
<210> 44
<211> 33
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<220>
<223> synthetic construct
<220>
<221> MISC _ feature
<222> (33)..(33)
<223> Xaa can be 2 to 15 Arg independently in L and/or D form
<400> 44
Tyr Thr Ile Trp Met Pro Glu Asn Pro Arg Pro Gly Thr Pro Cys Asp
1 5 10 15
Ile Phe Thr Asn Ser Arg Gly Lys Arg Ala Ser Asn Gly Gly Gly Gly
20 25 30
Xaa

Claims (94)

1. A composition comprising a gamma polyglutamated antifolate agent.
2. The composition of claim 1, wherein said antifolate is selected from the group consisting of: pirtrexin, pralatrexate, AG2034, GW1843, and LY309887, or stereoisomers thereof.
3. The composition of claim 1, wherein said antifolate is selected from the group consisting of: PMX, MTX, RTX and LTX, or stereoisomers thereof.
4. The composition of any one of claims 1-3, wherein said antifolate is selected from the group consisting of: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof.
5. The composition of claim 1, wherein said antifolate is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89.
6. The composition of any one of claims 1-5, wherein said gamma polyglutamated antifolate contains 4, 5, 2-10, 4-6, or more than 5 glutamyl groups.
7. The composition of any one of claims 1-6, wherein the gamma polyglutamated antifolate agent:
(a) is a gamma-tetraglutaminic antifolate;
(b) is a gamma-pentaglutaminated antifolate; or
(c) Is a gamma-hexaglutaminated antifolate.
8. The composition of any one of claims 1-7, wherein said gamma polyglutamated antifolate comprises 1-10 glutamyl groups having gamma carboxy linkages.
9. The composition of any one of claims 1-8, wherein:
(a) at least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form,
(b) each glutamyl group of the gamma polyglutamated antifolate is in the L form,
(c) At least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form,
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate, or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form.
10. The composition of any one of claims 1-9, wherein the polyglutamate is linear.
11. The composition of any one of claims 1-9, wherein the polyglutamate is branched.
12. A liposome composition comprising the gamma polyglutamated antifolate (Lp-gamma pantol) according to any one of claims 1-11.
13. The Lp- γ PANTIFOL composition of claim 12, wherein the polyglutamated antifolate agent is selected from:
(a) AG2034, pirtroxine, pralatrexate, GW1843, antifolate and LY 309887; or
(b) PMX, MTX, RTX and LTX, or stereoisomers thereof.
14. The Lp- γ PANTIFOL composition of claim 12 or 13, wherein the polyglutamated antifolate agent is selected from: LV (etoposide), L-folinic acid (L-5-formyltetrahydrofolic acid); 5-CH3-THF, 5-methyltetrahydrofolate; FA, folic acid; PteGlu, pteroylglutamic acid (FA); MTX, methotrexate; 2-dMTX, 2-deamino-MTX; 2-CH3-MTX, 2-deamino-2-methyl-MTX; AMT, aminopterin; 2-dAMT, 2-deamination-AMT; 2-CH3-AMT, 2-deamino-2-methyl-AMT; 10-EdAM, 10-ethyl-10-deazaaminopterin; PT523, na- (4-amino-4-deoxypteroyl) -N- (hemiphthaloyl) -L-ornithine; DDATHF (lometrexol), 5, 10-dideoxy-5, 6,7, 8-tetrahydrofolate; 5-d (i) H4PteGlu, 5-deaza-5, 6,7, 8-tetrahydroisofolic acid; N9-CH3-5-d (i) H4PteGlu, N9-methyl-5-deaza-5, 6,7, 8-tetrahydroisofolic acid; 5-dPteHCysA, na- (5-deaza pteroyl) -L-homocysteine; 5-dPteAPBA, na- (5-deaza pteroyl) -DL-2-amino-4-phosphonobutanoic acid; 5-dPteOrn, na- (5-deaza pteroyl) -L-ornithine; 5-dH4PteHCysA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-homocysteine; 5-dH4PteAPBA, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -DL-2-amino-4-phosphobutyric acid; 5-dH4PteOro, na- (5-deaza-5, 6,7, 8-tetrahydropteroyl) -L-ornithine; CB3717, N10-propargyl-5, 8-dideoxy folic acid; ICI-198,583, 2-deamino-2-methyl-N10-propargyl-5, 8-dideoxynitridofolic acid; 4-H-ICI-198,583, 4-deoxy-ICI-198,583; 4-OCH3-ICI-198,583, 4-methoxy-ICI-198,583 Glu-to-Val-ICI-198,583; valine-ICI-198; 583; Glu-to-Sub-ICI-198,583, 2-amino-suberic acid-ICI-198,583; 7-CH3-ICI-198,583, 7-methyl-ICI-198,583; ZD1694, N- [5(N- (3, 4-dihydro-2-methyl-4-oxoquinazolin-6-yl-methyl) amino) 2-thienyl) ] -L-glutamic acid; 2-NH2-ZD1694, 2-amino-ZD 1694; BW1843U89, (S) -2[5- (((1, 2-dihydro-3-methyl-1-oxobenzo (f) quinazolin-9-yl) methyl) amino-) -1-oxo-2-isoindolinyl ] -glutaric acid; LY231514, N- (4- (2- (2-amino-4, 7-dihydro-4-oxo-3H-pyrrolo [2,3-D ] pyrimidin-5-yl) ethyl) -benzoyl ] -L-glutamic acid, IAHQ, 5, 8-deaza-iso-folic acid, 2-dIAHQ, 2-deaza-IAHQ, 2-CH3-dIAHQ, 2-deaza-2-methyl-IAHQ, 5-D (i) PteGlu, 5-deaza-iso-folic acid, N9-CH3-5-D (i) PteGlu, N9-methyl-5-deaza-iso-folic acid, N9-CHO-5-D (i) PteGlu, N9-formyl-5-deaza-iso-folic acid, AG337, 3, 4-dihydro-2-amino-6-methyl-4-oxo-5- (4-pyridylthio) quinazoline; and AG377, 2, 4-diamino-6 [ N- (4- (phenylsulfonyl) benzyl) ethyl) amino ] quinazoline; or a stereoisomer thereof.
15. The Lp- γ PANTIFOL composition of any one of claims 12-14, wherein the antifolate agent is selected from the group consisting of: methotrexate, raltitrexed, pramipexole, pemetrexed, lometrexol (LMX; 5, 10-dideoxynitridotetrahydrofolate), cyclopenta [ g ] quinazoline with dipeptide ligand, CB3717, CB300945 or stereoisomers thereof, such as 6-R, S-BGC 945(ONX-0801), CB300638 and BW1843U 89.
16. The Lp- γ PANTIFOL composition of any one of claims 12-15, wherein the liposomes comprise a gamma polyglutamated antifolate agent containing 4, 5, 2-10, 4-6, or more than 5 gamma glutamyl groups.
17. The Lp- γ PANTIFOL composition of any one of claims 12-16, wherein the liposomes comprise a γ tetraglutamated antifolate.
18. The Lp- γ PANTIFOL composition of any one of claims 12-16, wherein the liposomes comprise a gamma pentaglutamated antifolate.
19. The Lp- γ PANTIFOL composition of any one of claims 12-16, wherein the liposomes comprise a gamma hexaglutaminated antifolate.
20. The Lp- γ PANTIFOL composition of any one of claims 12-19, wherein the gamma polyglutamated antifolate comprises 1-10 glutamyl groups having a gamma carboxy linkage.
21. The Lp- γ PANTIFLO composition of any one of claims 12-20, wherein:
(a) at least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form;
(b) each glutamyl group of the gamma polyglutamated antifolate agent is in the L form;
(c) at least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form;
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate; or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form.
22. The Lp- γ PANTIFOL composition of any one of claims 12-21, wherein
(a) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form;
(b) each glutamyl group of the gamma polyglutamated antifolate agent is in the L form;
(c) at least 1 glutamyl group of said gamma polyglutamated antifolate agent is in the D form;
(d) each glutamyl group of the gamma polyglutamated antifolate is in the D form, in addition to a glutamyl group of the antifolate; or
(e) At least 2 glutamyl groups of the gamma polyglutamated antifolate agent are in the L form and at least 1 glutamyl group is in the D form.
23. The Lp- γ PANTIFLOL composition of any one of claims 12-22, wherein the liposome is pegylated (PLp- γ PANTIFLOL).
24. The Lp- γ PANTIFOL composition of any one of claims 12-22, wherein the liposome is non-pegylated.
25. The Lp- γ PANTIFOL composition of any one of claims 12-24, wherein the liposome has a diameter in the range of 20nm to 200 nm.
26. The Lp- γ PANTIFOL composition of any one of claims 12-25, wherein the liposome has a diameter in the range of 80nm to 120 nm.
27. The Lp- γ PANTIFOL composition of any one of claims 12-26, wherein the liposomes are formed from liposomal components.
28. The Lp- γ PANTIFOL composition of claim 27, wherein the liposomal composition comprises at least one of anionic lipids and neutral lipids.
29. The Lp- γ PANTIFOL composition of claim 27 or 28, wherein the liposome component comprises at least one selected from the group consisting of: DSPE; DSPE-PEG; DSPE-PEG-maleimide; HSPC; HSPC-PEG; cholesterol; cholesterol-PEG; and cholesterol-maleimide.
30. The Lp- γ PANTIFOL composition of any one of claims 27-29, wherein the liposome component comprises at least one selected from the group consisting of: DSPE; DSPE-PEG; DSPE-PEG-FITC; DSPE-PEG-maleimide; cholesterol; and HSPC.
31. The Lp- γ PANTIFOL composition of any one of claims 27-30, wherein one or more liposomal components further comprises a steric stabilizer.
32. The Lp- γ PANTIFLOL composition of claim 31, wherein the steric stabilizer is at least one selected from the group consisting of: polyethylene glycol (PEG); poly-L-lysine (PLL); monosialoganglioside (GM 1); poly (vinyl pyrrolidone) (PVP); poly (acrylamide) (PAA); poly (2-methyl-2-oxazoline); poly (2-ethyl-2-oxazoline); a phosphatidylpolyglycerol; poly [ N- (2-hydroxypropyl) methacrylamide ]; amphiphilic poly-N-vinylpyrrolidone; an L-amino acid-based polymer; oligomerization of glycerol; copolymers comprising polyethylene glycol and polypropylene oxide; poloxamer 188; and polyvinyl alcohol.
33. The Lp- γ PANTIFLOL composition of claim 32, wherein the steric stabilizer is PEG and the PEG has a number average molecular weight (Mn) of 200 to 5000 daltons.
34. The Lp- γ PANTIFOL composition of any one of claims 12-33, wherein the liposome is anionic or neutral.
35. The Lp- γ PANTIFOL composition of any one of claims 12-33, wherein the liposomes have a zeta potential less than or equal to zero.
36. The Lp- γ PANTIFOL composition of any one of claims 12-33, wherein the liposomes have a zeta potential between 0 to-150 mV.
37. The Lp- γ PANTIFOL composition of any one of claims 12-33, wherein the liposomes have a zeta potential between-30 to-50 mV.
38. The Lp- γ PANTIFOL composition of any one of claims 12-33, wherein the liposome is cationic.
39. The Lp- γ PANTIFOL composition of any one of claims 12-38, wherein the liposome has an interior space comprising the γ polyglutamated antifolate agent and a pharmaceutically acceptable aqueous carrier.
40. The Lp- γ PANTIFOL composition of claim 39, wherein the pharmaceutically acceptable carrier comprises a tonicity agent such as dextrose, mannitol, glycerol, potassium chloride, sodium chloride at a concentration of greater than 1%.
41. The Lp- γ PANTIFOL composition of claim 39, wherein the pharmaceutically acceptable aqueous carrier is trehalose.
42. The Lp- γ PANTIFOL composition of claim 41, wherein the pharmaceutically acceptable carrier comprises 1% to 50% trehalose.
43. The Lp- γ PANTIFOL composition of any one of claims 39-42, wherein the pharmaceutically acceptable carrier comprises a 1% to 50% dextrose solution.
44. The Lp- γ PANTIFOL composition of any one of claims 39-43, wherein the interior space of the liposome comprises 5% dextrose suspended in HEPES buffer solution.
45. The Lp- γ PANTIFOL composition according to any one of claims 39-44, wherein the pharmaceutically acceptable carrier comprises a buffer, such as HEPES Buffered Saline (HBS) or the like, at a concentration of between 1 and 200mM and a pH of between 2 and 8.
46. The Lp- γ PANTIFOL composition of any one of claims 39-45, wherein the pharmaceutically acceptable carrier comprises sodium acetate and calcium acetate in a total concentration between 50mM to 500 mM.
47. The Lp- γ PANTIFLO composition of any one of claims 12-46, wherein the interior space of the liposome has a pH of 5-8 or a pH of 6-7, or any range therebetween.
48. The Lp- γ PANTIFOL composition of any one of claims 12-47, wherein the liposomes comprise less than 500,000 or less than 200,000 of the γ polyglutamated antifolate molecules.
49. The Lp- γ PANTIFOL composition of any one of claims 12-48, wherein the liposomes comprise between 10 to 100,000 of the γ polyglutamated antifolate molecules, or any range therebetween.
50. The Lp- γ PANTIFLOL composition of any one of claims 12-49, further comprising a targeting moiety, and wherein the targeting moiety has specific affinity for a surface antigen on a target cell of interest.
51. The Lp- γ PANTIFOL composition of 50, wherein the targeting moiety is attached to one or both of PEG and the exterior of the liposome, optionally wherein targeting moiety is attached to one or both of the PEG and the exterior of the liposome by a covalent bond.
52. The Lp- γ PANTIFLOL composition of claim 50 or 51, wherein the targeting moiety is a polypeptide.
53. The Lp- γ PANTIFLOL composition of any one of claims 50-52, wherein the targeting moiety is an antibody or an antigen-binding fragment of an antibody.
54. The Lp- γ PANTIFOL composition of any one of claims 50-53, wherein the use is for example
Figure FDA0002715408550000051
The targeting moiety was determined by assay to be at 0.5x10 -10To 10x10-6An equilibrium dissociation constant (Kd) in a range binds the surface antigen.
55. The Lp- γ PANTIFOL composition of any one of claims 50-54, wherein the targeting moiety specifically binds to one or more folate receptors selected from the group consisting of: folate receptor alpha (FR-. alpha.), folate receptor beta (FR-. beta.), and folate receptor (FR-).
56. The Lp- γ PANTIFOL composition of any one of claims 50-55, wherein the targeting moiety comprises one or more selected from the group consisting of: antibodies, humanized antibodies, antigen-binding fragments of antibodies, single chain antibodies, single domain antibodies, bispecific antibodies, synthetic antibodies, pegylated antibodies, and multimeric antibodies.
57. The Lp- γ PANTIFOL composition of any one of claims 50-56, wherein each pegylated liposome comprises 1 to 1000 or 30 to 200 targeting moieties.
58. The Lp- γ PANTIFLOL composition of any one of claims 39-57, further comprising one or more of an immunostimulatory agent, a detectable label, and a maleimide, wherein the immunostimulatory agent, the detectable label, or the maleimide is attached to the exterior of the PEG or the liposome.
59. The Lp- γ PANTIFOL composition of any one of claims 39-58, wherein the immunostimulant is at least one selected from the group consisting of: a protein immunostimulant; a nucleic acid immunostimulant; a chemical immunostimulant; a hapten; and an adjuvant.
60. The Lp- γ PANTIFLO composition of claim 58 or 59, wherein the immunostimulant is at least one selected from the group consisting of: fluorescein; fluorescein Isothiocyanate (FITC); DNP; beta glucan; beta-1, 3-glucan; beta-1, 6-glucan; resolvins (e.g., resolvins D such as D)n-6DPAOr Dn-3DPAResolvin E, or T series resolvin); and Toll-like receptor (TLR) modulationModerators such as oxidized low density lipoproteins (e.g., OXPAC, PGPC) and eritoran lipids (e.g., E5564).
61. The Lp- γ PANTIFOL composition of any one of claims 58 to 60, wherein the immunostimulatory agent and the detectable label are the same.
62. The Lp- γ PANTIFLOL composition of any one of claims 58-61, further comprising a hapten.
63. The Lp- γ PANTIFOL composition of claim 62, wherein the hapten comprises one or more of fluorescein or β 1, 6-glucan.
64. The Lp- γ PANTIFLOL composition of any one of claims 12-63, further comprising at least one cryoprotectant selected from the group consisting of: mannitol; trehalose; sorbitol; and sucrose.
65. A targeted composition comprising the composition of any one of claims 1-64.
66. A non-targeted composition comprising the composition of any one of claims 1-49.
67. The Lp- γ PANTIFLOL composition of any one of claims 12-66, further comprising carboplatin and/or pembrolizumab.
68. A pharmaceutical composition comprising the liposomal gamma-polyglutamated antifolate composition of any one of claims 12-67.
69. A pharmaceutical composition comprising the gamma polyglutamated antifolate composition of any one of claims 1-7.
70. The composition of any one of claims 1-69, for use in the treatment of a disease.
71. Use of the composition of any one of claims 1-70 in the manufacture of a medicament for treating a disease.
72. A method for treating or preventing a disease in a subject in need of such treatment or prevention, the method comprising administering to the subject the composition of any one of claims 1-70.
73. A method for treating or preventing a disease in a subject in need of such treatment or prevention, the method comprising administering to the subject the liposomal gamma polyglutamated antifolate composition of any one of claims 12-69.
74. A method of killing a hyperproliferative cell, comprising contacting a hyperproliferative cell with the composition of any one of claims 1-69.
75. A method of killing a hyperproliferative cell, comprising contacting a hyperproliferative cell with the liposomal gamma polyglutamated antifolate composition of any one of claims 12-69.
76. The method of claim 74 or 75, wherein the hyperproliferative cell is a cancer cell, a mammalian cell, and/or a human cell.
77. A method for treating cancer, the method comprising administering to a subject having or at risk of having cancer an effective amount of the composition of any one of claims 1-69.
78. A method for treating cancer, comprising administering to a subject having or at risk of having cancer an effective amount of the liposomal gamma polyglutamated antifolate composition of any one of claims 12-68.
79. The method of claim 77 or 78, wherein the cancer is selected from: non-hematologic malignancies including, for example, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, head and neck cancer, gastric cancer, gastrointestinal cancer, colorectal cancer, esophageal cancer, cervical cancer, liver cancer, kidney cancer, bile duct cancer, gall bladder cancer, sarcomas (e.g., osteosarcoma), brain cancer, central nervous system cancer, and melanoma; and hematological malignancies such as leukemia, lymphoma and other B cell malignancies, myeloma and other plasma cell dyscrasias.
80. The method of claim 77 or 78, wherein the cancer is selected from: lung cancer, breast cancer, colon cancer, pancreatic cancer, gastric cancer, bladder cancer, head and neck cancer, ovarian cancer, and cervical cancer.
81. The method of claim 77 or 78, wherein the cancer is selected from: colorectal cancer, lung cancer, breast cancer, head and neck cancer, and pancreatic cancer.
82. The method of claim 77 or 78, wherein the cancer is selected from: colorectal cancer, breast cancer, ovarian cancer, lung cancer, head and neck cancer, pancreatic cancer, gastric cancer, and mesothelioma.
83. A method for treating cancer, the method comprising administering to a subject having or at risk of having cancer cells expressing on their surface folate receptors bound by a targeting moiety an effective amount of the Lp- γ PANTIFOL composition of any one of claims 50-66.
84. A maintenance therapy for a subject undergoing or having undergone cancer therapy, the maintenance therapy comprising administering to a subject undergoing or having undergone cancer therapy an effective amount of the composition of any one of claims 1-69.
85. A maintenance therapy for a subject undergoing or having undergone cancer therapy, comprising administering to a subject undergoing or having undergone cancer therapy an effective amount of the liposomal gamma polyglutamated antifolate composition of any one of claims 12-69.
86. A method for treating an immune system disorder, the method comprising administering to a subject having or at risk of having an immune system disorder an effective amount of the composition of any one of claims 1-69, optionally wherein the immune system disorder is selected from: inflammation (e.g., acute and chronic inflammation), systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, systemic lupus erythematosus and takayasu's arteritis, and psoriasis.
87. A method for treating an immune system disorder, the method comprising administering to a subject having or at risk of an immune system disorder an effective amount of the liposomal gamma polyglutamated antifolate composition of any one of claims 8-69, optionally wherein the immune system disorder is selected from: inflammation (e.g., acute and chronic inflammation), systemic inflammation, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), crohn's disease, dermatomyositis/polymyositis, systemic lupus erythematosus and takayasu's arteritis, and psoriasis.
88. A method for treating the following diseases:
(a) an infectious disease, the method comprising administering to a subject having or at risk of having an infectious disease an effective amount of the composition of any one of claims 1-69;
(b) an infectious disease, a cardiovascular disease, a metabolic disease, or another disease, the method comprising administering to a subject having or at risk of having an infectious disease, a cardiovascular disease, or another disease an effective amount of the composition of any one of claims 1-69, wherein the disease is a member selected from: atherosclerosis, cardiovascular disease (CVD), coronary artery disease, myocardial infarction, stroke, metabolic syndrome, gestational trophoblastic disease, and ectopic pregnancy;
(c) An autoimmune disease, the method comprising administering to a subject having or at risk of having an autoimmune disease an effective amount of the composition of any one of claims 1-69;
(d) rheumatoid arthritis, the method comprising administering to a subject having or at risk of having rheumatoid arthritis an effective amount of the composition of any one of claims 1-69;
(e) an inflammatory disorder, the method comprising administering to a subject having or at risk of having inflammation an effective amount of a composition of any one of claims 1-69, optionally wherein the inflammation is acute, chronic, and/or systemic inflammation; or
(f) A skin condition, the method comprising administering to a subject having or at risk of having a skin condition an effective amount of the composition of any one of claims 1-69, optionally wherein the skin condition is psoriasis.
89. A method for treating an infectious disease, the method comprising administering to a subject having or at risk of having an infectious disease an effective amount of the liposomal gamma polyglutamated antifolate composition of any one of claims 12-69.
90. A method of delivering a gamma polyglutamated antifolate to a tumor expressing a folate receptor on the surface, the method comprising: administering to a subject having the tumor an amount of the Lp- γ PANTIFOL composition of any one of claims 1-69 to deliver a therapeutically effective dose of the γ -polyglutamated antifolate to the tumor.
91. A method of making a gamma polyglutamated antifolate composition comprising the liposomal gamma polyglutamated antifolate composition of any one of claims 12-69, the method comprising: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; and processing the mixture to form liposomes containing the gamma polyglutamated antifolate agent.
92. A method of making the composition of any one of claims 12-69, the method comprising the steps of: forming a mixture comprising a liposome component and a gamma polyglutamated antifolate agent in solution; homogenizing the mixture to form liposomes in the solution; processing the mixture to form liposomes encapsulating and/or encapsulating the gamma polyglutamated antifolate agent; and providing a targeting moiety on a surface of the liposome, the targeting moiety having a specific affinity for at least one of folate receptor alpha (FR-alpha), folate receptor beta (FR-beta), and folate receptor (FR-).
93. The method of 92, wherein the processing step comprises one or more of: film hydration, extrusion, on-line mixing, ethanol injection technology, freeze-thaw technology, reverse phase evaporation, dynamic high-pressure micro-jet, micro-jet mixing, multiple emulsion method, freeze-drying multiple emulsion method, 3D printing, membrane contactor method and stirring.
94. The method of 92, wherein the processing step comprises one or more steps of altering the size of the liposomes by one or more of extrusion, high pressure microfluidization, and/or sonication steps.
CN201980024669.5A 2018-02-07 2019-02-07 Gamma polyglutamated antifolate agent and its use Pending CN111936147A (en)

Applications Claiming Priority (29)

Application Number Priority Date Filing Date Title
US201862627733P 2018-02-07 2018-02-07
US201862627732P 2018-02-07 2018-02-07
US62/627,732 2018-02-07
US62/627,733 2018-02-07
US201862630652P 2018-02-14 2018-02-14
US201862630625P 2018-02-14 2018-02-14
US201862630751P 2018-02-14 2018-02-14
US201862630824P 2018-02-14 2018-02-14
US201862630620P 2018-02-14 2018-02-14
US201862630613P 2018-02-14 2018-02-14
US62/630,613 2018-02-14
US62/630,824 2018-02-14
US62/630,652 2018-02-14
US62/630,625 2018-02-14
US62/630,751 2018-02-14
US62/630,620 2018-02-14
US201862636289P 2018-02-28 2018-02-28
US62/636,289 2018-02-28
US201862662372P 2018-04-25 2018-04-25
US62/662,372 2018-04-25
US201862702774P 2018-07-24 2018-07-24
US201862702779P 2018-07-24 2018-07-24
US62/702,779 2018-07-24
US62/702,774 2018-07-24
US201862764951P 2018-08-17 2018-08-17
US201862764945P 2018-08-17 2018-08-17
US62/764,951 2018-08-17
US62/764,945 2018-08-17
PCT/US2019/017004 WO2019157148A1 (en) 2018-02-07 2019-02-07 Gamma polyglutamated antifolates and uses thereof

Publications (1)

Publication Number Publication Date
CN111936147A true CN111936147A (en) 2020-11-13

Family

ID=67548055

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201980024669.5A Pending CN111936147A (en) 2018-02-07 2019-02-07 Gamma polyglutamated antifolate agent and its use

Country Status (5)

Country Link
EP (1) EP3749321A4 (en)
JP (1) JP2021513533A (en)
CN (1) CN111936147A (en)
CA (1) CA3090505A1 (en)
WO (1) WO2019157148A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114555127A (en) * 2019-08-06 2022-05-27 L.E.A.F.控股集团公司 Method for preparing polyglutamated antifolates and use of compositions thereof

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112019002495A2 (en) 2016-08-12 2019-05-14 L.E.A.F. Holdings Group Llc polyglutamate antifolates and their uses
US20180236098A1 (en) 2016-08-12 2018-08-23 L.E.A.F. Holdings Group Llc Alpha and gamma-d polyglutamated antifolates and uses thereof
CA3090494A1 (en) 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated pemetrexed and uses thereof
WO2019157129A1 (en) 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated pralatrexate and uses thereof
WO2019160733A1 (en) * 2018-02-14 2019-08-22 L.E.A.F. Holdings Group Llc Gamma polyglutamated methotrexate and uses thereof
EP3752157A4 (en) 2018-02-14 2022-07-06 L.E.A.F Holdings Group LLC Gamma polyglutamated lometrexol and uses thereof
CA3090992A1 (en) 2018-02-14 2019-08-22 L.E.A.F. Holdings Group Llc Gamma polyglutamated tetrahydrofolates and uses thereof
US20210213019A1 (en) * 2018-02-14 2021-07-15 L.E.A.F. Holdings Group Llc Gamma polyglutamated aminopterin and uses thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050209239A1 (en) * 2004-03-12 2005-09-22 Barton Kamen Compositions and methods employing aminopterin
US20060160751A1 (en) * 2005-01-07 2006-07-20 Mcguire John J 5-Amino-4-imidazolecarboxamide riboside and its nucleobase as potentiators of antifolate transport and metabolism
US20080214585A1 (en) * 2007-01-19 2008-09-04 Chelsea Therapeutics, Inc. New classical antifolates
US20080234298A1 (en) * 2006-12-29 2008-09-25 Tracon Pharmaceuticals, Inc. Antifolate agent combinations in the treatment of cancer
US20080288177A1 (en) * 2006-08-30 2008-11-20 Hendrik Jan Guchelaar Pharacogenetic method for prediction of the efficacy of methotrexate monotherapy in recent-onset arthritis
CN106794164A (en) * 2014-08-14 2017-05-31 L.E.A.F.控股集团公司 Liposomal encapsulated compatibility medicine
US20180236098A1 (en) * 2016-08-12 2018-08-23 L.E.A.F. Holdings Group Llc Alpha and gamma-d polyglutamated antifolates and uses thereof

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6569432B1 (en) * 1995-02-24 2003-05-27 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen and uses thereof
US20020168761A1 (en) * 2000-01-24 2002-11-14 Gour Barbara J. Peptidomimetic modulators of cell adhesion
EP1791544A4 (en) * 2004-09-08 2007-09-12 Chelsea Therapeutics Inc Quinazoline derivatives as metabolically inert antifolate compounds
EP2437596B1 (en) * 2009-06-03 2017-05-03 Ex-Tek, LLC Skin treatment compositions
US9770414B2 (en) * 2010-05-13 2017-09-26 Pacira Pharmaceuticals, Inc. Sustained release formulation of methotrexate as a disease-modifying antirheumatic drug (DMARD) and an anti-cancer agent
CN103040748B (en) * 2012-12-18 2014-07-02 海南圣欣医药科技有限公司 Pemetrexed disodium liposome injection
WO2018031979A1 (en) * 2016-08-12 2018-02-15 L.E.A.F. Holdings Group Llc Alpha and gamma-d polyglutamated antifolates and uses thereof
BR112019002495A2 (en) * 2016-08-12 2019-05-14 L.E.A.F. Holdings Group Llc polyglutamate antifolates and their uses
EP3706801A4 (en) * 2017-11-08 2022-04-06 L.E.A.F Holdings Group LLC Platinum complexes and uses thereof
CA3090483A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Gamma polyglutamated pemetrexed and uses thereof
CA3090509A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated methotrexate and uses thereof
EP3749318A4 (en) * 2018-02-07 2022-07-06 L.E.A.F Holdings Group LLC Gamma polyglutamated raltitrexed and uses thereof
CA3090389A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated raltitrexed and uses thereof
WO2019157129A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated pralatrexate and uses thereof
US20210346381A1 (en) * 2018-02-07 2021-11-11 L.E.A.F. Holdings Group Llc Alpha polyglutamated tetrahydrofolates and uses thereof
CA3090384A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated aminopterin and uses thereof
CA3090381A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated lometrexol and uses thereof
CN111867593A (en) * 2018-02-07 2020-10-30 L.E.A.F.控股集团公司 Alpha polyglutamated antifolate agent and its use
CA3090494A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated pemetrexed and uses thereof
WO2019160733A1 (en) * 2018-02-14 2019-08-22 L.E.A.F. Holdings Group Llc Gamma polyglutamated methotrexate and uses thereof
US20210213019A1 (en) * 2018-02-14 2021-07-15 L.E.A.F. Holdings Group Llc Gamma polyglutamated aminopterin and uses thereof
CA3090753A1 (en) * 2018-02-14 2019-08-22 L.E.A.F. Holdings Group Llc Gamma polyglutamated pralatrexate and uses thereof
CA3090992A1 (en) * 2018-02-14 2019-08-22 L.E.A.F. Holdings Group Llc Gamma polyglutamated tetrahydrofolates and uses thereof
EP3752157A4 (en) * 2018-02-14 2022-07-06 L.E.A.F Holdings Group LLC Gamma polyglutamated lometrexol and uses thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050209239A1 (en) * 2004-03-12 2005-09-22 Barton Kamen Compositions and methods employing aminopterin
US20060160751A1 (en) * 2005-01-07 2006-07-20 Mcguire John J 5-Amino-4-imidazolecarboxamide riboside and its nucleobase as potentiators of antifolate transport and metabolism
US20080288177A1 (en) * 2006-08-30 2008-11-20 Hendrik Jan Guchelaar Pharacogenetic method for prediction of the efficacy of methotrexate monotherapy in recent-onset arthritis
US20080234298A1 (en) * 2006-12-29 2008-09-25 Tracon Pharmaceuticals, Inc. Antifolate agent combinations in the treatment of cancer
US20080214585A1 (en) * 2007-01-19 2008-09-04 Chelsea Therapeutics, Inc. New classical antifolates
CN106794164A (en) * 2014-08-14 2017-05-31 L.E.A.F.控股集团公司 Liposomal encapsulated compatibility medicine
US20180236098A1 (en) * 2016-08-12 2018-08-23 L.E.A.F. Holdings Group Llc Alpha and gamma-d polyglutamated antifolates and uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
D S DUCH,等: "Biochemical and cellular pharmacology of 1843U89, a novel benzoquinazoline inhibitor of thymidylate synthase" *
曹胜利,等: "抗叶酸剂类抗肿瘤药物的研究进展" *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114555127A (en) * 2019-08-06 2022-05-27 L.E.A.F.控股集团公司 Method for preparing polyglutamated antifolates and use of compositions thereof

Also Published As

Publication number Publication date
CA3090505A1 (en) 2019-08-15
WO2019157148A1 (en) 2019-08-15
EP3749321A4 (en) 2022-03-09
EP3749321A1 (en) 2020-12-16
JP2021513533A (en) 2021-05-27

Similar Documents

Publication Publication Date Title
US11779584B2 (en) Alpha polyglutamated pemetrexed and uses thereof
US20210038719A1 (en) Alpha polyglutamated antifolates and uses thereof
CN111936147A (en) Gamma polyglutamated antifolate agent and its use
US20210161899A1 (en) Gamma polyglutamated pemetrexed and uses thereof
JP7442822B2 (en) γ-polyglutaminated tetrahydrofolic acid and its uses
US20210154196A1 (en) Gamma polyglutamated methotrexate and uses thereof
US20210052592A1 (en) Alpha polyglutamated aminopterin and uses thereof
CN111954529A (en) Alpha polyglutamated methotrexate and uses thereof
US11771700B2 (en) Gamma polyglutamated lometrexol and uses thereof
US20210346381A1 (en) Alpha polyglutamated tetrahydrofolates and uses thereof
US20210213019A1 (en) Gamma polyglutamated aminopterin and uses thereof
US20210023006A1 (en) Gamma polyglutamated raltitrexed and uses thereof
US20210338675A1 (en) Alpha polyglutamated lometrexol and uses thereof
US20210169884A1 (en) Alpha polyglutamated raltitrexed and uses thereof
US20200360389A1 (en) Gamma polyglutamated antifolates and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination