WO2024062319A1 - Polymers with dual therapeutic agents - Google Patents

Polymers with dual therapeutic agents Download PDF

Info

Publication number
WO2024062319A1
WO2024062319A1 PCT/IB2023/058834 IB2023058834W WO2024062319A1 WO 2024062319 A1 WO2024062319 A1 WO 2024062319A1 IB 2023058834 W IB2023058834 W IB 2023058834W WO 2024062319 A1 WO2024062319 A1 WO 2024062319A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
occurrence
independently
linker
integer
Prior art date
Application number
PCT/IB2023/058834
Other languages
French (fr)
Inventor
Tracy Matray
Michael VANBRUNT
John Michael MCCUTCHEON
Original Assignee
Sony Group Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sony Group Corporation filed Critical Sony Group Corporation
Publication of WO2024062319A1 publication Critical patent/WO2024062319A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/605Medicinal 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 the macromolecule containing phosphorus in the main chain, e.g. poly-phosphazene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0041Xanthene dyes, used in vivo, e.g. administered to a mice, e.g. rhodamines, rose Bengal
    • A61K49/0043Fluorescein, used in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0054Macromolecular compounds, i.e. oligomers, polymers, dendrimers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0058Antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Targeted drug conjugates unlike, e.g., chemotherapy, deliver drugs to target cells, with little or no off-target activity.
  • targeted drug conjugates comprise a targeting molecule that is linked to a biologically active payload or drug.
  • ADCs Antibody-drug conjugates
  • ADCs for cancer treatment combine the targeting features of monoclonal antibodies with cancer-killing ability of cytotoxic agents to provide a therapeutic with several advantages over other chemotherapeutics.
  • challenges related to the complexity of ADC constructs, specifically the chemical linker between antibody and drug has caused significant difficulties for development of new and effective therapeutics.
  • Adcetris®, Besponsa®, Enhertu®, Mylotarg®, Padcev®, Polivy®, and Kadcyla® are commercially available globally (Zevalin® has been approved in China only).
  • compounds having the following structure (I) are provided: (I) or a stereoisomer, tautomer, or salt thereof, wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , L 1a , L 1b , L 2 , L 3 , L 4 , L 5 , L 6 , L 7 , M 1 , M 2 , M 3 , q, w, p, m, and n are as defined herein.
  • compositions which comprise a compound of structure (I) and a pharmaceutically acceptable carrier.
  • a method of treating a disease comprising administering to a subject in need thereof a therapeutically effective amount of a compound of structure (I) or a composition comprising a compound of structure (I), wherein each M 1 or M 3 is independently a biologically active moiety effective for treating the disease.
  • Amino refers to the -NH 2 group.
  • Carboxy refers to the -CO 2 H group.
  • Cyano refers to the -CN group.
  • Niro refers to the -NO 2 group.
  • Alkyl refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms (C 1 -C 12 alkyl), one to eight carbon atoms (C 1 -C 8 alkyl) or one to six carbon atoms (C 1 -C 8 alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl,
  • alkyl groups are optionally substituted.
  • Alkylene or "alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation, and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, alkylene is optionally substituted.
  • alkenylene or “alkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond and having from two to twelve carbon atoms, e.g., ethenylene, propenylene, n-butenylene, and the like.
  • the alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond.
  • the points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • alkenylene is optionally substituted.
  • "Alkynylene” or “alkynylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond and having from two to twelve carbon atoms, e.g., ethynylene, propynylene, n-butynylene, and the like.
  • the alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond.
  • alkynylene refers to any alkyl group as defined above, wherein at least one carbon- carbon bond is replaced with a carbon-oxygen bond.
  • the carbon-oxygen bond may be on the terminal end (as in an alkoxy group) or the carbon oxygen bond may be internal (i.e., C-O-C).
  • Alkylethers include at least one carbon oxygen bond, but may include more than one.
  • PEG polyethylene glycol
  • an alkylether group is optionally substituted.
  • Alkoxy refers to a group of the formula ⁇ OR a where R a is an alkyl group as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group is optionally substituted.
  • Heteroalkyl refers to an alkyl group, as defined above, comprising at least one heteroatom (e.g., N, O, P or S) within the alkyl group or at a terminus of the alkyl group.
  • the heteroatom is within the alkyl group (i.e., the heteroalkyl comprises at least one carbon-[heteroatom]x-carbon bond, where x is 1, 2 or 3).
  • the heteroatom is at a terminus of the alkyl group and thus serves to join the alkyl group to the remainder of the molecule (e.g., M1-H-A), where M1 is a portion of the molecule, H is a heteroatom and A is an alkyl group).
  • heteroalkyl group is optionally substituted.
  • exemplary heteroalkyl groups include ethylene oxide (e.g., polyethylene oxide), optionally including phosphorous-oxygen bonds, such as phosphodiester bonds.
  • Heteroalkoxy refers to a group of the formula ⁇ ORa where Ra is a heteroalkyl group as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a heteroalkoxy group is optionally substituted.
  • Heteroalkylene refers to an alkylene group, as defined above, comprising at least one heteroatom (e.g., Si, N, O, P or S) within the alkylene chain or at a terminus of the alkylene chain.
  • the heteroatom is within the alkylene chain (i.e., the heteroalkylene comprises at least one carbon-[heteroatom]-carbon bond, where x is 1, 2 or 3).
  • the heteroatom is at a terminus of the alkylene and thus serves to join the alkylene to the remainder of the molecule (e.g., M 1 -H-A-M 2 , where M 1 and M 2 are portions of the molecule, H is a heteroatom and A is an alkylene).
  • a heteroalkylene group is optionally substituted.
  • heteroalkylene groups include ethylene oxide (e.g., polyethylene oxide) and the "C,” "HEG,” and "PEG 1K” linking groups illustrated below: Multimers of the above C-linker, HEG linker and/or PEG 1K linker are included in various embodiments of heteroalkylene linkers.
  • n ranges from 19-25, for example n is 19, 20, 21, 22, 23, 24, or 25.
  • Multimers may comprise, for example, the following structure: wherein x is 0 or an integer greater than 0, for example, x ranges from 0-100 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10).
  • a “linker” refers to a contiguous chain of at least one atom, such as carbon, oxygen, nitrogen, sulfur, phosphorous, and combinations thereof, which connects a portion of a molecule to another portion of the same molecule or to a different molecule, moiety or solid support (e.g., microparticle). Linkers may connect the molecule via a covalent bond or other means, such as ionic or hydrogen bond interactions.
  • the linker is a heteroatomic linker (e.g., comprising 1-10 Si, N, O, P, or S atoms), a heteroalkylene (e.g., comprising 1-10 Si, N, O, P, or S atoms and an alkylene chain) or an alkylene linker (e.g., comprising 1-12 carbon atoms).
  • a heteroalkylene linker comprises the following structure: wherein: x 9 and x 10 are each independently an integer greater than 0.
  • Physiologically cleavable linker refers to a molecular linkage that can be split or separated a prescribed manner, resulting in two or more separate molecules while in the presence of an in vivo or in vitro environment of an organism or cell system.
  • physiological conditions that induce such a cleavage or scission event may include a temperature ranging from about 20 to 40°C, an atmospheric pressure of about 1 atm (101 kPa or 14.7 psi), a pH of about 6 to 8, a glucose concentration of about 1 to 20 mM, atmospheric oxygen concentration, and earth gravity.
  • physiological conditions include enzymatic conditions (i.e., enzymatic cleavage).
  • Bond cleavage or scission can be homolytic or heterolytic.
  • “Heteroalkenylene” is a heteroalkylene, as defined above, comprising at least one carbon- carbon double bond. Unless stated otherwise specifically in the specification, a heteroalkenylene group is optionally substituted.
  • Heteroalkynylene is a heteroalkylene comprising at least one carbon-carbon triple bond. Unless stated otherwise specifically in the specification, a heteroalkynylene group is optionally substituted.
  • Rd is a counter ion (e.g., Na + and the like) and provided that: i) Ra is S; ii) Rb is S- or SRd; iii)Rc is SH, S- or SRd; or iv) a combination of
  • Carbocyclic refers to a stable 3- to 18-membered aromatic or non-aromatic ring comprising 3 to 18 carbon atoms.
  • a carbocyclic ring may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems, and may be partially or fully saturated.
  • Non-aromatic carbocyclyl radicals include cycloalkyl, while aromatic carbocyclyl radicals include aryl.
  • a carbocyclic group is optionally substituted.
  • Cycloalkyl refers to a stable non-aromatic monocyclic or polycyclic carbocyclic ring, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond.
  • Monocyclic cycloalkyls include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Polycyclic cycloalkyls include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl- bicyclo-[2.2.1]heptanyl, and the like. Unless stated otherwise specifically in the specification, a cycloalkyl group is optionally substituted.
  • Aryl refers to a ring system comprising at least one carbocyclic aromatic ring. In some embodiments, an aryl comprises from 6 to 18 carbon atoms. The aryl ring may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
  • Aryls include, but are not limited to, aryls derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, an aryl group is optionally substituted.
  • Heterocyclic refers to a stable 3- to 18-membered aromatic or non-aromatic ring comprising one to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclic ring may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclic ring may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclic ring may be partially or fully saturated.
  • heteroaryls examples include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, pyrazolopyrimidinyl, quinuclidinyl, thiazolidin
  • heteroaryl refers to a 5- to 14-membered ring system comprising one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring.
  • the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized.
  • Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, benzoxazolinonyl, benzimidazolthionyl, carbazolyl, cinnolin
  • a heteroaryl group is optionally substituted.
  • the suffix "-ene” refers to a particular structural feature (e.g., alkyl, aryl, heteroalkyl, heteroaryl) attached to the rest of the molecule through a single bond and attached to a radical group through a single bond.
  • the suffix "-ene” refers to a linker having the structural features of the moiety to which it is attached. The points of attachment of the "-ene" chain to the rest of the molecule and to the radical group can be through one atom of or any two atoms within the chain.
  • a heteroarylene refers to a linker comprising a heteroaryl moiety as defined herein.
  • “Fused” refers to a ring system comprising at least two rings, wherein the two rings share at least one common ring atom, for example two common ring atoms.
  • the fused ring is a heterocyclyl ring or a heteroaryl ring
  • the common ring atom(s) may be carbon or nitrogen.
  • Fused rings include bicyclic, tricyclic, tertracyclic, and the like.
  • substituted means any of the above groups (e.g., alkyl, alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene, alkoxy, alkylether, phosphoalkyl, phosphoalkylether, thiophosphoalkyl, thiophosphoalkylether, carbocyclic, cycloalkyl, aryl, heterocyclic and/or heteroaryl) wherein at least one hydrogen atom (e.g., 1, 2, 3 or all hydrogen atoms) is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and
  • Substituted also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • a higher-order bond e.g., a double- or triple-bond
  • nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • R g and R h are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl.
  • Substituted further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl group.
  • each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
  • Conjugation refers to the overlap of one p-orbital with another p-orbital across an intervening sigma bond. Conjugation may occur in cyclic or acyclic compounds.
  • a “degree of conjugation” refers to the overlap of at least one p-orbital with another p-orbital across an intervening sigma bond. For example, 1, 3-butadine has one degree of conjugation, while benzene and other aromatic compounds typically have multiple degrees of conjugation.
  • Fluorescent and colored compounds typically comprise at least one degree of conjugation.
  • Fluorescent refers to a molecule which is capable of absorbing light of a particular frequency and emitting light of a different frequency.
  • Biomolecule refers to any of a variety of biological materials, including nucleic acids, carbohydrates, amino acids, polypeptides, glycoproteins, hormones, aptamers and mixtures thereof. More specifically, the term is intended to include, without limitation, RNA, DNA, oligonucleotides, modified or derivatized nucleotides, enzymes, receptors, prions, receptor ligands (including hormones), antibodies, antigens, and toxins, as well as bacteria, viruses, blood cells, and tissue cells.
  • the visually detectable biomolecules of the disclosure are prepared, as further described herein, by contacting a biomolecule with a compound having a reactive group that enables attachment of the biomolecule to the compound via any available atom or functional group, such as an amino, hydroxy, carboxyl, or sulfhydryl group on the biomolecule.
  • a "reactive group” is a moiety capable of reacting with a second reactive groups (e.g., a "complementary reactive group”) to form one or more covalent bonds, for example by a displacement, oxidation, reduction, addition or cycloaddition reaction.
  • Exemplary reactive groups are provided in Table 1, and include for example, nucleophiles, electrophiles, dienes, dienophiles, aldehyde, oxime, hydrazone, alkyne, amine, azide, acylazide, acylhalide, nitrile, nitrone, sulfhydryl, disulfide, sulfonyl halide, isothiocyanate, imidoester, activated ester, ketone, ⁇ , ⁇ -unsaturated carbonyl, alkene, maleimide, ⁇ -haloamide, epoxide, aziridine, tetrazine, tetrazole, phosphine, biotin, thiirane and the like.
  • Bio-conjugation or “bio-conjugate” and related variations refer to a chemical reaction strategy for forming a stable covalent bond between two molecules.
  • bio-conjugation is generally used when one of the molecules is a biomolecule (e.g., an antibody), but can be used to describe forming a covalent bond with a non-biomolecule (e.g., a polymeric resin).
  • the product or compound resulting from such a reaction strategy is a "conjugate,” “bio-conjugate” or a grammatical equivalent.
  • visible and “visually detectable” are used herein to refer to substances that are observable by visual inspection, without prior illumination, or chemical or enzymatic activation.
  • Such visually detectable substances absorb and emit light in a region of the spectrum ranging from about 300 to about 900 nm.
  • such substances are intensely colored, preferably having a molar extinction coefficient of at least about 40,000, more preferably at least about 50,000, still more preferably at least about 60,000, yet still more preferably at least about 70,000, and most preferably at least about 80,000 M -1 cm -1 .
  • the compounds of the disclosure may be detected by observation with the naked eye, or with the aid of an optically based detection device, including, without limitation, absorption spectrophotometers, transmission light microscopes, digital cameras and scanners.
  • Visually detectable substances are not limited to those which emit and/or absorb light in the visible spectrum.
  • UV ultraviolet
  • IR infrared
  • other regions of the electromagnetic spectrum are also included with the scope of "visually detectable” substances.
  • photostable visible dye refers to a chemical moiety that is visually detectable, as defined hereinabove, and is not significantly altered or decomposed upon exposure to light.
  • the photostable visible dye does not exhibit significant bleaching or decomposition after being exposed to light for at least one hour.
  • the visible dye is stable after exposure to light for at least 12 hours, still more preferably at least 24 hours, still yet more preferably at least one week, and most preferably at least one month.
  • photostable visible dyes suitable for use in the compounds and methods of the disclosure include azo dyes, thioindigo dyes, quinacridone pigments, dioxazine, phthalocyanine, perinone, diketopyrrolopyrrole, quinophthalone, and truarycarbonium.
  • the term "perylene derivative" is intended to include any substituted perylene that is visually detectable. However, the term is not intended to include perylene itself.
  • anthracene derivative is used analogously.
  • a derivative e.g., perylene, pyrene, anthracene or naphthalene derivative
  • an imide bisimide or hydrazamimide derivative of perylene, anthracene, naphthalene, or pyrene.
  • the visually detectable molecules of various embodiments of the disclosure are useful for a wide variety of analytical applications, such as biochemical and biomedical applications, in which there is a need to determine the presence, location, or quantity of a particular analyte (e.g., biomolecule).
  • the disclosure provides a method for visually detecting a biomolecule, comprising: (a) providing a biological system with a visually detectable biomolecule comprising the compound of structure (I) linked to a biomolecule; and (b) detecting the biomolecule by its visible properties.
  • detecting the biomolecule by its visible properties means that the biomolecule, without illumination or chemical or enzymatic activation, is observed with the naked eye, or with the aid of an optically based detection device, including, without limitation, absorption spectrophotometers, transmission light microscopes, digital cameras and scanners.
  • a densitometer may be used to quantify the amount of visually detectable biomolecule present.
  • the relative quantity of the biomolecule in two samples can be determined by measuring relative optical density. If the stoichiometry of dye molecules per biomolecule is known, and the extinction coefficient of the dye molecule is known, then the absolute concentration of the biomolecule can also be determined from a measurement of optical density.
  • biological system is used to refer to any solution or mixture comprising one or more biomolecules in addition to the visually detectable biomolecule. Nonlimiting examples of such biological systems include cells, cell extracts, tissue samples, electrophoretic gels, assay mixtures, and hybridization reaction mixtures.
  • Solid support or solid resin refers to any solid substrate known in the art for solid- phase support of molecules, for example a “microparticle” refers to any of a number of small particles useful for attachment to compounds of the disclosure, including, but not limited to, glass beads, magnetic beads, polymeric beads, nonpolymeric beads, and the like.
  • a microparticle comprises polystyrene beads.
  • the solid support or solid resin is controlled pore glass or macroporous polystyrene.
  • a "solid support residue” refers to the functional group remaining attached to a molecule when the molecule is cleaved from the solid support.
  • Solid support residues are known in the art and can be easily derived based on the structure of the solid support and the group linking the molecule thereto.
  • a “targeting moiety” is a moiety that selectively binds or associates with a particular target, such as an analyte molecule.
  • "Selectively" binding or associating means a targeting moiety preferentially associates or binds with the desired target relative to other targets.
  • the compounds disclosed herein include linkages to targeting moieties for the purpose of selectively binding or associating the compound with an analyte of interest (i.e., the target of the targeting moiety), thus allowing detection of the analyte.
  • targeting moieties include, but are not limited to, antibodies, antigens, nucleic acid sequences, enzymes, proteins, cell surface receptor antagonists, and the like.
  • the targeting moiety is a moiety, such as an antibody, that selectively binds or associates with a target feature on or in a cell, for example a target feature on a cell membrane or other cellular structure, thus allowing for detection of cells of interest.
  • Small molecules that selectively bind or associate with a desired analyte are also contemplated as targeting moieties in certain embodiments.
  • One of skill in the art will understand other analytes, and the corresponding targeting moiety, that will be useful in various embodiments.
  • Base pairing moiety refers to a heterocyclic moiety capable of hybridizing with a complementary heterocyclic moiety via hydrogen bonds (e.g., Watson-Crick base pairing).
  • Base pairing moieties include natural and unnatural bases.
  • Non-limiting examples of base pairing moieties are RNA and DNA bases such adenosine, guanosine, thymidine, cytosine and uridine and analogues thereof.
  • Embodiments of the disclosure disclosed herein are also meant to encompass all compounds being isotopically-labeled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 Cl, 123 I, and 125 I, respectively.
  • Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described below and in the following Examples using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Optional” or “optionally” means that the subsequently described event or circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • “optionally substituted alkyl” means that the alkyl group may or may not be substituted and that the description includes both substituted alkyl groups and alkyl groups having no substitution.
  • Salt includes both acid and base addition salts.
  • Acid addition salt refers to those salts which are formed with inorganic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4- acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucohepton
  • Base addition salt refers to those salts which are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • basic ion exchange resins such as
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. Crystallizations may produce a solvate of the compounds described herein. Embodiments of the present disclosure include all solvates of the described compounds.
  • the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the disclosure with one or more molecules of solvent.
  • the solvent may be water, in which case the solvate may be a hydrate.
  • the solvent may be an organic solvent.
  • the compounds of the present disclosure may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms.
  • the compounds of the disclosure may be true solvates, while in other cases the compounds of the disclosure may merely retain adventitious water or another solvent or be a mixture of water plus some adventitious solvent.
  • Embodiments of the compounds of the disclosure may contain one or more stereocenters and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids.
  • Embodiments of the present disclosure are meant to include all such possible isomers, as well as their racemic and optically pure forms.
  • Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization.
  • Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
  • HPLC high pressure liquid chromatography
  • a “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are non-superimposable mirror images of one another.
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule.
  • the present disclosure includes tautomers of any said compounds.
  • Various tautomeric forms of the compounds are easily derivable by those of ordinary skill in the art.
  • Step refers to a biologically active organic compound with four fused rings arrangement.
  • the core structure of the steroid comprises three six-member cyclohexane rings (i.e., rings A, B, and C) and one five-member cyclopentane ring.
  • Steroids vary by functional groups attached to the core structure and the oxidation stated of the rings.
  • steroids used in some embodiments include cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone.
  • "Vitamin” refers to a group of organic compounds which are essential for normal growth and nutrition and are required in small quantities in the diet because they cannot be synthesized by the body. Such compounds include Vitamin A, B1, B2, B3, B5, B6, B7, B9, B12, C, D, E, and K.
  • Vitamin E is fat soluble compounds including four tocopherols and four tocotrienols.
  • the core structure of the Vitamin E comprises 3,4-dihydro 2H-1 benzopyran.
  • the chemical structural difference between tocotrienols and tocopherols is that tocotrienols have an unsaturated isoprenoid side chain with three carbon-carbon double bonds off of the 3,4-dihydro 2H-1 benzopyran core structure whereas tocopherols have a saturated isoprenoid side chain.
  • Both tocotrienols and tocopherols have four structural isomers including ⁇ -tocopherol, ⁇ - tocopherol, ⁇ -tocopherol, ⁇ -tocopherol, ⁇ -tocotrienol, ⁇ -tocotrienol, ⁇ -tocotrienol, and ⁇ - tocotrienol.
  • An “amino acid side chain” or “side chain” refers to substituents attached to the ⁇ -carbon, ⁇ -carbon, or ⁇ -carbon of an amino acid. Amino acid side chains can be those associated with natural or unnatural amino acids.
  • An “amino acid sequence” or “peptide sequence” refers to the primary structure of linked amino acid residues along a backbone formed via peptide bonds.
  • Sequences are generally denoted from the N-terminal end to the C-terminal end.
  • Embodiments of the present invention include compounds comprising certain amino acid sequences where indicated. Amino acid sequences are indicated, where appropriate, by 3-letter or 1-letter abbreviations.
  • a “letter code,” “1-letter code,” or “3-letter code” refers to an indication or abbreviation for an amino acid or amino acid residue in an amino acid sequence.
  • a general list of 1 and 3- letter codes and the amino acid they correspond to is found below: A “beta sheet,” “ ⁇ -sheet,” “beta pleated sheet,” or “ ⁇ -pleated sheet” refers to secondary structure of amino acid sequences that forms via intra molecular folding.
  • Strands of amino acids are connected laterally by hydrogen bonding forming a generally twisted, pleated sheet.
  • Strands of amino acids that form this secondary structure are generally 3 to 10 amino acid residues in length.
  • the present invention includes amino acid sequences that include amino acid residues with beta sheet forming propensities where beta sheet moieties are indicated. These residues include, but are not limited to, glycine, methionine, serine, valine, tyrosine, phenylalanine, tryptophan, threonine, and isoleucine.
  • An “alpha helix” or “ ⁇ -helix” refers to secondary structure of amino acid chains that forms via intra molecular folding.
  • Amino acid side chain sequence influences formation of alpha helical structure.
  • the present invention includes amino acid sequences that include residues with helix-forming propensities where alpha helix moieties are indicated. These residues include, but are not limited to, glycine, methionine, alanine, arginine, histidine, leucine, glutamate, glutamic acid, phenylalanine, valine, tyrosine, and lysine.
  • the chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C.
  • M 1 is, at each occurrence, independently either i) H or ii) a moiety comprising a biologically active moiety, provided that M 1 is absent when L 1b is H;
  • M 2 is, at each occurrence, independently a moiety comprising a fluorescent dye;
  • M 3 is, at each occurrence, independently a moiety comprising a biologically active moiety.
  • M 1 or M 3 is an alkylating agent (e.g., monomethyl auristatin F, monomethyl auristatin E, SN38, pyrrolo benzo diazepine and the like) and
  • M 2 is fluorescent dye (e.g., fluorescein and the like).
  • the biological active moieties have multiple alkylating agents as the biological active moieties which are allowed to form interstrand and/or intrastrand DNA crosslinks resulting in greater DNA stabilization.
  • the biological active moieties can be attached to the polymer backbone via physiologically cleavable or non-cleavable linkers.
  • the procedures described in the present disclosure provide the ability to selectively install the physiologically cleavable and/or non- cleavable linkers. This allows for synthesizing a compound which has both physiologically cleavable and non-cleavable linkers with one or more biological active moieties.
  • the biological active moieties can be cleaved sequentially depending on physiological conditions.
  • the ratio of a biological active moiety on M 1 and a biological active moiety on M 2 can be fine-tuned based on a type of disease, progression of the disease, and a therapeutic method.
  • a compound with both multiple biological active moieties and a fluorescent moiety attached by physiologically cleavable and/or non-cleavable linkers can be synthesized.
  • Some embodiments of the present disclosure provide combinations of therapeutic agents, targeting moieties, and dye moieties (e.g., chromophores or fluorophores) that can be used for simultaneous targeting, treatment, and detection.
  • M 2 is a chromophore or fluorophore (e.g., FITC, 5-FAM, 6-FAM, and the like).
  • FITC chromophore or fluorophore
  • 5-FAM 5-FAM
  • 6-FAM fluorophore
  • the compounds of certain embodiments also provide other desirable properties, including enhanced permeability and retention effects.
  • the chemical features of embodiments of the present compounds can be adjusted to modulate the compound’s ability to permeate diseased cells/tissue and be retained within the same.
  • Ra is O or S;
  • Rb is OH, SH, O-, S-, ORd or SRd;
  • Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether;
  • Rd is a counter ion;
  • R 3 is, at each occurrence, independently H, alkyl or alkoxy;
  • R 3 is, at each occurrence, independently H, alkyl or alkoxy;
  • R 3 is, at each
  • the various linkers and substituents e.g., R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , L 1a , L 1b , L 2 , L 3 , L 4 , L 5 , L 6 , L 7 , M 1 , M 2 , M 3 , and Q
  • the optional substituent is selected to optimize the water solubility or other property of the compound of structure (I).
  • each chromophore, alkyl, alkoxy, alkylether, heteroarylene, heteroalkyl, alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene, alkoxyalkylether, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether in the compound of structure (I) is optionally substituted with one more substituent selected from the group consisting of hydroxyl, alkoxy, alkylether , alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether.
  • at least one occurrence of L 1a is an optionally substituted 5-9 membered heteroarylene linker.
  • L 1a is a substituted 5-membered heteroarylene linker.
  • L 1a is a substituted 6-membered heteroarylene linker.
  • L 1a is a substituted 7-membered heteroarylene linker.
  • L 1a is a substituted 8-membered heteroarylene linker.
  • L 1a is a substituted 9-membered heteroarylene linker.
  • L 1a is substituted with oxo, alkyl (e.g., methyl, ethyl, etc.) or combinations thereof.
  • L 1a is, at each occurrence, unsubstituted.
  • L 1a is, at each occurrence independently a pyrimidine.
  • L 1a is, at each occurrence, independently cytosine or thymine.
  • L 1a is, at each occurrence, independently selected from cytosine and thymine such that the compound comprises a sequence of cytosine and thymine bases capable of triplex formation with a target DNA sequence.
  • L 1a has one of the following structures: or .
  • L 1a has the following structure when L 1b and M 1 are absent: wherein R 8 is, at each occurrence, independently O, NH, or NR e ; R 9 is, at each occurrence, independently H, alkyl, or optionally substituted alkyl; R 10 is, at each occurrence, independently H or F; and R e is, at each occurrence, independently alkyl or optionally substituted alkyl.
  • at least one occurrence of R 8 is O.
  • at least one occurrence of R 8 is NH.
  • at least one occurrence of R 8 is NR e .
  • R 10 is F. In some more specific embodiments, R 10 is, at each occurrence, independently H. In some more specific embodiments, R 10 is, at each occurrence, independently F. In certain embodiments, R 8 is O, R 9 is H, and R 10 is H. Some embodiments provide a compound having the following structure (IA): (IA) or a stereoisomer, salt or tautomer thereof. Some embodiments provide a compound having the following structure (IB): (IB) or a stereoisomer, salt or tautomer thereof. Compounds of structure (IB) have a gemcitabine moiety incorporated to the polymeric backbone, which can act a therapeutic agent.
  • At least one occurrence of R 6 or R 7 is F or H. In certain embodiments, each occurrence of R 6 or R 7 is F or H. In some more specific embodiments, wherein at least each occurrence of R 6 and R 7 are F, R 8 is O, R 9 is H, and R 10 is H.
  • Some embodiments provide a compound having the following structure (IC): (IC) or a stereoisomer, salt or tautomer thereof, wherein k is, at each occurrence, an integer of zero or greater.
  • at least one occurrence of L 4 is heteroalkylene. In some embodiments, each occurrence of L 4 is heteroalkylene. In some embodiments, at least one occurrence of L 4 comprises alkylene oxide.
  • the alkylene oxide of L 4 is ethylene oxide. In some more specific embodiments, the ethylene oxide is polyethylene oxide. In certain embodiments, L 4 , at each occurrence, has the following structure: , wherein: z is an integer from 1 to 100; and * indicates a bond to the adjacent phosphorous atom. In some embodiments, z is an integer from 3 to 8. In some embodiments, z is 3. In some embodiments, z is 4. In some embodiments, z is 5. In some embodiments, z is 6. In some embodiments, z is 7. In some embodiments, z is 8. In some other embodiments, z is an integer from 22 to 26. In some embodiments, z is 22. In some embodiments, z is 23.
  • z is 24. In some embodiments, z is 25. In some embodiments, z is 26. In some other embodiments, z ranges from 19 to 28. In certain embodiments, the average z is 23. In some embodiments, the average z is 19, 20, 21, 22, 23, 24, 25, 26, 27, or 28. In other embodiments, at least one occurrence of L 4 is an alkylene linker (e.g., methylene). In some more specific embodiments, L 4 is an alkylene linker at each occurrence (e.g., methylene). In some embodiments, at least one occurrence of in compounds of structure (I) comprising one of the following structures: ; ; ; or wherein z is an integer from 1 to 100.
  • alkylene linker e.g., methylene
  • L 4 is an alkylene linker at each occurrence (e.g., methylene). In some embodiments, at least one occurrence of in compounds of structure (I) comprising one of the following structures: ; ; ; or where
  • z is an integer from 3 to 8. In some other embodiments, z is an integer from 22 to 26. In some other embodiments, z ranges from 19 to 28. In some embodiments, at each occurrence, in compounds of structure (I) comprising one of the following structures: ; ; ; or wherein z is an integer from 1 to 100. In some embodiments, z is an integer from 3 to 8. In some other embodiments, z is an integer from 22 to 26. In some other embodiments, z ranges from 19 to 28. Some embodiments provide a compound having one of the following structures (ID), (IE), or (IF): (ID) (IE) or (IF) or a stereoisomer, salt or tautomer thereof.
  • L 3 , L 5 , and L 6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers.
  • L 3 , L 5 or L 6 are, at each occurrence, independently alkylene.
  • L 3 , L 5 or L 6 are, at each occurrence, independently C 1 -C 6 alkylene, C2-C6 alkenylene or C2-C6 alkynylene.
  • the alkylene linker of L 3 and L 5 are C1 alkyl linker.
  • the alkylene linker of L 3 and L 5 are C2 alkyl linker. In some more specific embodiments, the alkylene linker of L 3 and L 5 are C3 alkyl linker. In some more specific embodiments, the alkylene linker of L 3 and L 5 are C4 alkyl linker. In some more specific embodiments, the alkylene linker of L 3 and L 5 are C5 alkyl linker. In some more specific embodiments, the alkylene linker of L 3 and L 5 are C6 alkyl linker. In some specific embodiments, the alkylene linker of L 6 are C1 alkyl linker. In some specific embodiments, the alkylene linker of L 6 are C2 alkyl linker.
  • the alkylene linker of L 6 are C3 alkyl linker.
  • at least one occurrence of L 6 is a direct bond.
  • each occurrence of L 6 is a direct bond.
  • Some embodiments provide a compound having one of the following structures (IG), (IH), or (IJ): (IG) (IH) or (IJ) or a stereoisomer, salt or tautomer thereof, wherein: y 1 is, at each occurrence, independently an integer from 0 to 6; and y 2 and y 3 are, at each occurrence, independently an integer from 1 to 6. In some embodiments, y 1 is, at each occurrence, an integer 0 (i.e., L 6 is a direct bond).
  • y 1 is, at each occurrence, an integer 1. In some embodiments, y 1 is, at each occurrence, an integer 2. In some embodiments, y 1 is, at each occurrence, an integer 3. In some embodiments, y 1 is, at each occurrence, an integer 4. In some embodiments, y 1 is, at each occurrence, an integer 5. In some embodiments, y 1 is, at each occurrence, an integer 6. In some embodiments, y 2 and y 3 are at each occurrence, an integer 1 In some embodiments, y 2 and y 3 are at each occurrence, an integer 2. In some embodiments, y 2 and y 3 are at each occurrence, an integer 3. In some embodiments, y 2 and y 3 are at each occurrence, an integer 4.
  • y 2 and y 3 are at each occurrence, an integer 5. In some embodiments, y 2 and y 3 are at each occurrence, an integer 6. In some specific embodiments, y 1 is, at each occurrence, an integer 0 or 1; and y 2 , and y 3 are, at each occurrence, 1.
  • the linkers L 1b can be used as a point of attachment of the M 1 moiety to the remainder of the compound.
  • a synthetic precursor which is described as structure (II) hereinbelow, to the compound of structure (I) is prepared, and the M 1 moiety is attached to the synthetic precursor using any number of coupling methods known in the art for example methods referred to as “click chemistry.”
  • click chemistry any reaction which is rapid and substantially irreversible can be used to attach M 1 to the synthetic precursor to form a compound of structure (I).
  • Exemplary reactions include the copper catalyzed reaction of an azide and alkyne to form a triazole (Huisgen 1, 3-dipolar cycloaddition), reaction of a diene and dienophile (Diels-Alder), strain-promoted alkyne-nitrone cycloaddition, reaction of a strained alkene with an azide, tetrazine or tetrazole, alkene and azide [3+2] cycloaddition, alkene and tetrazine inverse-demand Diels-Alder, alkene and tetrazole photoreaction and various displacement reactions, such as displacement of a leaving group by nucleophilic attack on an electrophilic atom.
  • a triazole Huisgen 1, 3-dipolar cycloaddition
  • Diels-Alder Diels-Alder
  • strain-promoted alkyne-nitrone cycloaddition reaction
  • Exemplary displacement reactions include reaction of an amine with: an activated ester; an N-hydroxysuccinimide ester; an isocyanate; an isothioscyanate or the like.
  • the reaction to form L 1b may be performed in an aqueous environment.
  • L 1b is, at each occurrence, a linker comprising a functional group capable of formation by reaction of two complementary reactive groups, for example a functional group which is the product of one of the foregoing “click” reactions.
  • At least one occurrence of L 1b comprises a functional group formed by reaction of an aldehyde, oxime, hydrazone, alkyne, amine, azide, acylazide, acylhalide, nitrile, nitrone, sulfhydryl, disulfide, sulfonyl halide, isothiocyanate, imidoester, activated ester, ketone, ⁇ ⁇ ⁇ -unsaturated carbonyl, alkene, maleimide, ⁇ -haloamide, epoxide, aziridine, tetrazine, tetrazole, phosphine, biotin, or thiirane with a complementary reactive group.
  • At least one occurrence of L 1b comprises a functional group formed by a reaction of an alkyne and an azide.
  • at least one occurrence of L 1b is a linker comprising a triazolyl functional group.
  • at least one occurrence of L 1b -M 1 comprises one of the following structures: or , wherein L c and L d are each independently optional linkers.
  • L c or L d , or both is absent.
  • L c or L d , or both is present.
  • L c and L d when present, are each independently alkylene or heteroalkylene.
  • L c and L d independently have one of the following structures: ; ; ; ; ; or .
  • M 1 -L 1b has one of the following structures: ; ; or , wherein a, b, c, and d are each independently an integer ranging from 1-6.
  • a, b, c, and d are each independently an integer 1.
  • a, b, c, and d are each independently an integer 2.
  • a, b, c, and d are each independently an integer 3.
  • a, b, c, and d are each independently an integer 4.
  • a, b, c, and d are each independently an integer 5. In some embodiments, a, b, c, and d are each independently an integer 6. In some embodiments, at least one occurrence of M 1 -L 1b has one of the following structures: ; ; ; ; or . In some embodiments, each occurrence of M 1 -L 1b has one of the following structures: ; ; ; or . In other embodiments, L 1b is, at least one occurrence, H. In this regard, the compound has structure (IB), (IC), (IE), (IF), (IH), or (IJ). In other embodiments, L 1b is, at each occurrence, H.
  • the compound has structure (IB), (IE), or (IH).
  • Compounds of structure (III) have a peptide-based backbone.
  • the various linkers and substituents e.g., M 2 , M 3 , Q, R 1 , R 2 , R 21 , L 1a , L 1b , L 2 , L 3 , L 5 , L 6 , L 7 and L'
  • the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (III).
  • each alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (III) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether, alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether.
  • substituents are selected to increase cellular or tissue permeation. In related embodiments, substituents are selected to increase cellular or tissue retention.
  • At least one R 21 is a neutral amino acid side chain. In some embodiments, at least one R 21 is a charged amino acid side chain. In some embodiments, R 21 is, at each occurrence, independently H, alkyl, -CH 2 CO 2 -, -CH 2 CH 2 CO 2 -, -CH 2 CH 2 CH 2 CH 2 NH 3 + ,
  • R 21 , L 5 and m are selected such that has an amino acid sequence of (G)io, (GDGDGDGDGD) or (GKGKGKGKGK).
  • R 21 , L 5 and m are selected such that has an amino acid sequence capable of forming an ⁇ -helix or 0- sheet secondary structure.
  • the amino acid sequence is (GGEEFMLVYKFARKHGG) or (GGMSMVVSGG).
  • L 5 or L 3 , or both is present for at least one occurrence.
  • L 5 or L 3 , or both is a heteroalkylene linker.
  • the heteroalkylene linker comprises a functional group capable of maintaining a positive or negative charge at pH values ranging from 3 to 11 in aqueous solution.
  • at least one occurrence of L 5 or L 3 , or both has the following structure:
  • L 5 or L 3 has the following structure: .
  • L 6 and L 2 are independently absent or a heteroalkylene linker.
  • the heteroalkylene linker is a peptidyl linker.
  • Some embodiments provide a compound having structure (IV): (IV) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: M 2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M 3 is, at each occurrence, independently a moiety comprising a biologically active moiety; L 7 is, at each occurrence, independently a linker; L 2 , L 5 and L 6 are, at each occurrence, independently a direct bond orindependently an optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker; L 8 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker comprising one or more charged moieties, provided at least one charged moiety is not a phosphate ester; R 3 is, at each occurrence, independently H, alkyl or alkoxy
  • the various linkers and substituents e.g., M 2 , M 3 , Q, R 1 , R 2 , R 3 , L 1a , L 1b , L 2 , L 5 , L 6 , L 7 , L 8 , and L'
  • the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (IV).
  • each alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker or alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (IV) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether , alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether.
  • substituents are selected to increase cellular or tissue permeation.
  • substituents are selected to increase cellular or tissue retention.
  • the alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl and alkyloxycarbonyl are optionally substituted with hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, or combinations thereof.
  • the charged moieties are positively charged.
  • the charged moieties comprise a protonated amine or quaternary amine functional group.
  • the charged moieties independently have one of the following structures: ; or wherein: R is, at each occurrence, independently H or C 1 -C 6 alkyl.
  • the charged moieties are negatively charged.
  • the charged moieties comprise a carboxylic acid, phosphate or sulfate functional group.
  • the charged moieties have the following structure: .
  • the charged moieties comprise a combination of positively charged and negatively charged moieties.
  • the charged moieties are pendant to the backbone of the compound (e.g., attached to a linker via an alkylene or heteroalkylene linker).
  • the charged moiety is part of the backbone of the compound (e.g., part of the contiguous chain of the linker).
  • L 8 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; or ’ wherein: R is, at each occurrence, independently H or C 1 -C 6 alkyl; x is an integer from 0 to 6; and m is an integer of 1 or greater, provided that m is selected such that the compound comprises at least two charged moieties.
  • the linker L 7 can be used as a point of attachment of the M 2 and M 3 moieties to the remainder of the compound.
  • a synthetic precursor to the compound of structure (I) is prepared, and the M 2 and M 3 moieties are attached to the synthetic precursor using any number of coupling methods known in the art.
  • at least one occurrence of L 7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof.
  • the functional group comprises an amide or an ester.
  • At least one occurrence of L 7 or L 1b comprises one of the following structures: ; ; ; ; ; ; ; ; or .
  • each occurrence of L 7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof.
  • the functional group comprises an amide.
  • each occurrence of L 7 or L 1b comprises one of the following structures: ; ; ; ; ; ; ; or .
  • at least one occurrence of R 3 is H.
  • at least one occurrence of R 3 is alkyl.
  • at least one occurrence of R 3 is alkoxy.
  • each occurrence of R 3 is H.
  • Rc is OL'.
  • L' is an alkylene or heteroalkylene linker to: Q, a targeting moiety, an analyte (e.g., analyte molecule), a solid support, a solid support residue, a nucleoside or a further compound of structure (I), (III), or (IV).
  • the linker L' can be any linker suitable for attaching Q, a targeting moiety, an analyte (e.g., analyte molecule), a solid support, a solid support residue, a nucleoside or a further compound of structure (I), (III), or (IV) to the compound of structure (I), (III), or (IV).
  • analyte e.g., analyte molecule
  • solid support e.g., alyte molecule
  • solid support residue e.g., a nucleoside or a further compound of structure (I), (III), or (IV) to the compound of structure (I), (III), or (IV).
  • Advantageously certain embodiments include use of L' moieties selected to increase or optimize water solubility of the compound.
  • L' is a heteroalkylene moiety.
  • L' comprises an alkylene oxide or phosphodiester moiety, or combinations thereof.
  • R 2 is L'.
  • L' is a linker to a targeting moiety.
  • R 2 is –NH 2 .
  • one of R 2 or R 1 is L' and L' is a linker comprising a covalent bond to a solid support.
  • the solid support is a polymeric bead or non-polymeric bead.
  • L' is a linker to a targeting moiety, the linker comprising an alkylene oxide or phosphodiester moiety, or combinations thereof.
  • L' has one of the following structures: ; ; ; or wherein: x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer from 1 to 10; R b is H, an electron pair or a counter ion; L'' is the targeting moiety or a linkage to the targeting moiety. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 1.
  • x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 2. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 3. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 4. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 5.
  • x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 6. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 7. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 8. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 9.
  • x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 10.
  • R b is H.
  • R b is a counter ion.
  • R b is Na + .
  • R b is K + .
  • L'' is the targeting moiety.
  • L'' is a linkage to the targeting moiety.
  • the targeting moiety is an antibody.
  • the targeting moiety is cell surface receptor antagonist.
  • the antibody or cell surface receptor antagonist is an epidermal growth factor receptor (EGFR) inhibitor, a hepatocyte growth factor receptor (HGFR) inhibitor, an insulin-like growth factor receptor (IGFR) inhibitor, a folate, or a MET inhibitor.
  • EGFR epidermal growth factor receptor
  • HGFR hepatocyte growth factor receptor
  • IGFR insulin-like growth factor receptor
  • folate a folate
  • MET inhibitor a monoclonal antibody.
  • the monoclonal antibody is Abciximab, Adalimumab, Alemtuzumab, Alirocumab, Avibactam, Basiliximab, Benralizumab, Bezlotoxumab, Blinatumomab, Brodalumab, Burosumab, Canakinumab, Caplacizumab, Certolizumab pegol, Daclizumab, Denosumab, Dupilumab, Eculizumab, Emicizumab, Erenumab, Evolocumab, Fremanezumab, Galcanezumab, Golimumab, Guselkumab, Ibalizumab, Idarucizumab, Infliximab, Itolizumab, Ixekizumab, Lanadelumab, Lokivetmab, Mepolizumab, Natalizumab, Obiltoxaximab
  • R 1 or R 2 has one of the following structures: ; ; ; ; ; ; ; or , wherein R a is H or a solid support.
  • R 2 has one of the following structures: ; or . .
  • Certain embodiments of compounds of structure (I) can be prepared according to solid- phase synthetic methods analogous to those known in the art for preparation of oligonucleotides. Accordingly, in some embodiments, L' is a linkage to a solid support, a solid support residue or a nucleoside.
  • Solid supports comprising an activated deoxythymidine (dT) group are readily available, and in some embodiments can be employed as starting material for preparation of compounds of structure (I). Accordingly, in some specific embodiments, R 1 has the following structure: .
  • dT group depicted above is included for ease of synthesis and economic efficiencies only, and is not required.
  • Other solid supports can be used and would result in a different nucleoside or solid support residue being present on L', or the nucleoside or solid support residue can be removed or modified post synthesis.
  • the analyte molecule is a nucleic acid, amino acid or a polymer thereof.
  • the analyte molecule is an enzyme, receptor, receptor ligand, antibody, glycoprotein, aptamer or prion.
  • the targeting moiety is an antibody or cell surface receptor antagonist.
  • the solid support is a polymeric bead or non-polymeric bead.
  • Q is, at each occurrence, independently a moiety comprising a reactive group capable of forming a covalent bond with an analyte molecule or a solid support.
  • Q is, at each occurrence, independently a moiety comprising a reactive group capable of forming a covalent bond with a complementary reactive group Q′.
  • Q′ is present on a further compound of structure (I) (e.g., in the R 1 or R 2 position), and Q and Q′ comprise complementary reactive groups such that reaction of the compound of structure (I) and the further compound of structure (I) results in covalently bound dimer of the compound of structure (I).
  • Multimer compounds of structure (I) can also be prepared in an analogous manner and are included within the scope of embodiments of the disclosure.
  • the type of Q group and connectivity of the Q group to the remainder of the compound of structure (I) is not limited, provided that Q comprises a moiety having appropriate reactivity for forming the desired bond.
  • Q is a moiety which is not susceptible to hydrolysis under aqueous conditions, but is sufficiently reactive to form a bond with a corresponding group on an analyte molecule or solid support (e.g., an amine, azide or alkyne).
  • analyte molecule or solid support e.g., an amine, azide or alkyne.
  • Certain embodiments of compounds of structure (I) comprise Q groups commonly employed in the field of bioconjugation.
  • Q comprises a nucleophilic reactive group, an electrophilic reactive group or a cycloaddition reactive group.
  • Q comprises a sulfhydryl, disulfide, activated ester, isothiocyanate, azide, alkyne, alkene, diene, dienophile, acid halide, sulfonyl halide, phosphine, ⁇ -haloamide, biotin, amino or maleimide functional group.
  • the activated ester is an N-succinimide ester, imidoester or polyfluorophenyl ester.
  • the alkyne is an alkyl azide or acyl azide.
  • Q groups can be conveniently provided in protected form to increase storage stability or other desired properties, and then the protecting group removed at the appropriate time for conjugation with, for example, a targeting moiety or analyte.
  • Q groups include "protected forms" of a reactive group, including any of the reactive groups described above and in the Table 1 below.
  • a "protected form” of Q refers to a moiety having lower reactivity under predetermined reaction conditions relative to Q, but which can be converted to Q under conditions, which preferably do not degrade or react with other portions of the compound of structure (I).
  • One of skill in the art can derive appropriate protected forms of Q based on the particular Q and desired end use and storage conditions.
  • a protected form of Q includes a disulfide, which can be reduced to reveal the SH moiety using commonly known techniques and reagents.
  • Exemplary Q moieties are provided in Table I below. Table 1.
  • Exemplary Q Moieties It should be noted that in some embodiments, wherein Q is SH, the SH moiety will tend to form disulfide bonds with another sulfhydryl group, for example on another compound of structure (I). Accordingly, some embodiments include compounds of structure (I), which are in the form of disulfide dimers, the disulfide bond being derived from SH Q groups.
  • the Q moiety is conveniently masked (e.g., protected) as a disulfide moiety, which can later be reduced to provide an activated Q moiety for binding to a desired analyte molecule or targeting moiety.
  • the Q moiety may be masked as a disulfide having the following structure: wherein R is an optionally substituted alkyl group.
  • Q is provided as a disulfide moiety having the following structure: where n is an integer from 1 to 10.
  • M 2 is, at one or more occurrences, independently a moiety comprising four or more aryl or heteroaryl rings, or combinations thereof.
  • M 2 is, at one or more occurrences, independently fluorescent or colored. In certain embodiments, M 2 is, at one or more occurrences, fluorescent. In other embodiments, M 2 is, at one or more occurrences, independently comprise a fused-multicyclic aryl or heteroaryl moiety comprising at least four fused rings.
  • M 2 is, at each occurrence, independently selected from the group consisting of phenyl, (bis-fluorophenyl-difluorobora- diaza-indacene)phenyl, quaterphenyl, bi-benzothiazole, ter-benzothiazole, bi-naphthyl, bi- anthracyl, squaraine, squarylium, 9,10-ethynylanthracene, and ter-naphthyl moiety.
  • M 2 is, at each occurrence, independently selected from the group consisting of p-terphenyl, perylene, azobenzene, phenazine, phenanthroline, acridine, thioxanthrene, chrysene, rubrene, coronene, cyanine, perylene imide, perylene amide, and derivatives thereof.
  • M 2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, resorufin dye, dipyrrometheneboron difluoride dye, ruthenium bipyridyl dye, thiazole orange dye, polymethine, and N-aryl-1,8-naphthalimide dye.
  • M 2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, boron-dipyrromethene, rhodamine, cyanine, pyrene, perylene, perylene monoimide, 6-carboxyfluorescein (6-FAM), 5-carboxyfluorescein (5-FAM), 6- fluorescein isothiocyanate (6-FITC), 5-fluorescein isothiocyanate (5-FITC), and derivatives thereof.
  • M 2 at each occurrence, independently has one of the following structures:
  • At least one occurrence of M 2 has the following structure: . In some more specific embodiments, each occurrence of M 2 has the following structure: . In some embodiments, at least one occurrence of –L 7 - M 2 has one of the following structures: . In some more specific embodiments, each occurrence of –L 7 - M 2 has one of the following structures: . In some more specific embodiments, each occurrence of –L 7 - M 2 has one of the following structures: . M 1 and M 3 are selected based on the desired alkylating properties.
  • M 1 and M 3 are the same at each occurrence; however, it is important to note that each occurrence of M 1 and M 3 need not be an identical M 1 and M 3 , and certain embodiments include compounds wherein M 1 and M 3 are not the same at each occurrence.
  • each M 1 and M 3 are not the same and the different M 1 and M 3 moieties are selected to have different alkylating agents.
  • Exemplary M 1 and M 3 moieties can be appropriately selected by one of ordinary skill in the art based on the desired end use.
  • alkylating agents are protected by protecting groups such as an allyloxycarbonyl group (–Alloc) and a tert-butyl dimethyl silyl ether (-TBS) in order to survive during the DNA synthesis cycle.
  • Alloc protecting group can be easily cleaved by a palladium catalyst, for example, Pd(PPh 3 ) 4 with PhSiH 3 to afford the corresponding amine.
  • TBS protecting group can be cleaved by a fluoride source such as tetra-n-butylammonium fluoride (TBAF) to afford the corresponding alcohol.
  • M 1 or M 3 moieties are an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
  • each occurrence of M 1 or M 3 is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
  • At least one occurrence of M 1 or M 3 is a nitrogen mustard, a nitrosourea, a tetrazine, an aziridine, a cisplatin or cisplatin derivative, or a non-classical alkylating agent.
  • At least one occurrence of M 1 or M 3 is mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide, busulfan, N-nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU), semustine (MeCCNU), fotemustine, streptozotocin, dacarbazine, mitozolomide, temozolomide, thiotepa, mytomycin, diaziquone (AZQ), cisplatin, carboplatin, oxaliplatin, procarbazine, or hexamethylmelamine.
  • MNU N-nitroso-N-methylurea
  • BCNU carmustine
  • CCNU lomustine
  • Semustine MeCCNU
  • fotemustine streptozotocin
  • dacarbazine mitozolomide
  • temozolomide temozolomide
  • At least one occurrence of M 1 or M 3 is an anti-folate, a fluoropyrimidines, a deoxynucleoside analogue, or a thiopurine.
  • at least one occurrence of M 1 or M 3 is methotrexate, pemetrexed, fluorouracil, capecitabine, cytarabine, gemcitabine, decitabine, azacitidine, fludarabine, nelarabine, cladribine, clofarabine, pentostatin, thioguanine, and mercaptopurine.
  • At least one occurrence of M 1 or M 3 is an auristatin, a Vinca alkaloid, or a taxane. In certain specific embodiments, at least one occurrence of M 1 or M 3 is auristatin F, auristatin E, vincristine, vinblastine, vinorelbine, vindesine, vinflunine, paclitaxel, docetaxel, etoposide, or teniposide. In some more specific embodiments, at least one occurrence of M 1 or M 3 is irinotecan, SN 38, topotecan, camptothecin, doxorubicin, mitoxantrone, teniposide.
  • At least one occurrence of M 1 or M 3 is an anthracycline or a bleomycin. In some embodiments, at least one occurrence of M 1 or M 3 is doxorubicin, daunorubicin, epirubicin, idarubicin, pirarubicin, aclarubicin, or mitoxantrone.
  • At least one occurrence of M 1 or M 3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan.
  • each occurrence of M 1 or M 3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan.
  • M 1 or M 3 may be attached to the remainder of the molecule from any position (i.e., atom) on M 1 or M 3 , respectively.
  • One of skill in the art will recognize means for attaching M 1 or M 3 to the remainder of molecule.
  • M 1 or M 3 may be attached to the remainder of the molecule through nitrogen of diazepine, oxygen of diazepine or phenyl ring, or carbon of pyrrolidine ring.
  • at least one occurrence of M 1 or M 3 has one of the following structures: ; ; ; ; ; ; ; or .
  • each occurrence of M 1 or M 3 has one of the following structures: ; ; ; ; ; ; or .
  • the compound is a compound selected from Table 2A.
  • the compounds in Table 2A are prepared according to the procedures set forth in the Examples.
  • M 3 has the following structure: , wherein M 1 has one of the following structures: ; ; ; ; ; ; or . Further in some embodiments, at least one occurrence of M 3 is an anti-inflammatory compound.
  • M 3 is a steroid which is a biologically active organic compound with four rings arranged in specific molecular configuration as shown below.
  • At least one occurrence of M 3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone.
  • At least one occurrence of M 3 has one of the following structures: or , wherein: R 11 is H or halogen; R 12 and R 13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R 14 is H, halogen, OH, or alkyl; R 15 is H or OH; R 16 , R 17 , and R 18 , are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond.
  • at least one occurrence of M 3 has one of the following structures: ; ; ; ; ; ; ; ; or .
  • the compound is a compound selected from Table 2B. The compounds in Table 2B are prepared according to the procedures set forth in the Examples.
  • M 3 has one of the following structures: ; ; ; ; or , wherein or is selected from the following structures: ; ; ; ; ; ; ; ; or , wherein M 1 has one of the following structures: ; ; ; ; ; ; ; ; or .
  • at least one occurrence of M 3 is a Vitamin.
  • at least one occurrence of M 3 is a Vitamin E.
  • at least one occurrence of M 3 is a tocopherol.
  • At least one occurrence of M 3 is ⁇ -tocopherol, ⁇ -tocopherol, ⁇ -tocopherol, ⁇ -tocopherol, ⁇ -tocotrienol, ⁇ - tocotrienol, ⁇ -tocotrienol, or ⁇ -tocotrienol.
  • at least one occurrence of M 3 has the following structure: , wherein: R 19 and R 20 are independently H or CH3; and represents a single or double carbon-carbon bond.
  • at least one occurrence of M 3 has one of the following structures: ; ; ; ; ; ; or .
  • the compound is a compound selected from Table 2C. The compounds in Table 2C are prepared according to the procedures set forth in the Examples.
  • M 3 has one of the following structures: ; ; ; ; or , wherein is selected from the following structures: ; ; ; ; ; ; or , wherein M 1 has one of the following structures: ; ; ; ; ; ; ; ; or .
  • the compounds of structure (III) are selected from Table 2D.
  • the compounds in Table 2D are prepared according to the procedures set forth in the Examples.
  • the compounds of structure (IV) are selected from Table 2E.
  • the compounds in Table 2E are prepared according to the procedures set forth in the Examples.
  • n is an integer of one or greater for structure (I). In certain embodiments, n is an integer from 1 to 100 for structure (I). In other embodiments, n is an integer from 1 to 10 for structure (I). In some embodiments, n is 1 for structure (I). In some embodiments, n is 2 for structure (I).
  • n is 3 for structure (I). In some embodiments, n is 4 for structure (I). In some embodiments, n is 5 for structure (I). In some embodiments, n is 6 for structure (I). In some embodiments, n is 7 for structure (I). In some embodiments, n is 8 for structure (I). In some embodiments, n is 9 for structure (I). In some embodiments, n is 10 for structure (I). In some embodiments of structures (III) and (IV), n is an integer of two or greater. In certain embodiments, n is an integer from 2 to 100 for structures (III) and (IV). In other embodiments, n is an integer from 2 to 10 for structures (III) and (IV).
  • n is 2 for structures (III) and (IV). In some embodiments, n is 3 for structures (III) and (IV). In some embodiments, n is 4 for structures (III) and (IV). In some embodiments, n is 5 for structures (III) and (IV). In some embodiments, n is 6 for structures (III) and (IV). In some embodiments, n is 7 for structures (III) and (IV). In some embodiments, n is 8 for structures (III) and (IV). In some embodiments, n is 9 for structures (III) and (IV). In some embodiments, n is 10 for structures (III) and (IV). The fluorescence or the effectiveness toward alkylation of DNA may also be tuned by selection of values for m.
  • m has the ability to control the spacing between neighboring M 1 , M 2 , or M 3 .
  • m is an integer of zero or greater. In certain embodiments, m is an integer from 0 to 100. In certain embodiments, m is an integer from 0 to 10. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments, m is 5. In some embodiments, m is 6. In some embodiments, m is 7. In some embodiments, m is 8. In some embodiments, m is 9. In some embodiments, m is 10. In some certain embodiments, m is 1 or 2.
  • the fluorescence intensity may be tuned how many fluorescent dye moieties are attached to the polymeric backbone.
  • the value for q has the ability to control the brightness of compounds.
  • q is, at each occurrence, an integer of zero or greater. In some more specific embodiments, q is from 0 to 10. In some embodiments, q is from 0 to 5. For example, in some embodiments, q is 0. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, q is 3. In some embodiments, q is 4. In some embodiments, q is 5. In some other embodiments, q is 0, 1, 2, 3, or 4. In some other embodiments, q is 1 or 2.
  • the effectiveness toward alkylation of DNA may also be tuned by selection of values for p.
  • p is, at each occurrence, an integer of zero or greater. In some more specific embodiments, p is from 0 to 10. In some embodiments, p is from 0 to 5. For example, in some embodiments, p is 0. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, p is 4. In some embodiments, p is 5. In some other embodiments, p is 1, 2, 3, or 4. In some other embodiments, p is 2 or 3.
  • the effectiveness toward alkylation of DNA may also be tuned by selection of values for w. In some embodiments, w is, at each occurrence, an integer of zero or greater. In some more specific embodiments, w is from 0 to 10.
  • w is from 0 to 5. In some embodiments, w is from 1 to 5. For example, in some embodiments, w is 1. In some embodiments, w is 2. In some embodiments, w is 3. In some embodiments, w is 4. In some embodiments, w is 5. In some other embodiments, w is 1, 2, 3, or 4. In some other embodiments, w is 1 or 2.
  • v is an integer of one or greater. For example, in some embodiments, v is an integer from 1 to 100. Further in some embodiments, v is an integer from 1 to 10. In some embodiments, v is 1. In some embodiments, v is 2. In some embodiments, v is 3. In some embodiments, v is 4.
  • v is 5. In some embodiments, v is 6. In some embodiments, v is 7. In some embodiments, v is 8. In some embodiments, v is 9. In some embodiments, v is 10.
  • the value n, q, w, p, and m are closely related to provide the ability to control the fluorescence and effectiveness toward alkylation of DNA. In some specific embodiments, n is 1; q is 2; w is 2, p is 2; and m is 1. In some other specific embodiments, n is 1; q is 2; w is 1, p is 3; and m is 1.
  • the ratio of a biological active moiety on M 1 , a fluorescent dye M 2 , and a biological active moiety on M 3 can be fine-tuned based on a type of disease, progression of the disease, a therapeutic method, and/or diagnostic method used for a treatment.
  • a ratio of M 1 :M 2 :M 3 is 1:1:1.
  • M 1 :M 2 :M 3 is 1:2:3.
  • M 1 :M 2 :M 3 is 2:1:1.
  • M 1 :M 2 :M 3 is 1:0:1 when q is 0.
  • M 1 :M 2 :M 3 is 1:0:2 when q is 0.
  • M 1 :M 2 :M 3 is 2:0:1 when q is 0.
  • Pharmaceutical Compositions One embodiment provides a composition comprising the compound according to any one of the embodiments disclosed herein (e.g., a compound of Structure (I), (III), or (IV)) and a pharmaceutically acceptable carrier. Other embodiments are directed to pharmaceutical compositions.
  • the pharmaceutical composition comprises any one (or more) of the compounds of Structure (I), (III), or (IV) and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is formulated for oral administration. In other embodiments, the pharmaceutical composition is formulated for injection.
  • the pharmaceutical compositions comprise a compound of Structure (I), (III), or (IV) and an additional therapeutic agent (e.g., anticancer agent).
  • additional therapeutic agent e.g., anticancer agent
  • suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • a compound of Structure (I), (III), or (IV) is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation.
  • long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the drug is delivered in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody. In such embodiments, the liposomes are targeted to and taken up selectively by the organ.
  • the compound of Structure (I), (III), or (IV) is provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation.
  • the compound of Structure (I), (III), or (IV) is administered topically.
  • the compounds of Structure (I), (III), or (IV) are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that are used in some embodiments. An exemplary dosage is 10 to 30 mg per day.
  • a compound of Structure (I), (III), or (IV) is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes are used as appropriate.
  • a single dose of a compound of Structure (I), (III), or (IV) may also be used for treatment of an acute condition.
  • a compound of Structure (I), (III), or (IV) is administered in multiple doses.
  • dosing is about once, twice, three times, four times, five times, six times, or more than six times per day. In other embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day.
  • a compound of Structure (I), (III), or (IV) and another agent are administered together about once per day to about 6 times per day.
  • the administration of a compound of Structure (I), (III), or (IV) and an agent continues for less than about 7 days.
  • the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • a compound of Structure (I) is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, a compound of Structure (I), (III), or (IV) is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, a compound of Structure (I), (III), or (IV) is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects. In some embodiments, the compounds of Structure (I), (III), or (IV) are administered in dosages. It is known in the art that due to inter-subject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy.
  • the compounds of Structure (I), (III), or (IV) are formulated into pharmaceutical compositions.
  • pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • compositions comprising a compound of Structure (I), (III), or (IV) and a pharmaceutically acceptable diluent(s), excipient(s), or carrier(s).
  • the compounds described are administered as pharmaceutical compositions in which compounds of Structure (I), (III), or (IV) are mixed with other active ingredients, as in combination therapy.
  • the pharmaceutical compositions include one or more compounds of Structure (I), (III), or (IV).
  • a pharmaceutical composition refers to a mixture of a compound of Structure (I), (III), or (IV) with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • the pharmaceutical composition facilitates administration of the compound to an organism.
  • therapeutically effective amounts of compounds of Structure (I), (III), or (IV) provided herein are administered in a pharmaceutical composition to a mammal having a disease, disorder or medical condition to be treated.
  • the mammal is a human.
  • therapeutically effective amounts vary depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors.
  • the compounds of Structure (I), (III), or (IV) are used singly or in combination with one or more therapeutic agents as components of mixtures.
  • one or more compounds of Structure (I), (III), or (IV) is formulated in an aqueous solution.
  • the aqueous solution is selected from, by way of example only, a physiologically compatible buffer, such as Hank’s solution, Ringer’s solution, or physiological saline buffer.
  • one or more compound of Structure (I), (III), or (IV) is/are formulated for transmucosal administration.
  • transmucosal formulations include penetrants that are appropriate to the barrier to be permeated.
  • appropriate formulations include aqueous or non-aqueous solutions.
  • such solutions include physiologically compatible buffers and/or excipients.
  • compounds described herein are formulated for oral administration. Compounds described herein are formulated by combining the active compounds with, e.g., pharmaceutically acceptable carriers or excipients.
  • the compounds described herein are formulated in oral dosage forms that include, by way of example only, tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like.
  • pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate.
  • disintegrating agents are optionally added.
  • Disintegrating agents include, by way of example only, cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • dosage forms such as dragee cores and tablets, are provided with one or more suitable coating.
  • concentrated sugar solutions are used for coating the dosage form.
  • the sugar solutions optionally contain additional components, such as by way of example only, gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs and/or pigments are also optionally added to the coatings for identification purposes.
  • the dyestuffs and/or pigments are optionally utilized to characterize different combinations of active compound doses.
  • therapeutically effective amounts of at least one of the compounds described herein are formulated into other oral dosage forms.
  • Oral dosage forms include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • push-fit capsules contain the active ingredients in admixture with one or more filler. Fillers include, by way of example only, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • soft capsules contain one or more active compound that is dissolved or suspended in a suitable liquid.
  • suitable liquids include, by way of example only, one or more fatty oil, liquid paraffin, or liquid polyethylene glycol.
  • stabilizers are optionally added.
  • therapeutically effective amounts of at least one of the compounds described herein are formulated for buccal or sublingual administration.
  • Formulations suitable for buccal or sublingual administration include, by way of example only, tablets, lozenges, or gels.
  • the compounds described herein are formulated for parental injection, including formulations suitable for bolus injection or continuous infusion.
  • formulations for injection are presented in unit dosage form (e.g., in ampoules) or in multi-dose containers.
  • Preservatives are, optionally, added to the injection formulations.
  • the pharmaceutical compositions are formulated in a form suitable for parenteral injection as sterile suspensions, solutions or emulsions in oily or aqueous vehicles.
  • Parenteral injection formulations optionally contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
  • suspensions of the active compounds e.g., compounds of Structure (I), (III), or (IV) are prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles for use in the pharmaceutical compositions described herein include, by way of example only, fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension contains suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the compounds of Structure (I), (III), or (IV) are administered topically.
  • the compounds described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
  • Such pharmaceutical compositions optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • the compounds of Structure (I), (III), or (IV) are formulated for transdermal administration.
  • transdermal formulations employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive.
  • patches are constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • the transdermal delivery of the compounds of Structure (I), (III), or (IV) is accomplished by means of iontophoretic patches and the like.
  • transdermal patches provide controlled delivery of the compounds of Structure (I), (III), or (IV).
  • the rate of absorption is slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel.
  • absorption enhancers are used to increase absorption.
  • Absorption enhancers or carriers include absorbable pharmaceutically acceptable solvents that assist passage through the skin.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • the compounds of Structure (I), (III), or (IV) are formulated for administration by inhalation.
  • Various forms suitable for administration by inhalation include, but are not limited to, aerosols, mists or powders.
  • compositions of any of compound of Structure (I), (III), or (IV) are 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).
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit of a pressurized aerosol is determined by providing a valve to deliver a metered amount.
  • capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator is formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • a powder mix of the compound such as lactose or starch.
  • the compounds of Structure (I), (III), or (IV) are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like.
  • a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with melted cocoa butter.
  • pharmaceutical compositions are formulated in any conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are optionally used as suitable.
  • compositions comprising a compound of Structure (I), (III), or (IV) are manufactured in a conventional manner, such as, by way of example only, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • Pharmaceutical compositions include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of Structure (I), (III), or (IV), described herein as an active ingredient.
  • the active ingredient is in free-acid or free-base form, or in a pharmaceutically acceptable salt form.
  • compositions described herein include the use of N-oxides, crystalline forms (also known as polymorphs), as well as active metabolites of these compounds having the same type of activity. All tautomers of the compounds described herein are included within the scope of the compounds presented herein. Additionally, the compounds described herein encompass unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • compositions optionally include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, buffers, and/or other therapeutically valuable substances.
  • Methods for the preparation of compositions comprising the compounds described herein include formulating the compounds with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid or liquid.
  • Solid compositions include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • Liquid compositions include solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein.
  • Semi-solid compositions include, but are not limited to, gels, suspensions and creams.
  • the form of the pharmaceutical compositions described herein include liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions also optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth.
  • composition comprising at least one compound of Structure (I), (III), or (IV) illustratively takes the form of a liquid where the agents are present in solution, in suspension or both.
  • a liquid composition includes a gel formulation.
  • the liquid composition is aqueous.
  • useful aqueous suspensions contain one or more polymers as suspending agents.
  • Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross- linked carboxyl-containing polymers.
  • Certain pharmaceutical compositions described herein comprise a mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
  • Useful pharmaceutical compositions also, optionally, include solubilizing agents to aid in the solubility of a compound of Structure (I), (III), or (IV).
  • solubilizing agent generally includes agents that result in formation of a micellar solution or a true solution of the agent.
  • Certain acceptable nonionic surfactants for example polysorbate 80, are useful as solubilizing agents, as can ophthalmically acceptable glycols, polyglycols, e.g., polyethylene glycol 400, and glycol ethers.
  • useful pharmaceutical compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
  • useful compositions also, optionally, include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • Other useful pharmaceutical compositions optionally include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
  • Still other useful compositions include one or more surfactants to enhance physical stability or for other purposes.
  • Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40.
  • Still other useful compositions include one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example only, ascorbic acid and sodium metabisulfite.
  • aqueous suspension compositions are packaged in single-dose non-reclosable containers.
  • multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
  • other delivery systems for hydrophobic pharmaceutical compounds are employed. Liposomes and emulsions are examples of delivery vehicles or carriers useful herein.
  • organic solvents such as N-methylpyrrolidone are also employed.
  • the compounds described herein are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials are useful herein. In some embodiments, sustained-release capsules release the compounds for a few weeks up to over 100 days.
  • the formulations described herein comprise one or more antioxidants, metal chelating agents, thiol containing compounds and/or other general stabilizing agents.
  • stabilizing agents include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1% w/v methionine, (c) about 0.1% to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v.
  • polysorbate 20 (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (l) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.
  • the concentration of one or more compounds provided in the pharmaceutical compositions is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%,14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v or v/v.
  • the concentration of one or more compounds is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%, 4.25%, 4%,
  • the concentration of one or more compounds is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v.
  • the concentration of one or more compounds is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
  • the amount of one or more compounds is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.008 g
  • the amount of one or more compounds is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075 g, 0.
  • the amount of one or more compounds ranges from 0.0001 to 10 g, 0.0005 to 9 g, 0.001 to 8 g, 0.005 to 7 g, 0.01 to 6 g, 0.05 to 5 g, 0.1 to 4 g, 0.5 to 4 g, or 1 to 3 g.
  • Method of Treatment Certain compounds of the present disclosure are useful for treating disease (i.e., compounds of Structure (I), (III), or (IV)). Those compounds disclosed herein offer a targeted approach to drug delivery strategies.
  • one embodiment provides a method of treating a disease (or the symptoms thereof) comprising administering to a mammal (e.g., a human) in need thereof a therapeutically effective amount of a compound of Structure (I), (III), or (IV).
  • a mammal e.g., a human
  • a therapeutically effective amount of a compound of Structure (I), (III), or (IV) comprising administering to a mammal (e.g., a human) in need thereof a therapeutically effective amount of a compound of Structure (I), (III), or (IV).
  • the disclosure provides a method of treating solid tumors, multiple myeloma, gliomas, clear cell renal cell carcinoma, prostate cancer, ovarian cancer, non-small cell lung cancer, GI malignancies, acute lymphoblastic leukemia, acute myelogenous leukemia, renal cell carcinoma, colorectal carcinoma, epithelial cancers, pancreatic and gastric cancers, renal cell carcinoma, non-Hodgkin’s lymphoma, metastatic renal cell carcinoma, malignant mesothelioma, pancreatic, ovarian, and/or lung adenocarcinoma, B-cell malignancies, breast cancer, melanoma, recurrent multiple myeloma, small cell lung cancer, CD22-positive B cell malignancies, Hodgkin’s lymphoma/anaplastic large cell lymphoma, or HER2-positive breast cancer.
  • GI malignancies acute lymphoblastic leukemia, acute myelogenous leukemia, renal cell carcinoma, color
  • the disease is cancer.
  • the cancer is breast cancer, non-Hodgkin's lymphoma, acute myeloid leukemia, multiple myeloma, gastric cancer, renal cell carcinoma, solid tumors, ovarian cancer, prostate cancer, colorectal cancer, pancreatic cancer, small cell lung cancer, diffuse large B-cell lymphoma, a neoplasm, urothelial cancer, ALL, CLL, glioblastoma, Hodgkin's lymphoma, lymphoma, mesothelioma, non-small cell lung cancer, recurrent head and neck cancer, or a combination thereof.
  • Certain embodiments also relate to a method of treating a hyperproliferative disorder in a mammal (e.g., a human) that comprises administering to said mammal a therapeutically effective amount of a compound of Structure (I), (III), or (IV), or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • a mammal e.g., a human
  • administering to said mammal a therapeutically effective amount of a compound of Structure (I), (III), or (IV), or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • said method relates to the treatment of cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g., Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal
  • cancer such
  • said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
  • the lung cancer is a small cell lung carcinoma.
  • Other lung cancers treatable with the disclosed compounds include, but are not limited to, glandular tumors, carcinoid tumors and undifferentiated carcinomas.
  • A is an antibody or a cell surface receptor antagonist.
  • EGFR epidermal growth factor receptor
  • HGFR hepatocyte growth factor receptor
  • IGFR insulin-like growth factor receptor
  • the method further comprises inducing apoptosis.
  • the method of treatment comprises treating a tumor having tumor cells with tumor cell receptors.
  • the tumor cells have receptors ranging from 1,000 to 100,000, from 1,000 to 50,000, from 1,000 to 25,000 receptors, 1,000 to 10,000 receptors per cell.
  • the tumor cells have about 1,000, about 10,000, or less than 100,000 receptors per cell.
  • Further therapeutic agents that can be combined with a compound of the disclosure are found in Goodman and Gilman’s "The Pharmacological Basis of Therapeutics" Tenth Edition edited by Hardman, Limbird and Gilman or the Physician’s Desk Reference, both of which are incorporated herein by reference in their entirety.
  • the compounds of Structure (I), (III), or (IV) described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated.
  • the one or more compounds of the disclosure will be co-administered with other agents as described above.
  • the compounds described herein are administered with the second agent simultaneously or separately.
  • This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously.
  • a compound of the disclosure and any of the agents described above can be simultaneously administered, wherein both the agents are present in separate formulations.
  • a compound of the present disclosure can be administered just followed by and any of the agents described above, or vice versa.
  • a compound of the disclosure and any of the agents described above are administered a few minutes apart, or a few hours apart, or a few days apart.
  • the method further comprises administering an additional therapeutic agent selected from the group consisting of an antineoplastic agent, an enediyne antitumor antibiotic, a maytansinoid, a topoisomerase inhibitor, a kinase inhibitor, an anthracycline, and EGFR inhibitor, an alkylating agent and combinations thereof.
  • the method further comprises administering an additional therapeutic agent selected from the group consisting of an antineoplastic agent, an enediyne antitumor antibiotic, a maytansinoid, a topoisomerase inhibitor, a kinase inhibitor, an anthracycline, and EGFR inhibitor, an alkylating agent and combinations thereof.
  • an additional therapeutic agent selected from the group consisting of an antineoplastic agent, an enediyne antitumor antibiotic, a maytansinoid, a topoisomerase inhibitor, a kinase inhibitor, an anthracycline, and EGFR inhibitor, an alkylating agent and combinations thereof.
  • the additional therapeutic agent comprises auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan.
  • auristatin F monomethyl auristatin F
  • monomethyl auristatin E monomethyl auristatin E
  • paciltaxol SN-38
  • calicheamicin anthramycin, abbeymycin, chica
  • a pharmaceutical composition comprising the compound of any of the forementioned structures, and a pharmaceutically acceptable carrier, diluent, or excipient.
  • a method of treating a disease or disorder comprising administering a therapeutically effective amount of a compound of any of the forementioned structures, or the pharmaceutical composition of the same, to a subject in need thereof.
  • the disease or disorder is cancer.
  • the cancer is breast cancer, stomach cancer, lung cancer, ovarian cancer, lymphoma, and bladder cancer.
  • At least one occurrence of L 4 is heteroalkylene. In some embodiments, each occurrence of L 4 is heteroalkylene. In some embodiments, at least one occurrence of L 4 comprises alkylene oxide. In some more specific embodiments, the alkylene oxide of L 4 is ethylene oxide. In some more specific embodiments, the ethylene oxide is polyethylene oxide. In certain embodiments, L 4 , at each occurrence, has the following structure: , wherein: z is an integer from 1 to 100; and * indicates a bond to the adjacent phosphorous atom. In some embodiments, z is an integer from 3 to 8. In some embodiments, z is 3. In some embodiments, z is 4. In some embodiments, z is 5. In some embodiments, z is 6.
  • z is 7. In some embodiments, z is 8. In some other embodiments, z is an integer from 22 to 26. In some embodiments, z is 22. In some embodiments, z is 23. In some embodiments, z is 24. In some embodiments, z is 25. In some embodiments, z is 26. In some other embodiments, z ranges from 19 to 28. In certain embodiments, the average z is 23. In some embodiments, the average z is 19, 20, 21, 22, 23, 24, 25, 26, 27, or 28. In other embodiments, at least one occurrence of L 4 is an alkylene linker (e.g., methylene).
  • alkylene linker e.g., methylene
  • L 4 is an alkylene linker at each occurrence (e.g., methylene).
  • z is an integer from 3 to 8.
  • z is an integer from 22 to 26.
  • z ranges from 19 to 28.
  • z is an integer from 3 to 8.
  • z is an integer from 22 to 26. In some other embodiments, z ranges from 19 to 28.
  • Some embodiments provide a compound having the following structure (IIA): (IIA) or a stereoisomer, salt or tautomer thereof.
  • L 3 , L 5 , and L 6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers. In some embodiments, L 3 , L 5 or L 6 are, at each occurrence, independently alkylene.
  • L 3 , L 5 or L 6 are, at each occurrence, independently C 1 -C 6 alkylene, C2-C6 alkenylene or C2-C6 alkynylene.
  • the alkylene linker of L 3 and L 5 are C1 alkyl linker.
  • the alkylene linker of L 3 and L 5 are C2 alkyl linker.
  • the alkylene linker of L 3 and L 5 are C3 alkyl linker.
  • the alkylene linker of L 3 and L 5 are C4 alkyl linker.
  • the alkylene linker of L 3 and L 5 are C5 alkyl linker.
  • the alkylene linker of L 3 and L 5 are C6 alkyl linker. In some specific embodiments, the alkylene linker of L 6 are C1 alkyl linker. In some specific embodiments, the alkylene linker of L 6 are C2 alkyl linker. In some specific embodiments, the alkylene linker of L 6 are C3 alkyl linker. In other embodiments, at least one occurrence of L 6 is a direct bond. In some other embodiments, each occurrence of L 6 is a direct bond.
  • Some embodiments provide a compound having the following structure (IIB): (IIB) or a stereoisomer, salt or tautomer thereof, wherein: y 1 is, at each occurrence, independently an integer from 0 to 6; and y 2 and y 3 are, at each occurrence, independently an integer from 1 to 6.
  • y 1 is, at each occurrence, an integer 0 (i.e., L 6 is a direct bond).
  • y 1 is, at each occurrence, an integer 1.
  • y 1 is, at each occurrence, an integer 2.
  • y 1 is, at each occurrence, an integer 3.
  • y 1 is, at each occurrence, an integer 4.
  • y 1 is, at each occurrence, an integer 5. In some embodiments, y 1 is, at each occurrence, an integer 6. In some embodiments, y 2 and y 3 are at each occurrence, an integer 1 In some embodiments, y 2 and y 3 are at each occurrence, an integer 2. In some embodiments, y 2 and y 3 are at each occurrence, an integer 3. In some embodiments, y 2 and y 3 are at each occurrence, an integer 4. In some embodiments, y 2 and y 3 are at each occurrence, an integer 5. In some embodiments, y 2 and y 3 are at each occurrence, an integer 6.
  • y 1 is, at each occurrence, an integer 0 or 1; and y 2 , and y 3 are, at each occurrence, 1.
  • compounds of (III) can be prepared by oligomerization using well known phosphoramidite chemistry. Applicants have discovered intermediate compounds (V) useful for synthesis of compounds of structures (III).
  • some embodiments provide intermediate compounds having structure (V): (V) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: G 2 is, at each occurrence, independently a moiety reactive under cycloaddition; G 3 is, at each occurrence, independently a moiety reactive under cycloaddition; L 1a is, at each occurrence, independently a heteroalkylene or heteroarylene linker; L 1b is, at each occurrence, independently either i) absent or ii) a linker; L 7 is, at each occurrence, independently a linker; L 2 , L 3 , L 5 and L 6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; R 21 is, at each occurrence, independently a natural or unnatural amino acid side chain; R 1 and R 2 are each independently H, OH, SH, –NH 2
  • Compounds of structure (V) have a peptide-based backbone.
  • the various linkers and substituents e.g., G 2 , G 3 , Q, R 1 , R 2 , R 21 , L 2 , L 3 , L 5 , L 6 , L 7 and L'
  • the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (V).
  • each alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (V) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether, alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether.
  • substituents are selected to increase cellular or tissue permeation.
  • substituents are selected to increase cellular or tissue retention.
  • at least one R 21 is a neutral amino acid side chain.
  • at least one R 21 is a charged amino acid side chain.
  • R 21 , L 5 and m are selected such that has an amino acid sequence of (G)10, (GDGDGDGDGD) or (GKGKGKGKGK).
  • R 21 , L 5 and m are selected such that has an amino acid sequence capable of forming an ⁇ -helix or ⁇ -sheet secondary structure.
  • the amino acid sequence is (GGEEFMLVYKFARKHGG) or (GGMSMVVSGG).
  • L 5 or L 3 , or both is present for at least one occurrence.
  • L 5 or L 3 , or both is a heteroalkylene linker.
  • the heteroalkylene linker comprises a functional group capable of maintaining a positive or negative charge at pH values ranging from 3 to 11 in aqueous solution.
  • At least one occurrence of L 5 or L 3 , or both has the following structure: . In some embodiments, at least one occurrence of L 5 or L 3 , or both, has the following structure: . In some embodiments, L 6 and L 2 are independently absent or a heteroalkylene linker. In more specific embodiments, the heteroalkylene linker is a peptidyl linker.
  • compounds of (IV) can be prepared by oligomerization using well known phosphoramidite chemistry. Applicants have discovered intermediate compounds (VI) useful for synthesis of compounds of structures (IV).
  • some embodiments provide a compound having structure (VI): (VI) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: G 2 is, at each occurrence, independently a moiety reactive under cycloaddition; G 3 is, at each occurrence, independently a moiety reactive under cycloaddition; L 7 is, at each occurrence, independently a linker; L 2 , L 5 and L 6 are, at each occurrence, independently a direct bond or independently an optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker; L 8 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker comprising one or more charged moieties, provided at least one charged moiety is not a phosphate ester; R 3 is, at each occurrence, independently H, alkyl or alk
  • the various linkers and substituents e.g., G 2 , G 3 , Q, R 1 , R 2 , R 3 , L 2 , L 5 , L 6 , L 7 , L 8 , and L'
  • the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (VI).
  • each alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker or alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (VI) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether, alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether.
  • substituents are selected to increase cellular or tissue permeation.
  • substituents are selected to increase cellular or tissue retention.
  • the alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl and alkyloxycarbonyl are optionally substituted with hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, or combinations thereof
  • the charged moieties are positively charged.
  • the charged moieties comprise a protonated amine or quaternary amine functional group.
  • the charged moieties independently have one of the following structures: ; or wherein: R is, at each occurrence, independently H or C 1 -C 6 alkyl.
  • the charged moieties are negatively charged.
  • the charged moieties comprise a carboxylic acid, phosphate or sulfate functional group.
  • the charged moieties have the following structure: .
  • the charged moieties comprise a combination of positively charged and negatively charged moieties.
  • the charged moieties are pendant to the backbone of the compound (e.g., attached to a linker via an alkylene or heteroalkylene linker).
  • the charged moiety is part of the backbone of the compound (e.g., part of the contiguous chain of the linker).
  • L 8 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; or ’ wherein: R is, at each occurrence, independently H or C 1 -C 6 alkyl; x is an integer from 0 to 6; and m is an integer of 1 or greater, provided that m is selected such that the compound comprises at least two charged moieties.
  • the linker L 7 can be used as a point of attachment of the M 2 and M 3 moieties or G 2 and G 3 moieties in case of structure (V) and (VI) to the remainder of the compound.
  • a synthetic precursor (II) to the compound of structure (I) is prepared, and the M 2 and M 3 moieties are attached to the synthetic precursor using any number of coupling methods known in the art.
  • At least one occurrence of L 7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof.
  • the functional group comprises an amide or an ester.
  • at least one occurrence of L 7 comprises one of the following structures: ; ; ; ; ; ; ; ; ; or .
  • each occurrence of L 7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof.
  • the functional group comprises an amide.
  • each occurrence of L 7 comprises one of the following structures: ; ; ; ; ; ; ; ; or .
  • at least one occurrence of R 3 is H.
  • at least one occurrence of R 3 is alkyl.
  • Rc is OL'.
  • Q a targeting moiety
  • an analyte e.g., analyte molecule
  • solid support e.g., a solid support residue
  • nucleoside or a further compound of structure II), (V), or (VI).
  • the linker L' can be any linker suitable for attaching Q, a targeting moiety, an analyte (e.g., analyte molecule), a solid support, a solid support residue, a nucleoside or a further compound of structure (II), (V), or (VI) to the compound of structure (II), (V), or (VI).
  • analyte e.g., analyte molecule
  • solid support e.g., alyte molecule
  • a solid support residue e.g., a further compound of structure (II), (V), or (VI) to the compound of structure (II), (V), or (VI).
  • Advantageously certain embodiments include use of L' moieties selected to increase or optimize water solubility of the compound.
  • L' is a heteroalkylene moiety.
  • L' comprises an alkylene oxide or phosphodiester moiety, or combinations thereof.
  • R 2 is L'.
  • L' is a linker to a targeting moiety.
  • L' is a linker to a targeting moiety, the linker comprising an alkylene oxide or phosphodiester moiety, or combinations thereof.
  • L' has one of the following structures: ; ; ; or wherein: x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer from 1 to 10; R b is H, an electron pair or a counter ion; L'' is the targeting moiety or a linkage to the targeting moiety.
  • x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 1. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 2. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 3. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 4.
  • x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 5. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 6. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 7. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 8.
  • x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 9. In some embodiments, x 1 , x 2 , x 3 , x 4 , x 5 , x 6 , x 7 and x 8 are independently an integer 10.
  • R b is H. In some embodiments, R b is a counter ion. For example, in some embodiments, R b is Na + . In some embodiments, R b is K + .
  • L'' is the targeting moiety. In some other embodiments, L'' is a linkage to the targeting moiety.
  • the targeting moiety is an antibody.
  • the targeting moiety is cell surface receptor antagonist.
  • the antibody or cell surface receptor antagonist is an epidermal growth factor receptor (EGFR) inhibitor, a hepatocyte growth factor receptor (HGFR) inhibitor, an insulin-like growth factor receptor (IGFR) inhibitor, a folate, or a MET inhibitor.
  • the targeting moiety is a monoclonal antibody.
  • the monoclonal antibody is Abciximab, Adalimumab, Alemtuzumab, Alirocumab, Avibactam, Basiliximab, Benralizumab, Bezlotoxumab, Blinatumomab, Brodalumab, Burosumab, Canakinumab, Caplacizumab, Certolizumab pegol, Daclizumab, Denosumab, Dupilumab, Eculizumab, Emicizumab, Erenumab, Evolocumab, Fremanezumab, Galcanezumab, Golimumab, Guselkumab, Ibalizumab, Idarucizumab, Infliximab, Itolizumab, Ixekizumab, Lanadelumab, Lokivetmab, Mepolizumab, Natalizumab, Obiltoxaximab
  • R 1 or R 2 has one of the following structures: ; ; ; ; ; ; ; or , wherein R a is H or a solid support.
  • R 2 has one of the following structures: ; or . .
  • Certain embodiments of compounds of structure (II), (V), or (VI) can be prepared according to solid-phase synthetic methods analogous to those known in the art for preparation of oligonucleotides. Accordingly, in some embodiments, L' is a linkage to a solid support, a solid support residue or a nucleoside.
  • Solid supports comprising an activated deoxythymidine (dT) group are readily available, and in some embodiments can be employed as starting material for preparation of compounds of structure (II), (V), or (VI). Accordingly, in some specific embodiments, R 1 has the following structure: .
  • dT group depicted above is included for ease of synthesis and economic efficiencies only, and is not required.
  • Other solid supports can be used and would result in a different nucleoside or solid support residue being present on L', or the nucleoside or solid support residue can be removed or modified post synthesis.
  • the analyte molecule is a nucleic acid, amino acid or a polymer thereof.
  • the analyte molecule is an enzyme, receptor, receptor ligand, antibody, glycoprotein, aptamer or prion.
  • the targeting moiety is an antibody or cell surface receptor antagonist.
  • the solid support is a polymeric bead or non-polymeric bead.
  • M 2 is, at one or more occurrences, independently a moiety comprising four or more aryl or heteroaryl rings, or combinations thereof. In some embodiments, M 2 is, at one or more occurrences, independently fluorescent or colored. In certain embodiments, M 2 is, at one or more occurrences, fluorescent.
  • M 2 is, at one or more occurrences, independently comprise a fused-multicyclic aryl or heteroaryl moiety comprising at least four fused rings.
  • M 2 is, at each occurrence, independently selected from the group consisting of phenyl, (bis-fluorophenyl-difluorobora- diaza-indacene)phenyl, quaterphenyl, bi-benzothiazole, ter-benzothiazole, bi-naphthyl, bi- anthracyl, squaraine, squarylium, 9,10-ethynylanthracene, and ter-naphthyl moiety.
  • M 2 is, at each occurrence, independently selected from the group consisting of p-terphenyl, perylene, azobenzene, phenazine, phenanthroline, acridine, thioxanthrene, chrysene, rubrene, coronene, cyanine, perylene imide, perylene amide, and derivatives thereof.
  • M 2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, resorufin dye, dipyrrometheneboron difluoride dye, ruthenium bipyridyl dye, thiazole orange dye, polymethine, and N-aryl-1,8-naphthalimide dye.
  • M 2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, boron-dipyrromethene, rhodamine, cyanine, pyrene, perylene, perylene monoimide, 6-carboxyfluorescein (6-FAM), 5-carboxyfluorescein (5-FAM), 6- fluorescein isothiocyanate (6-FITC), 5-fluorescein isothiocyanate (5-FITC), and derivatives thereof.
  • M 2 at each occurrence, independently has one of the following structures: ; ; ; ; ; ; or .
  • at least one occurrence of M 2 has the following structure: .
  • each occurrence of M 2 has the following structure: .
  • at least one occurrence of –L 7 - M 2 has one of the following structures: .
  • each occurrence of –L 7 - M 2 has one of the following structures: .
  • M 3 is selected based on the desired alkylating properties. Exemplary M 3 moiety can be appropriately selected by one of ordinary skill in the art based on the desired end use. In some embodiments, at least one occurrence of M 3 moiety is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
  • each occurrence of M 3 is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
  • at least one occurrence of M 1 or M 3 is a nitrogen mustard, a nitrosourea, a tetrazine, an aziridine, a cisplatin or cisplatin derivative, or a non-classical alkylating agent.
  • At least one occurrence of M 3 is mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide, busulfan, N-nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU), semustine (MeCCNU), fotemustine, streptozotocin, dacarbazine, mitozolomide, temozolomide, thiotepa, mytomycin, diaziquone (AZQ), cisplatin, carboplatin, oxaliplatin, procarbazine, or hexamethylmelamine.
  • MNU N-nitroso-N-methylurea
  • BCNU carmustine
  • CCNU lomustine
  • Semustine MeCCNU
  • fotemustine streptozotocin
  • dacarbazine mitozolomide
  • temozolomide temozolomide
  • thiotepa myto
  • At least one occurrence of M 3 is an anti-folate, a fluoropyrimidines, a deoxynucleoside analogue, or a thiopurine.
  • at least one occurrence of M 3 is methotrexate, pemetrexed, fluorouracil, capecitabine, cytarabine, gemcitabine, decitabine, azacitidine, fludarabine, nelarabine, cladribine, clofarabine, pentostatin, thioguanine, and mercaptopurine.
  • at least one occurrence of M 3 is an auristatin, a Vinca alkaloid, or a taxane.
  • At least one occurrence of M 3 is auristatin F, auristatin E, vincristine, vinblastine, vinorelbine, vindesine, vinflunine, paclitaxel, docetaxel, etoposide, or teniposide.
  • at least one occurrence of M 3 is irinotecan, SN 38, topotecan, camptothecin, doxorubicin, mitoxantrone, teniposide. novobiocin, merbarone, or aclarubicin.
  • at least one occurrence of M 3 is an anthracycline or a bleomycin.
  • At least one occurrence of M 3 is doxorubicin, daunorubicin, epirubicin, idarubicin, pirarubicin, aclarubicin, or mitoxantrone.
  • at least one occurrence of M 3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rub
  • each occurrence of M 3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan.
  • M 3 may be attached to the remainder of the molecule from any position (i.e., atom) on M 3 .
  • One of skill in the art will recognize means for attaching M 3 to the remainder of molecule.
  • M 3 may be attached to the remainder of the molecule through nitrogen of diazepine, oxygen of diazepine or phenyl ring, or carbon of pyrrolidine ring.
  • at least one occurrence of M 3 is an anti-inflammatory compound.
  • M 3 is a steroid which is a biologically active organic compound with four rings arranged in specific molecular configuration as shown below.
  • At least one occurrence of M 3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone.
  • At least one occurrence of M 3 has one of the following structures: or , wherein: R 11 is H or halogen; R 12 and R 13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R 14 is H, halogen, OH, or alkyl; R 15 is H or OH; R 16 , R 17 , and R 18 , are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond.
  • at least one occurrence of M 3 has one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or .
  • each occurrence of M 3 has one of the following structures: ; ; ; ; ; ; ; ; ; or .
  • G, G 2 , or G 3 is a reactive moiety that undergoes the cycloaddition.
  • the cycloaddition is click reaction between an azide and an alkyne.
  • G, G 2 , or G 3 has one of the following structures: ; or .
  • G, G 2 , or G 3 at each occurrence, has the structure of .
  • G, G 2 , or G 3 at each occurrence, has the structure of .
  • the fluorescence intensity or the effectiveness toward alkylation of DNA can also be tuned by selection of different values of n.
  • n is an integer of one or greater.
  • n is an integer from 1 to 100.
  • n is an integer from 1 to 10.
  • n is 1.
  • n is 2.
  • n is 3.
  • n is 4.
  • n is 5.
  • n is 6.
  • n is 7.
  • n is 8.
  • n is 9. In some embodiments, n is 10.
  • the fluorescence or the effectiveness toward alkylation of DNA may also be tuned by selection of values for m.
  • the value for m has the ability to control the spacing between neighboring M 1 , M 2 , or M 3 (or G, G 2 , or G 3 ).
  • m is an integer of zero or greater.
  • m is an integer from 0 to 100.
  • m is an integer from 0 to 10.
  • m is 0.
  • m is 1.
  • m is 2.
  • m is 3.
  • m is 4.
  • m is 5.
  • m is 6.
  • m is 7.
  • m is 8.
  • m is 9. In some embodiments, m is 10.
  • the fluorescence intensity may be tuned how many fluorescent dye moieties are attached to the polymeric backbone.
  • the value for q has the ability to control the brightness of compounds.
  • q is, at each occurrence, an integer of zero or greater. In some more specific embodiments, q is from 0 to 10. In some embodiments, q is from 0 to 5. For example, in some embodiments, q is 0. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, q is 3. In some embodiments, q is 4. In some embodiments, q is 5. In some other embodiments, q is 0, 1, 2, 3, or 4. In some other embodiments, q is 1 or 2.
  • the effectiveness toward alkylation of DNA may also be tuned by selection of values for p.
  • p is, at each occurrence, an integer of zero or greater. In some more specific embodiments, p is from 0 to 10. In some embodiments, p is from 0 to 5. For example, in some embodiments, p is 0. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, p is 4. In some embodiments, p is 5. In some other embodiments, p is 1, 2, 3, or 4. In some other embodiments, p is 2 or 3.
  • the effectiveness toward alkylation of DNA may also be tuned by selection of values for w.
  • w is, at each occurrence, an integer of zero or greater. In some more specific embodiments, w is from 0 to 10. In some embodiments, w is from 0 to 5. In some embodiments, w is from 1 to 5. For example, in some embodiments, w is 1. In some embodiments, w is 2. In some embodiments, w is 3. In some embodiments, w is 4. In some embodiments, w is 5. In some other embodiments, w is 1, 2, 3, or 4. In some other embodiments, w is 1 or 2.
  • the effectiveness toward alkylation of DNA may also be tuned by selection of values for k. In some embodiments, k is, at each occurrence, an integer of zero or greater. In some more specific embodiments, k is from 0 to 10.
  • k is from 0 to 5. For example, in some embodiments, k is 0. In some embodiments, k is 1. In some embodiments, k is 2. In some embodiments, k is 3. In some embodiments, k is 4. In some embodiments, k is 5. In other embodiments, k is 0, 1, 2, 3, or 4. In some other embodiments, k is 0 or 1. In some embodiments of structure (V) or (VI), v is an integer of one or greater. For example, in some embodiments, v is an integer from 1 to 100. Further in some embodiments, v is an integer from 1 to 10. In some embodiments, v is 1. In some embodiments, v is 2. In some embodiments, v is 3. In some embodiments, v is 4.
  • v is 5. In some embodiments, v is 6. In some embodiments, v is 7. In some embodiments, v is 8. In some embodiments, v is 9. In some embodiments, v is 10.
  • the value n, q, w, p, k, and m are closely related to provide the ability to control the fluorescence and effectiveness toward alkylation of DNA. In some specific embodiments, n is 1; q is 2; w is 2, k is 0, p is 2; and m is 1. In some other specific embodiments, n is 1; q is 2; w is 1, k is 0, p is 3; and m is 1.
  • n is 3; q is 1 for the first occurrence, q is 0 for the second occurrence, q is 0 for the third occurrence; w is 0 for the first occurrence, w is 0 for the second occurrence, and w is 1 for the third occurrence; k is 0 for the first occurrence, k is 1 for the second occurrence, and k is 0 for the third occurrence; p is 1 for the first occurrence, p is 0 for the second occurrence, and p is 0 for the third occurrence; and m is 1 for all occurrences.
  • the sum of p and w is an integer of two or greater, and the compound comprises at least two different biologically active moieties.
  • At least one occurrence of R 6 or R 7 is F or H. In other embodiments, at least each occurrence of R 6 and R 7 are F or H. In certain embodiments, at least each occurrence of R 6 and R 7 are F, R 8 is O, R 9 is H, and R 10 is H. In some other embodiments, at least each occurrence of R 6 and R 7 are H, R 8 is O, R 9 is H, and R 10 is H. In some embodiments, x 9 is, at each occurrence, an integer from 1 to 6. For example, in some embodiments, each occurrence of x 9 is 1. In some embodiments, each occurrence of x 9 is 2. In some embodiments, each occurrence of x 9 is 3.
  • each occurrence of x 9 is 4. In some embodiments, each occurrence of x 9 is 5. In some embodiments, each occurrence of x 9 is 6. In some other embodiments, each occurrence of x 9 is 2,4, or 6. In some other embodiments, each occurrence of x 9 is 2 or 4.
  • the compound is a compound selected from Table 3A. The compounds in Table 3A are prepared according to the procedures set forth in the Examples.
  • M 3 has the following structure: .
  • at least one occurrence of M 3 is an anti-inflammatory compound.
  • M 3 is a steroid which is a biologically active organic compound with four rings arranged in specific molecular configuration as shown below.
  • at least one occurrence of M 3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone.
  • At least one occurrence of M 3 has one of the following structures: or , wherein: R 11 is H or halogen; R 12 and R 13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R 14 is H, halogen, OH, or alkyl; R 15 is H or OH; R 16 , R 17 , and R 18 , are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond.
  • at least one occurrence of M 3 has one of the following structures:
  • the compound is a compound selected from Table 3B.
  • the compounds in Table 3B are prepared according to the procedures set forth in the Examples
  • M 3 has one of the following structures: ; ; ; ; or , wherein or is selected from the following structures: ; ; ; ; ; ; ; or .
  • at least one occurrence of M 3 is a Vitamin.
  • at least one occurrence of M 3 is a Vitamin E.
  • at least one occurrence of M 3 is a tocopherol.
  • At least one occurrence of M 3 is ⁇ -tocopherol, ⁇ -tocopherol, ⁇ -tocopherol, ⁇ -tocopherol, ⁇ -tocotrienol, ⁇ - tocotrienol, ⁇ -tocotrienol, or ⁇ -tocotrienol.
  • at least one occurrence of M 3 has the following structure: , wherein R 19 and R 20 are independently H or CH 3 ; and represents a single or double carbon-carbon bond.
  • at least one occurrence of M 3 has one of the following structures: ; ; ; ; ; ; or .
  • the compound is a compound selected from Table 3C. The compounds in Table 3C are prepared according to the procedures set forth in the Examples.
  • M 3 has one of the following structures: ; ; ; ; or , wherein is selected from the following structures: ; ; ; ; ; ; or .
  • the compounds of structure (V) are selected from Table 3D.
  • the compounds in Table 3D are prepared according to the procedures set forth in the Examples.
  • the compounds of structure (VI) are selected from Table 3E.
  • the compounds in Table 3E are prepared according to the procedures set forth in the Examples.
  • a REPRESENTATIVE DNA SYNTHESIS CYCLE Oligomerization is initiated, typically, through the removal of a protecting group (e.g., a dimethoxytrityl group, DMTr) to reveal a free –OH (hydroxyl) group (Step 1, DETRITYLATION).
  • a phosphoramidite monomer is introduced that reacts with the free OH group making a new covalent bond to phosphorus, with concomitant loss of the diisopropyl amine group (Step 2, COUPLING).
  • the resultant, phosphite triester is oxidized (e.g., with I2 and pyridine) to the more stable phosphate ester (Step 3, OXIDATION) and a capping step renders unreactive any remaining free OH groups (Step 4, CAPPING).
  • the new product, phosphate oligomer contains a DMTr protected OH group that can be deprotected to reinitiate the synthetic cycle so another phosphoramidite monomer can be appended to the oligomer. Customization occurs at step 2 through the choice of phosphoramidite monomer.
  • L i.e., a linker group
  • M i.e., a chemotherapeutic agent
  • L i.e., a linker group
  • M i.e., a chemotherapeutic agent
  • L i.e., a linker group
  • M i.e., a chemotherapeutic agent
  • M can be optionally absent to incorporate desired spacing between M moieties.
  • a person of ordinary skill in the art can select multiple monomer types to arrive at compounds of the disclosure containing multiple therapeutic agents and/or other moieties (e.g., fluorophores or chromophores) with concurrent variability in linker groups.
  • Several steroid phosphoramidites and vitamin phosphoramidites are commercially available and can be used in the above DNA synthesis cycle.
  • G 1 represents a desired moiety containing a carboxylic acid functional group (e.g., a drug moiety such as Auristatin F or SN 38), L represents a bivalent linker moiety (e.g., an alkylene, or alkylene ether), X represents a leaving group (e.g., halo such as Cl), and PG represents a protecting group (e.g., 4,4'-dimethoxytriphenylmethyl).
  • Step 1 of Reaction Scheme I starts with an activation of the carboxylic acid functional group of the first compound shown using known reagents under basic conditions (e.g., HATU and DIPEA in DMF).
  • the activated acid is then reacted with an amine to provide the reaction product of Step 1.
  • the resulting diol is then protected under standard conditions (e.g., 4,4'-dimethoxytriphenylmethyl chloride and pyridine).
  • the protected product is then reacted with 3- ((chloro(diisopropylamino)phosphaneyl)oxy)propanenitrile (or other appropriate reagent) to yield a desired compound of Structure (VII) as shown above.
  • the resultant compound of Structure (VII) can then be used to synthesize a desired compound of Structure (I) by reaction under well-known (automated) DNA synthesis conditions.
  • additional repeat units may be incorporated to achieve a final compound of Structure (I).
  • compounds having the following structure may be used: wherein: L is a desired linker moiety (e.g., including PEG or dye-containing moiety).
  • L is a desired linker moiety (e.g., including PEG or dye-containing moiety).
  • the following compound may be used in the synthesis of a compound of Structure (I): .
  • EXAMPLES General Methods Mass spectral analysis is performed on a Waters/Micromass Quattro micro MS/MS system (in MS only mode) using MassLynx 4.1 acquisition software.
  • Mobile phase used for LC/MS on dyes is 100 mM 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP), 8.6 mM triethylamine (TEA), pH 8.
  • Phosphoramidites and precursor molecules are also analyzed using a Waters Acquity UHPLC system with a 2.1 mm ⁇ 50 mm Acquity BEH-C18 column held at 45 °C, employing an acetonitrile / water mobile phase gradient.
  • Molecular weights for monomer intermediates are obtained using tropylium cation infusion enhanced ionization on a Waters/Micromass Quattro micro MS/MS system (in MS only mode).
  • EXAMPLE 1 S YNTHESIS OF DMT PROTECTED GEMCITABINE C OMPOUND Gencitabine (0.5057 mmole) is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of pyridine, anhydrous (5.06 mL). The reaction flask is then transferred to an ice water bath (0°C) and allowed to cool with mixing until thermally equalized (approximately 10 minutes). Then, 4,4′-Dimethoxytrityl chloride (0.257 g, 0.759 mmole) is added to the cooled mixture with continuous mixing under inert gas. The reaction mixture is allowed to warm to room temperature then sampled for TLC analysis.
  • the separated organic phase is washed with sodium chloride (5.06 mL, saturated aq.) and separated.
  • the separated organic phase is dried over sodium sulfate, anhydrous and the sodium sulfate filtered off.
  • the product containing organic phase is sampled for TLC and LC-UV/MS analysis.
  • Solvent is removed by rotary evaporation, resulting in crude DMT protected gemcitabine.
  • This crude material is then combined with crude material from a small-scale test reaction.
  • the combined crude material is purified by silica gel flash chromatography, dichloromethane / methanol mobile phase, product containing fractions are pooled and solvent is removed by rotary evaporation, and then placed on vacuum line for at least 24 hours to yield DMT protected gemcitabine.
  • the organic phases are combined and dried over sodium sulfate, anhydrous, and then the sodium sulfate filtered off.
  • the product containing organic phase is sampled for TLC and LC-UV/MS analysis.
  • dichloromethane is stripped off by rotary evaporation and proceeded to purification without crude weight.
  • This crude material is then combined with crude material from a small-scale test reaction.
  • the combined crude material is purified by silica gel solid phase extraction, dichloromethane/ methanol / triethylamine mobile phase, product containing fractions are pooled.
  • the mobile phase is stripped off by rotary evaporation, and then placed on vacuum line for at least 24 hours to yield DMT protected gemcitabine phosphoramidite.
  • DMT protected gemcitabine phosphoramidite can be purchased from Glen Research and used as is with no additional purification.
  • EXAMPLE 3 SYNTHESIS OF AURISTATIN F PHOSPHORAMIDITE Auristatin F (0.501 g, 0.671 mmole) was added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF (6.10 mL). The Auristatin F was allowed to dissolve completely under inert gas at room temperature.
  • the crude compound (a portion of crude material from previous step, 0.444 g, 0.5057 mmole theoretical) was added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of pyridine, anhydrous (5.06 mL). The reaction flask was then transferred to an ice water bath (0°C) and allowed to cool with mixing until thermally equalized (approximately 10 minutes). Then, 4,4′-Dimethoxytrityl chloride (0.257 g, 0.759 mmole) was added to the cooled mixture with continuous mixing under inert gas. The reaction mixture was allowed to warm to room temperature then sampled for TLC analysis.
  • the purified DMT protected compound (portion of the material 0.267, 0.226 mmole), dried under vacuum for at least 24 hours was dissolved in dichloromethane (2.26 mL), under inert gas blanket, with magnetic stir bar, followed by addition of DIPEA (0.117 g), and then addition of Cl-Phos. (0.107 g). The reaction was allowed to mix for approximately 15 minutes and then sampled for TLC analysis (TLC showed reaction completion). When reaction completion was verified, the reaction mixture was washed by adding directly to sodium bicarbonate (2.26 mL, saturated aq.) and organic phase separated, repeated one time. The organic phases were combined and dried over sodium sulfate, anhydrous, and then the sodium sulfate filtered off.
  • the product containing organic phase was sampled for TLC and LC-UV/MS analysis (analytical LC-UV248nm chromatogram showed two product peaks (diastereomers) ⁇ 64% by total peak area, identified by MS). Then, dichloromethane stripped off by rotary evaporation and proceeded to purification without crude weight. This crude material was then combined with crude material from a small-scale test reaction. The combined crude material was purified by silica gel solid phase extraction, dichloromethane/ methanol / triethylamine mobile phase, product containing fractions were pooled (determined by TLC) sampled for TLC and LC- UV/MS analysis.
  • the mobile phase striped off by rotary evaporation, and then placed on vacuum line for at least 24 hours to yield 0.363 g of Auristatin F phosphoramidite.
  • Auristatin F phosphoramidite (Analytical LC-UV248nm chromatogram showed 68% target product by total peak area, identified by MS).
  • SN38 phosphoramidites, ibuprofen phosphoramidite, and naproxen phosphoramidite are synthesized according to the above procedure.
  • the SN38 phosphoramidites, ibuprofen phosphoramidite, and naproxen phosphoramidite include the following:
  • Step 1 Into a 500 mL round bottomed flask was placed Boc-Ala-Ala-OH (4.0g, 15.4mmol, Chem-Impex Cat#04505), DMF (150mL) and a magnetic stir bar. HATU coupling agent (7.0g, 18.4mmol) was added and the mixture was stirred for 5min before adding Fmoc-1,2- diaminoethane-HCl (4.9g, 15.4mmol) and diisopropylethylamine (8.0mL, 46.1mmol). After stirring overnight, TLC (silica plats with F254 and dichloromethane:methanol elution 9:1) indicated the reaction was complete.
  • Step 2 In a 250 mL round bottomed flask with magnetic stir bar was placed the Boc-Ala-Ala derivative prepared in the previous step (2.5g, 4.8mmol). Dichloromethane (30mL) and DMF (10mL) were added and the mixture stirred. To this was added 4M hydrochloric acid in dioxane (30mL, Sigma Cat#345547) and the mixture was stirred.
  • Step 3 Into a 250 mL round bottomed flask was placed the amine from the previous step (1.5 g, 3.5 mmol), DMF (35mL) and a magnetic stir bar.
  • Succinic anhydride (1.8g, 17.7mmol) was added in a single portion followed by triethylamine (6.6 mL, 47.2 mmol). The mixture was stirred for 2h at which point TLC indicated the reaction was complete (TLC elution 4:1 DCM:MeOH). The mixture was concentrated on the rotovap and then treated with potassium carbonate solution (1M, 50 mL). The mixture was allowed to stir for 60min. The mixture was acidified with hydrochloric acid (20%, 50 mL) and the product precipitated. The mixture was cooled on ice for 30 min and the solids were collected medium frit glass filter. Solid weight 1.32g. The product was confirmed by LC-MS.
  • Step 4 In a small 20 mL glass vial was placed 6,7-dihydroxy-4-oxaheptylamine (443 mg, 3.0 mmol, Berry & Associates Cat#LK4010) and DMF (5 mL). The sample was warmed on a heat plate set a 60 °C for 10 min prior to assembling the other components of the reaction. In a 200 mL round bottomed flask was placed the Succinylated Ala-Ala derivative prepared in the prior step (1.3 g, 2.5 mmol), a magnetic stir bar and DMF (18 mL) . HATU (1.1 g, 3.0 mmol, Anaspec) was added and the mixture was stirred for 5 min.
  • Step 5 Fmoc protecting group is cleaved with a base such as piperidine, 4-methylpiperidine, piperazine, 1,8-diazabicyclo[5.4.0]undec-7-ene, or morpholine.
  • a chose of the base depends on how acidic a substrate is resistant towards.
  • the Fmoc protected diol is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF. In a separate dried round bottom flask with a magnetic star bar, 20% piperidine in 80% DMF is prepared.
  • Step 6 Carbonylated Camptothecin derivative prepared according to the above carbonylation is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF (6.10 mL). The carbonylated Camptothecin derivative is allowed to dissolve completely under inert gas at room temperature. Then, to the mixture is added DIPEA (0.351 g, 2.013 mmole), followed by addition of HATU (0.278 g, 0.732 mmole). The amine diol (0.610 mmole) is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF (6.10 mL), and dissolve completely at room temperature.
  • Step 7 In a 200 mL round bottomed flask with magnetic stir bar is placed the diol prepared in the prior step (1.9 mmol) and pyridine (38 mL). 4,4-dimethoxytrityl chloride is added to the solution in a single portion. The flask is capped and allowed to stir overnight. The mixture is concentrated under reduced pressure and the product is isolated by flash chromatography 24 g silica column (Teledyne-ISCO) and eluted with a dichloromethane:methanol gradient resulting in crude product. Step 8 The tritylated product is placed in 50 mL RB flask with magnetic stir bar (0.28 mmol).
  • EXAMPLE 6 S YNTHESIS OF DOXORUBICIN POLYMER An exemplary alkynyl-containing polymer is coupled to doxorubicin-PEG-azide. Reaction conditions include CuSO 4 , tris(3-hydroxypropyltriazolylmethyl)amine (THPTA), and sodium ascorbate. The reaction is carried out in phosphate buffered aqueous solvent with 60% DMS at a pH of 7.6. The reaction is run at room temperature and the presence of the desired product is confirmed by LC-MS. D’ indicates another biologically active moiety attached to the backbone structure (I)-(VI) different from doxorubicin.
  • Reaction conditions include CuSO 4 , tris(3-hydroxypropyltriazolylmethyl)amine (THPTA), and sodium ascorbate. The reaction is carried out in phosphate buffered aqueous solvent with 60% DMS at a pH of 7.6. The reaction is run at room temperature and the presence of the desired product is confirmed by
  • SN38 azide, alanine linked SN38, and monomethyl auristatin E (MMAE) azide were also coupled to the compound of structure (II), (V), or (VI) via click reaction as shown above.
  • the filtrate is removed and a third 10 mL 100 mM NaOH aliquot is added to the retentate.
  • the setup is centrifuged as before and the filtrate removed.
  • a fourth 10 mL 100 mM NaOH aliquot is added to the retentate and centrifuged as before.
  • the filtrate is removed and 10 mL of water are added to the filtration setup.
  • the mixture is centrifuged as before.
  • the retentate is removed, the filtration vessel is washed with water and the rinesates are added to the final volume (3.5 mL).
  • the desired product is confirmed by LC-MS and absorbance is used to determine concentration.
  • EXAMPLE 8 ACTIVATION AND ANTIBODY CONJUGATION OF COMPOUND I-1
  • the maleimide functionalized Compound I-1 is prepared according to the method described in Example 1.
  • an UCHT-1 antibody is treated with bis-maleimidoethane ("BMOE") to reduce disulfide bonds.
  • the reduced antibody is reacted with Compound I-1 in a 5:1 molar ratio of polymer to antibody.
  • the reaction results in a final product having a polymer to antibody ratio of 1:1 as detected by size exclusion chromatography.
  • anti-CD33, anti-CD70, or anti-CD123 may be used with bis-maleimidoethane ("BMOE”) to reduce disulfide bonds.
  • the crude product obtained was optionally further purified by recrystallization, but the crude product had enough purity to use without further purification. Presence of compound I-29 was confirmed by a mass spec. The mass spectra of the crude product compound I-29 after 4 hours is included hereinbelow. At 4 hours the reaction was a 1:1 mixture between conjugated and unconjugated polymer. Theoretical MW of I-29 is 5307 and observed MW is 5307.3.
  • Small porous beads are initially treated with functional units, which bind to the surface of the porous beads.
  • Peptide chains are built upon the functional units sites and remain covalently bonded to the bead until they are cleaved.
  • a peptide chain is immobilized on the solid phase and retained during a filtration process, wherein liquid reagents and by-products of the synthesis are washed away.
  • the general cycle of solid phase synthesis is one of repeated cycles of deprotection-wash- coupling-wash.
  • a free N-terminal amine of a peptide, attached to a solid support is coupled to an N-protected amino acid group (e.g., with Fmoc or Boc).
  • the newly introduced amino acid unit is deprotected to reveal a new N-terminal amine, which is further reacted with additional amino acids.
  • the process is repeated and the peptide chain is elongated.
  • the peptide chain has incorporated all desired amino acid and monomer units, it is cleaved from the bead.
  • Cleaving reagents such as anhydrous hydrogen fluoride or trifluoroacetic acid can be used to cleave peptide chains from beads.
  • the peptide chain is then collected, purified and characterized.

Abstract

Conjugates of biologically active compounds are disclosed. The compounds can have the following structure (I): Formula (I) or a stereoisomer, tautomer, or salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, L1a, L1b, L2, L3, L4, L5, L6, L7, M1, M2, M3, p, q, w, m and n are as defined herein. Compounds of structure (I) find utility in a number of applications, including use as therapeutic agents for various treatment methods.

Description

POLYMERS WITH DUAL THERAPEUTIC AGENTS BACKGROUND Field Embodiments of the present disclosure are generally directed to polymeric biologically active compounds having alkylating agents, and methods for their preparation. Description of the Related Art Targeted drug conjugates, unlike, e.g., chemotherapy, deliver drugs to target cells, with little or no off-target activity. Typically, targeted drug conjugates comprise a targeting molecule that is linked to a biologically active payload or drug. By combining the unique targeting capability with the therapeutic effectiveness of a biologically active drug (or moiety), conjugates can deliver the drug only to the intended target and minimize potential side effects. Antibody-drug conjugates (ADCs) are one class of targeted drug conjugates that are of particular interest, for example for cancer treatment. ADCs for cancer treatment combine the targeting features of monoclonal antibodies with cancer-killing ability of cytotoxic agents to provide a therapeutic with several advantages over other chemotherapeutics. However, challenges related to the complexity of ADC constructs, specifically the chemical linker between antibody and drug, has caused significant difficulties for development of new and effective therapeutics. Although the first ADC was approved in 2001, it took almost a decade before the next ADC was approved. As of today, only Adcetris®, Besponsa®, Enhertu®, Mylotarg®, Padcev®, Polivy®, and Kadcyla® are commercially available globally (Zevalin® has been approved in China only). Thus, there exists a need in the art for developing potent, targeting drug conjugates having a high therapeutic index and methods of preparing the same. The present disclosure fulfills this need and provides further related advantages. BRIEF SUMMARY In one embodiment, compounds having the following structure (I) are provided: (I) or a stereoisomer, tautomer, or salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, L1a, L1b, L2, L3, L4, L5, L6, L7, M1, M2, M3, q, w, p, m, and n are as defined herein. Compounds of structure (I) find utility in a number of applications, including use as therapeutic agents for various treatment methods. In another embodiment, compositions are provided which comprise a compound of structure (I) and a pharmaceutically acceptable carrier. In yet another embodiment, a method of treating a disease is provided, the method comprising administering to a subject in need thereof a therapeutically effective amount of a compound of structure (I) or a composition comprising a compound of structure (I), wherein each M1 or M3 is independently a biologically active moiety effective for treating the disease. In yet another embodiment, compounds having the following structure (II). (II) or salt, tautomer, or stereoisomer thereof, wherein R1, R2, R3, R4, R5, R6, R7, L2, L3, L4, L5, L6, L7, G, M2, M3, q, w, p, m, and n are as defined herein. Compounds of structure (II) are synthetic intermediates which are used to form compounds of structure (I). These and other aspects of the disclosure will be apparent upon reference to the following detailed description. DETAILED DESCRIPTION In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments of the disclosure. However, one skilled in the art will understand that the disclosure may be practiced without these details. Unless the context requires otherwise, throughout the present specification and claims, the word "comprise" and variations thereof, such as, "comprises" and "comprising" are to be construed in an open, inclusive sense, that is, as "including, but not limited to". Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present disclosure. Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
"Amino" refers to the -NH2 group.
"Carboxy" refers to the -CO2H group.
"Cyano" refers to the -CN group.
"Formyl" refers to the -C(=O)H group.
"Hydroxy" or "hydroxyl" refers to the -OH group.
"Imino" refers to the =NH group.
"Nitro" refers to the -NO2 group.
"Oxo" refers to the =O substituent group.
"Sulfhydryl" refers to the -SH group.
"Thioxo" refers to the =S group.
"Alkyl" refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms (C1-C12 alkyl), one to eight carbon atoms (C1-C8 alkyl) or one to six carbon atoms (C1-C8 alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl,
1-methylethyl (iso-propyl), n-butyl, n-pentyl, 1,1 -dimethylethyl (/-butyl), 3 -methylhexyl,
2-methylhexyl, and the like. Unless stated otherwise specifically in the specification, alkyl groups are optionally substituted.
"Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation, and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, alkylene is optionally substituted. "Alkenylene" or "alkenylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond and having from two to twelve carbon atoms, e.g., ethenylene, propenylene, n-butenylene, and the like. The alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond. The points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, alkenylene is optionally substituted. "Alkynylene" or "alkynylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond and having from two to twelve carbon atoms, e.g., ethynylene, propynylene, n-butynylene, and the like. The alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond. The points of attachment of the alkynylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, alkynylene is optionally substituted. "Alkylether" refers to any alkyl group as defined above, wherein at least one carbon- carbon bond is replaced with a carbon-oxygen bond. The carbon-oxygen bond may be on the terminal end (as in an alkoxy group) or the carbon oxygen bond may be internal (i.e., C-O-C). Alkylethers include at least one carbon oxygen bond, but may include more than one. For example, polyethylene glycol (PEG) is included within the meaning of alkylether. Unless stated otherwise specifically in the specification, an alkylether group is optionally substituted. For example, in some embodiments an alkylether is substituted with an alcohol or ˗OP(=Ra)(Rb)Rc, wherein each of Ra, Rb and Rc is as defined for compounds of structure (I). "Alkoxy" refers to a group of the formula ˗ORa where Ra is an alkyl group as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group is optionally substituted. "Alkoxyalkylether" refers to a group of the formula ˗ORaRb where Ra is an alkylene group as defined above containing one to twelve carbon atoms, and Rb is an alkylether group as defined herein. Unless stated otherwise specifically in the specification, an alkoxyalkylether group is optionally substituted, for example substituted with an alcohol or ˗OP(=Ra)(Rb)Rc, wherein each of Ra, Rb and Rc is as defined for compounds of structure (I). "Heteroalkyl" refers to an alkyl group, as defined above, comprising at least one heteroatom (e.g., N, O, P or S) within the alkyl group or at a terminus of the alkyl group. In some embodiments, the heteroatom is within the alkyl group (i.e., the heteroalkyl comprises at least one carbon-[heteroatom]x-carbon bond, where x is 1, 2 or 3). In other embodiments, the heteroatom is at a terminus of the alkyl group and thus serves to join the alkyl group to the remainder of the molecule (e.g., M1-H-A), where M1 is a portion of the molecule, H is a heteroatom and A is an alkyl group). Unless stated otherwise specifically in the specification, a heteroalkyl group is optionally substituted. Exemplary heteroalkyl groups include ethylene oxide (e.g., polyethylene oxide), optionally including phosphorous-oxygen bonds, such as phosphodiester bonds. "Heteroalkoxy" refers to a group of the formula ˗ORa where Ra is a heteroalkyl group as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a heteroalkoxy group is optionally substituted. "Heteroalkylene" refers to an alkylene group, as defined above, comprising at least one heteroatom (e.g., Si, N, O, P or S) within the alkylene chain or at a terminus of the alkylene chain. In some embodiments, the heteroatom is within the alkylene chain (i.e., the heteroalkylene comprises at least one carbon-[heteroatom]-carbon bond, where x is 1, 2 or 3). In other embodiments, the heteroatom is at a terminus of the alkylene and thus serves to join the alkylene to the remainder of the molecule (e.g., M1-H-A-M2, where M1 and M2 are portions of the molecule, H is a heteroatom and A is an alkylene). Unless stated otherwise specifically in the specification, a heteroalkylene group is optionally substituted. Exemplary heteroalkylene groups include ethylene oxide (e.g., polyethylene oxide) and the "C," "HEG," and "PEG 1K" linking groups illustrated below:
Figure imgf000006_0001
Multimers of the above C-linker, HEG linker and/or PEG 1K linker are included in various embodiments of heteroalkylene linkers. In some embodiments of the PEG 1K linker, n ranges from 19-25, for example n is 19, 20, 21, 22, 23, 24, or 25. Multimers may comprise, for example, the following structure: wherein x is 0 or an integer greater than 0, for example, x ranges from 0-100 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10). A "linker" refers to a contiguous chain of at least one atom, such as carbon, oxygen, nitrogen, sulfur, phosphorous, and combinations thereof, which connects a portion of a molecule to another portion of the same molecule or to a different molecule, moiety or solid support (e.g., microparticle). Linkers may connect the molecule via a covalent bond or other means, such as ionic or hydrogen bond interactions. In some embodiments, the linker is a heteroatomic linker (e.g., comprising 1-10 Si, N, O, P, or S atoms), a heteroalkylene (e.g., comprising 1-10 Si, N, O, P, or S atoms and an alkylene chain) or an alkylene linker (e.g., comprising 1-12 carbon atoms). In some embodiments, a heteroalkylene linker comprises the following structure: wherein: x9 and x10 are each independently an integer greater than 0. In some embodiments, the heteroatomic linker is –O–, –S–, or –OP(=O)O-–O–. In some embodiments, the heteroalkylene linker comprises –OP(=O)O-–O–. In some embodiments, the heteroalkylene linker comprises at least one S-S bond. "Physiologically cleavable linker" refers to a molecular linkage that can be split or separated a prescribed manner, resulting in two or more separate molecules while in the presence of an in vivo or in vitro environment of an organism or cell system. Generally, physiological conditions that induce such a cleavage or scission event may include a temperature ranging from about 20 to 40°C, an atmospheric pressure of about 1 atm (101 kPa or 14.7 psi), a pH of about 6 to 8, a glucose concentration of about 1 to 20 mM, atmospheric oxygen concentration, and earth gravity. In some embodiments, physiological conditions include enzymatic conditions (i.e., enzymatic cleavage). Bond cleavage or scission can be homolytic or heterolytic. "Heteroalkenylene" is a heteroalkylene, as defined above, comprising at least one carbon- carbon double bond. Unless stated otherwise specifically in the specification, a heteroalkenylene group is optionally substituted. "Heteroalkynylene" is a heteroalkylene comprising at least one carbon-carbon triple bond. Unless stated otherwise specifically in the specification, a heteroalkynylene group is optionally substituted. "Heteroatomic" in reference to a "heteroatomic linker" refers to a linker group consisting of one or more heteroatoms. Exemplary heteroatomic linkers include single atoms selected from the group consisting of O, N, P and S, and multiple heteroatoms for example a linker having the formula ˗P(O-)(=O)O˗ or ˗OP(O-)(=O)O˗ and multimers and combinations thereof. "Phosphate" refers to the ˗OP(=O)(Ra)Rb group, wherein Ra is OH, O- or ORc; and Rb is OH, O-, ORc, a thiophosphate group or a further phosphate group, wherein Rc is a counter ion (e.g., Na+ and the like). "Phosphoalkyl" refers to the ˗OP(=O)(Ra)Rb group, wherein Ra is OH, O- or ORc; and Rb is ˗Oalkyl, wherein Rc is a counter ion (e.g., Na+ and the like). Unless stated otherwise specifically in the specification, a phosphoalkyl group is optionally substituted. For example, in certain embodiments, the ˗Oalkyl moiety in a phosphoalkyl group is optionally substituted with one or more of hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether, thiophosphoalkylether or ˗OP(=Ra)(Rb)Rc, wherein each of Ra, Rb and Rc is as defined for compounds of structure (I). "Phosphoalkylether" refers to the ˗OP(=O)(Ra)Rb group, wherein Ra is OH, O- or ORc; and Rb is ˗Oalkylether, wherein Rc is a counter ion (e.g., Na+ and the like). Unless stated otherwise specifically in the specification, a phosphoalkylether group is optionally substituted. For example, in certain embodiments, the -Oalkylether moiety in a phosphoalkylether group is optionally substituted with one or more of hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether, thiophosphoalkylether or ˗OP(=Ra)(Rb)Rc, wherein each of Ra, Rb and Rc is as defined for compounds of structure (I). "Thiophosphate" refers to the ˗OP(=Ra)(Rb)Rc group, wherein Ra is O or S, Rb is OH, O-, S-, ORd or SRd; and Rc is OH, SH, O-, S-, ORd, SRd, a phosphate group or a further thiophosphate group, wherein Rd is a counter ion (e.g., Na+ and the like) and provided that: i) Ra is S; ii) Rb is S- or SRd; iii)Rc is SH, S- or SRd; or iv) a combination of i), ii) and/or iii). "Thiophosphoalkyl" refers to the ˗OP(=Ra)(Rb)Rc group, wherein Ra is O or S, Rb is OH, O-, S-, ORd or SRd; and Rc is ˗Oalkyl, wherein Rd is a counter ion (e.g., Na+ and the like) and provided that: i) Ra is S; ii) Rb is S- or SRd; or iii)Ra is S and Rb is S- or SRd. Unless stated otherwise specifically in the specification, a thiophosphoalkyl group is optionally substituted. For example, in certain embodiments, the ˗Oalkyl moiety in a thiophosphoalkyl group is optionally substituted with one or more of hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether, thiophosphoalkylether or ˗OP(=Ra)(Rb)Rc, wherein each of Ra, Rb and Rc is as defined for compounds of structure (I). "Thiophosphoalkylether" refers to the ˗OP(=Ra)(Rb)Rc group, wherein Ra is O or S, Rb is OH, O-, S-, ORd or SRd; and Rc is ˗Oalkylether, wherein Rd is a counter ion (e.g., Na+ and the like) and provided that: i) Ra is S; ii) Rb is S- or SRd; or iii)Ra is S and Rb is S- or SRd. Unless stated otherwise specifically in the specification, a thiophosphoalkylether group is optionally substituted. For example, in certain embodiments, the -Oalkylether moiety in a thiophosphoalkyl group is optionally substituted with one or more of hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether, thiophosphoalkylether or ˗OP(=Ra)(Rb)Rc, wherein each of Ra, Rb and Rc is as defined for compounds of structure (I). "Carbocyclic" refers to a stable 3- to 18-membered aromatic or non-aromatic ring comprising 3 to 18 carbon atoms. Unless stated otherwise specifically in the specification, a carbocyclic ring may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems, and may be partially or fully saturated. Non-aromatic carbocyclyl radicals include cycloalkyl, while aromatic carbocyclyl radicals include aryl. Unless stated otherwise specifically in the specification, a carbocyclic group is optionally substituted. "Cycloalkyl" refers to a stable non-aromatic monocyclic or polycyclic carbocyclic ring, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Monocyclic cycloalkyls include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyls include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl- bicyclo-[2.2.1]heptanyl, and the like. Unless stated otherwise specifically in the specification, a cycloalkyl group is optionally substituted. "Aryl" refers to a ring system comprising at least one carbocyclic aromatic ring. In some embodiments, an aryl comprises from 6 to 18 carbon atoms. The aryl ring may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Aryls include, but are not limited to, aryls derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, an aryl group is optionally substituted. "Heterocyclic" refers to a stable 3- to 18-membered aromatic or non-aromatic ring comprising one to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclic ring may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclic ring may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclic ring may be partially or fully saturated. Examples of aromatic heterocyclic rings are listed below in the definition of heteroaryls (i.e., heteroaryl being a subset of heterocyclic). Examples of non-aromatic heterocyclic rings include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, pyrazolopyrimidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trioxanyl, trithianyl, triazinanyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocyclic group is optionally substituted. "Heteroaryl" refers to a 5- to 14-membered ring system comprising one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of certain embodiments of this disclosure, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, benzoxazolinonyl, benzimidazolthionyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, 1- oxidopyrazinyl, 1-oxidopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, pteridinonyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyridinonyl, pyrazinyl, pyrimidinyl, pryrimidinonyl, pyridazinyl, pyrrolyl, pyrido[2,3- d]pyrimidinonyl, quinazolinyl, quinazolinonyl, quinoxalinyl, quinoxalinonyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, thieno[3,2-d]pyrimidin-4-onyl, thieno[2,3-d]pyrimidin-4-onyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteroaryl group is optionally substituted. The suffix "-ene" refers to a particular structural feature (e.g., alkyl, aryl, heteroalkyl, heteroaryl) attached to the rest of the molecule through a single bond and attached to a radical group through a single bond. In other words, the suffix "-ene" refers to a linker having the structural features of the moiety to which it is attached. The points of attachment of the "-ene" chain to the rest of the molecule and to the radical group can be through one atom of or any two atoms within the chain. For example, a heteroarylene refers to a linker comprising a heteroaryl moiety as defined herein. "Fused" refers to a ring system comprising at least two rings, wherein the two rings share at least one common ring atom, for example two common ring atoms. When the fused ring is a heterocyclyl ring or a heteroaryl ring, the common ring atom(s) may be carbon or nitrogen. Fused rings include bicyclic, tricyclic, tertracyclic, and the like. The term "substituted" used herein means any of the above groups (e.g., alkyl, alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene, alkoxy, alkylether, phosphoalkyl, phosphoalkylether, thiophosphoalkyl, thiophosphoalkylether, carbocyclic, cycloalkyl, aryl, heterocyclic and/or heteroaryl) wherein at least one hydrogen atom (e.g., 1, 2, 3 or all hydrogen atoms) is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced with ˗NRgRh, ˗NRgC(=O)Rh, ˗NRgC(=O)NRgRh, ˗NRgC(=O)ORh, ˗NRgSO2Rh, ˗OC(=O)NRgRh, ˗ORg, ˗SRg, ˗SORg, ˗SO2Rg, ˗OSO2Rg, ˗SO2ORg, =NSO2Rg, and ˗SO2NRgRh. "Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with ˗C(=O)Rg, ˗C(=O)ORg, ˗C(=O)NRgRh, ˗CH2SO2Rg, ˗CH2SO2NRgRh. In the foregoing, Rg and Rh are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl. "Substituted" further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl group. In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents. "Conjugation" refers to the overlap of one p-orbital with another p-orbital across an intervening sigma bond. Conjugation may occur in cyclic or acyclic compounds. A "degree of conjugation" refers to the overlap of at least one p-orbital with another p-orbital across an intervening sigma bond. For example, 1, 3-butadine has one degree of conjugation, while benzene and other aromatic compounds typically have multiple degrees of conjugation. Fluorescent and colored compounds typically comprise at least one degree of conjugation. "Fluorescent" refers to a molecule which is capable of absorbing light of a particular frequency and emitting light of a different frequency. Fluorescence is well-known to those of ordinary skill in the art. "Colored" refers to a molecule which absorbs light within the colored spectrum (i.e., red, yellow, blue and the like). The term "biomolecule" refers to any of a variety of biological materials, including nucleic acids, carbohydrates, amino acids, polypeptides, glycoproteins, hormones, aptamers and mixtures thereof.

More specifically, the term is intended to include, without limitation, RNA,
 DNA, oligonucleotides, modified or derivatized nucleotides, enzymes, receptors, prions, receptor ligands (including hormones), antibodies, antigens, and toxins, as well as bacteria, viruses, blood cells, and tissue cells. The visually detectable biomolecules of the disclosure (e.g., compounds of structure (I) having a biomolecule linked thereto) are prepared, as further described herein, by contacting a biomolecule with a compound having a reactive group that enables attachment of the biomolecule to the compound via any available atom or functional group, such as an amino, hydroxy, carboxyl, or sulfhydryl group on the biomolecule. A "reactive group" is a moiety capable of reacting with a second reactive groups (e.g., a "complementary reactive group") to form one or more covalent bonds, for example by a displacement, oxidation, reduction, addition or cycloaddition reaction. Exemplary reactive groups are provided in Table 1, and include for example, nucleophiles, electrophiles, dienes, dienophiles, aldehyde, oxime, hydrazone, alkyne, amine, azide, acylazide, acylhalide, nitrile, nitrone, sulfhydryl, disulfide, sulfonyl halide, isothiocyanate, imidoester, activated ester, ketone, α,β-unsaturated carbonyl, alkene, maleimide, α-haloamide, epoxide, aziridine, tetrazine, tetrazole, phosphine, biotin, thiirane and the like. "Bio-conjugation" or "bio-conjugate" and related variations refer to a chemical reaction strategy for forming a stable covalent bond between two molecules. The term "bio-conjugation" is generally used when one of the molecules is a biomolecule (e.g., an antibody), but can be used to describe forming a covalent bond with a non-biomolecule (e.g., a polymeric resin). The product or compound resulting from such a reaction strategy is a "conjugate," "bio-conjugate" or a grammatical equivalent. The terms "visible" and "visually detectable" are used herein to refer to substances that are observable by visual inspection, without prior illumination, or chemical or enzymatic activation. Such visually detectable substances absorb and emit light in a region of the spectrum ranging from about 300 to about 900 nm. Preferably, such substances are intensely colored, preferably having a molar extinction coefficient of at least about 40,000, more preferably at least about 50,000, still more preferably at least about 60,000, yet still more preferably at least about 70,000, and most preferably at least about 80,000 M-1cm-1. The compounds of the disclosure may be detected by observation with the naked eye, or with the aid of an optically based detection device, including, without limitation, absorption spectrophotometers, transmission light microscopes, digital cameras and scanners. Visually detectable substances are not limited to those which emit and/or absorb light in the visible spectrum. Substances which emit and/or absorb light in the ultraviolet (UV) region (about 10 nm to about 400 nm), infrared (IR) region (about 700 nm to about 1 mm), and substances emitting and/or absorbing in other regions of the electromagnetic spectrum are also included with the scope of "visually detectable" substances. For purposes of embodiments of the disclosure, the term "photostable visible dye" refers to a chemical moiety that is visually detectable, as defined hereinabove, and is not significantly altered or decomposed upon exposure to light. Preferably, the photostable visible dye does not exhibit significant bleaching or decomposition after being exposed to light for at least one hour. More preferably, the visible dye is stable after exposure to light for at least 12 hours, still more preferably at least 24 hours, still yet more preferably at least one week, and most preferably at least one month. Non-limiting examples of photostable visible dyes suitable for use in the compounds and methods of the disclosure include azo dyes, thioindigo dyes, quinacridone pigments, dioxazine, phthalocyanine, perinone, diketopyrrolopyrrole, quinophthalone, and truarycarbonium. As used herein, the term "perylene derivative" is intended to include any substituted perylene that is visually detectable. However, the term is not intended to include perylene itself. The terms "anthracene derivative", "naphthalene derivative", and "pyrene derivative" are used analogously. In some preferred embodiments, a derivative (e.g., perylene, pyrene, anthracene or naphthalene derivative) is an imide, bisimide or hydrazamimide derivative of perylene, anthracene, naphthalene, or pyrene. The visually detectable molecules of various embodiments of the disclosure are useful for a wide variety of analytical applications, such as biochemical and biomedical applications, in which there is a need to determine the presence, location, or quantity of a particular analyte (e.g., biomolecule). In another aspect, therefore, the disclosure provides a method for visually detecting a biomolecule, comprising: (a) providing a biological system with a visually detectable biomolecule comprising the compound of structure (I) linked to a biomolecule; and (b) detecting the biomolecule by its visible properties. For purposes of the disclosure, the phrase "detecting the biomolecule by its visible properties" means that the biomolecule, without illumination or chemical or enzymatic activation, is observed with the naked eye, or with the aid of an optically based detection device, including, without limitation, absorption spectrophotometers, transmission light microscopes, digital cameras and scanners. A densitometer may be used to quantify the amount of visually detectable biomolecule present. For example, the relative quantity of the biomolecule in two samples can be determined by measuring relative optical density. If the stoichiometry of dye molecules per biomolecule is known, and the extinction coefficient of the dye molecule is known, then the absolute concentration of the biomolecule can also be determined from a measurement of optical density. As used herein, the term "biological system" is used to refer to any solution or mixture comprising one or more biomolecules in addition to the visually detectable biomolecule. Nonlimiting examples of such biological systems include cells, cell extracts, tissue samples, electrophoretic gels, assay mixtures, and hybridization reaction mixtures. "Solid support" or "solid resin" refers to any solid substrate known in the art for solid- phase support of molecules, for example a "microparticle" refers to any of a number of small particles useful for attachment to compounds of the disclosure, including, but not limited to, glass beads, magnetic beads, polymeric beads, nonpolymeric beads, and the like. In certain embodiments, a microparticle comprises polystyrene beads. In some embodiments, the solid support or solid resin is controlled pore glass or macroporous polystyrene. A "solid support residue" refers to the functional group remaining attached to a molecule when the molecule is cleaved from the solid support. Solid support residues are known in the art and can be easily derived based on the structure of the solid support and the group linking the molecule thereto. A "targeting moiety" is a moiety that selectively binds or associates with a particular target, such as an analyte molecule. "Selectively" binding or associating means a targeting moiety preferentially associates or binds with the desired target relative to other targets. In some embodiments the compounds disclosed herein include linkages to targeting moieties for the purpose of selectively binding or associating the compound with an analyte of interest (i.e., the target of the targeting moiety), thus allowing detection of the analyte. Exemplary targeting moieties include, but are not limited to, antibodies, antigens, nucleic acid sequences, enzymes, proteins, cell surface receptor antagonists, and the like. In some embodiments, the targeting moiety is a moiety, such as an antibody, that selectively binds or associates with a target feature on or in a cell, for example a target feature on a cell membrane or other cellular structure, thus allowing for detection of cells of interest. Small molecules that selectively bind or associate with a desired analyte are also contemplated as targeting moieties in certain embodiments. One of skill in the art will understand other analytes, and the corresponding targeting moiety, that will be useful in various embodiments. "Base pairing moiety" refers to a heterocyclic moiety capable of hybridizing with a complementary heterocyclic moiety via hydrogen bonds (e.g., Watson-Crick base pairing). Base pairing moieties include natural and unnatural bases. Non-limiting examples of base pairing moieties are RNA and DNA bases such adenosine, guanosine, thymidine, cytosine and uridine and analogues thereof. Embodiments of the disclosure disclosed herein are also meant to encompass all compounds being isotopically-labeled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 31P, 32P, 35S, 18F, 36Cl, 123I, and 125I, respectively. Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described below and in the following Examples using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed. "Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. "Optional" or "optionally" means that the subsequently described event or circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted alkyl" means that the alkyl group may or may not be substituted and that the description includes both substituted alkyl groups and alkyl groups having no substitution. "Salt" includes both acid and base addition salts. "Acid addition salt" refers to those salts which are formed with inorganic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4- acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like. "Base addition salt" refers to those salts which are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. Crystallizations may produce a solvate of the compounds described herein. Embodiments of the present disclosure include all solvates of the described compounds. As used herein, the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the disclosure with one or more molecules of solvent. The solvent may be water, in which case the solvate may be a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present disclosure may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compounds of the disclosure may be true solvates, while in other cases the compounds of the disclosure may merely retain adventitious water or another solvent or be a mixture of water plus some adventitious solvent. Embodiments of the compounds of the disclosure (e.g., compounds of structure I), or their salts, tautomers or solvates may contain one or more stereocenters and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. Embodiments of the present disclosure are meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are non-superimposable mirror images of one another. A "tautomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any said compounds. Various tautomeric forms of the compounds are easily derivable by those of ordinary skill in the art. "Steroid" refers to a biologically active organic compound with four fused rings arrangement. The core structure of the steroid comprises three six-member cyclohexane rings (i.e., rings A, B, and C) and one five-member cyclopentane ring. Steroids vary by functional groups attached to the core structure and the oxidation stated of the rings. For example, the 3 position on the A ring of the steroid may be a hydroxyl group (e.g., cholesterol, cholic acid, lanosterol, and β-sitosterol) or carbonyl (-C=O) group (e.g., testosterone, dexamethasone, progesterone, and medrogestone). Examples of steroids used in some embodiments include cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone. "Vitamin" refers to a group of organic compounds which are essential for normal growth and nutrition and are required in small quantities in the diet because they cannot be synthesized by the body. Such compounds include Vitamin A, B1, B2, B3, B5, B6, B7, B9, B12, C, D, E, and K. Vitamin E is fat soluble compounds including four tocopherols and four tocotrienols. The core structure of the Vitamin E comprises 3,4-dihydro 2H-1 benzopyran. The chemical structural difference between tocotrienols and tocopherols is that tocotrienols have an unsaturated isoprenoid side chain with three carbon-carbon double bonds off of the 3,4-dihydro 2H-1 benzopyran core structure whereas tocopherols have a saturated isoprenoid side chain. Both tocotrienols and tocopherols have four structural isomers including α-tocopherol, β- tocopherol, γ-tocopherol, δ-tocopherol, α -tocotrienol, β -tocotrienol, γ -tocotrienol, and δ - tocotrienol. An “amino acid side chain” or “side chain” refers to substituents attached to the α-carbon, β-carbon, or γ-carbon of an amino acid. Amino acid side chains can be those associated with natural or unnatural amino acids. An “amino acid sequence” or “peptide sequence” refers to the primary structure of linked amino acid residues along a backbone formed via peptide bonds. Sequences are generally denoted from the N-terminal end to the C-terminal end. Embodiments of the present invention include compounds comprising certain amino acid sequences where indicated. Amino acid sequences are indicated, where appropriate, by 3-letter or 1-letter abbreviations. A “letter code,” “1-letter code,” or “3-letter code” refers to an indication or abbreviation for an amino acid or amino acid residue in an amino acid sequence. A general list of 1 and 3- letter codes and the amino acid they correspond to is found below:
Figure imgf000019_0001
Figure imgf000020_0001
A “beta sheet,” “β-sheet,” “beta pleated sheet,” or “β-pleated sheet” refers to secondary structure of amino acid sequences that forms via intra molecular folding. Strands of amino acids are connected laterally by hydrogen bonding forming a generally twisted, pleated sheet. Strands of amino acids that form this secondary structure are generally 3 to 10 amino acid residues in length. The present invention includes amino acid sequences that include amino acid residues with beta sheet forming propensities where beta sheet moieties are indicated. These residues include, but are not limited to, glycine, methionine, serine, valine, tyrosine, phenylalanine, tryptophan, threonine, and isoleucine. An “alpha helix” or “α-helix” refers to secondary structure of amino acid chains that forms via intra molecular folding. A spiral conformation is formed wherein backbone N-H groups donate a hydrogen bond to backbone C=O groups of the amino acid four residues earlier. Amino acid side chain sequence influences formation of alpha helical structure. The present invention includes amino acid sequences that include residues with helix-forming propensities where alpha helix moieties are indicated. These residues include, but are not limited to, glycine, methionine, alanine, arginine, histidine, leucine, glutamate, glutamic acid, phenylalanine, valine, tyrosine, and lysine. The chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C. nomenclature system, using the ACD/Name Version 9.07 software program and/or ChemDraw Ultra Version 11.0 software naming program (CambridgeSoft). Common names familiar to one of ordinary skill in the art are also used. Compounds As noted above, in one embodiment of the present disclosure, compounds useful as covalent linkers between biologically active moieties such as alkylating agents and targeting moieties are provided. In other embodiments, compounds useful as synthetic intermediates for preparation of compounds comprising one or more biologically active moieties are provided. Accordingly, in some embodiments, M1 is, at each occurrence, independently either i) H or ii) a moiety comprising a biologically active moiety, provided that M1 is absent when L1b is H; M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety. In some embodiments, M1 or M3 is an alkylating agent (e.g., monomethyl auristatin F, monomethyl auristatin E, SN38, pyrrolo benzo diazepine and the like) and M2 is fluorescent dye (e.g., fluorescein and the like). Numerous advantages are afforded by embodiments disclosed herein, including the ability to control the number and type of biologically active moieties M1 and M3 that are attached to the polymer backbone and any subsequent targeting moiety, spacing between the neighboring biologically active moieties on the polymer backbone (e.g., how far or close each of the biologically active moieties M1 and M3 are), and spacing between the polymer backbone and the biological active moiety (e.g., the length of linkers off of the polymer backbone). This allows for constructing compounds with the biologically active moieties to facilitate an alkylation of guanine (G) of DNA such that the biological active moieties attached to the polymer backbone are located in a low energy position within the minor groove of DNA. Compounds disclosed in the present disclosure have multiple alkylating agents as the biological active moieties which are allowed to form interstrand and/or intrastrand DNA crosslinks resulting in greater DNA stabilization. The biological active moieties can be attached to the polymer backbone via physiologically cleavable or non-cleavable linkers. The procedures described in the present disclosure provide the ability to selectively install the physiologically cleavable and/or non- cleavable linkers. This allows for synthesizing a compound which has both physiologically cleavable and non-cleavable linkers with one or more biological active moieties. In this regard, the biological active moieties can be cleaved sequentially depending on physiological conditions. Further, the ratio of a biological active moiety on M1 and a biological active moiety on M2 can be fine-tuned based on a type of disease, progression of the disease, and a therapeutic method. Additionally, a compound with both multiple biological active moieties and a fluorescent moiety attached by physiologically cleavable and/or non-cleavable linkers can be synthesized. Some embodiments of the present disclosure provide combinations of therapeutic agents, targeting moieties, and dye moieties (e.g., chromophores or fluorophores) that can be used for simultaneous targeting, treatment, and detection. The ease of coupling polymer-drug constructs to targeting agents such as antibodies, antibody fragments, proteins or other clinically interesting agents provides utility to a wide variety of interesting applications (e.g., surface chemistries, assay development, etc.). Accordingly, in some embodiments, M2 is a chromophore or fluorophore (e.g., FITC, 5-FAM, 6-FAM, and the like). The compounds of certain embodiments also provide other desirable properties, including enhanced permeability and retention effects. In addition to providing necessary solubility characteristics, the chemical features of embodiments of the present compounds can be adjusted to modulate the compound’s ability to permeate diseased cells/tissue and be retained within the same. These features allow effective delivery of biologically active agents by increasing permeation and increasing efficacy by enhancing retention. Accordingly, it is understood that any embodiment of the compounds of Structures (I), (II), or (III), as set forth above, may be independently combined with other embodiments to form embodiments of the disclosure not specifically set forth above. It is understood that in the present description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds. Accordingly, one embodiment provides a compound having the following structure (I): (I) or a stereoisomer, pharmaceutically salt or tautomer thereof, wherein: R1 and R2 are each independently H, OH, SH, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; R3 is, at each occurrence, independently H, alkyl or alkoxy; R4 is, at each occurrence, independently OH, SH, O-, S-, ORd or SRd; R5 is, at each occurrence, independently oxo or thioxo; R6 and R7 are, at each occurrence, independently H, OH, or halo; L1a is, at each occurrence, independently a heteroalkylene or heteroarylene linker; L1b is, at each occurrence, independently either i) H or ii) a linker; L2, L3, L5, and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; L4 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker; L7 is, at each occurrence, independently a linker; M1 is, at each occurrence, independently either i) absent or ii) a moiety comprising a biologically active moiety, provided that M1 is absent when L1b is H; M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of structure (I); and m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, an integer of zero or greater; p is, at each occurrence, an integer of zero or greater; w is, at each occurrence, an integer of zero or greater; and n is an integer of one or greater, wherein the sum of p and w is an integer of two or greater, and the compound comprises at least two different biologically active moieties. The various linkers and substituents (e.g., R1, R2, R3, R4, R5, R6, R7, L1a, L1b, L2, L3, L4, L5, L6, L7, M1, M2, M3, and Q) in the compound of structure (I) are optionally substituted with one more substituent. For example, in some embodiments the optional substituent is selected to optimize the water solubility or other property of the compound of structure (I). In certain embodiments, each chromophore, alkyl, alkoxy, alkylether, heteroarylene, heteroalkyl, alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene, alkoxyalkylether, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether in the compound of structure (I) is optionally substituted with one more substituent selected from the group consisting of hydroxyl, alkoxy, alkylether , alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether. In certain embodiments the optional substituent is ˗OP(=Ra)(Rb)Rc, where Ra, Rb and Rc are as defined for the compound of structure (I). In some embodiments, at least one occurrence of L1a is an optionally substituted 5-9 membered heteroarylene linker. In certain embodiments, L1a is a substituted 5-membered heteroarylene linker. In certain embodiments, L1a is a substituted 6-membered heteroarylene linker. In certain embodiments, L1a is a substituted 7-membered heteroarylene linker. In certain embodiments, L1a is a substituted 8-membered heteroarylene linker. In certain embodiments, L1a is a substituted 9-membered heteroarylene linker. In some related embodiments, L1a is substituted with oxo, alkyl (e.g., methyl, ethyl, etc.) or combinations thereof. In certain embodiments, L1a is, at each occurrence, unsubstituted. In some more specific embodiments, L1a is, at each occurrence independently a pyrimidine. In some more specific embodiments, L1a is, at each occurrence, independently cytosine or thymine. In some embodiments, L1a is, at each occurrence, independently selected from cytosine and thymine such that the compound comprises a sequence of cytosine and thymine bases capable of triplex formation with a target DNA sequence. In some more specific embodiments, L1a has one of the following structures: or . In other embodiments, L1a has the following structure when L1b and M1 are absent: wherein R8 is, at each occurrence, independently O, NH, or NRe; R9 is, at each occurrence, independently H, alkyl, or optionally substituted alkyl; R10 is, at each occurrence, independently H or F; and Re is, at each occurrence, independently alkyl or optionally substituted alkyl. In some embodiments, at least one occurrence of R8 is O. In some embodiments, at least one occurrence of R8 is NH. In some embodiments, at least one occurrence of R8 is NRe. In some more specific embodiments, R8 is, at each occurrence, independently O. In some more specific embodiments, R8 is, at each occurrence, independently NH. In some more specific embodiments, R8 is, at each occurrence, independently NRe. In some embodiments, at least one occurrence of R9 is H. In some embodiments, at least one occurrence of R9 is alkyl. In some more specific embodiments, at least one occurrence of R9 is substituted alkyl. For example, in some embodiments, at least one occurrence of R9 is an acyl group (-C(=O)CH3, -C(=O)CH2CH3, or the like). In some embodiments, at least one occurrence of R10 is H. In some embodiments, at least one occurrence of R10 is F. In some more specific embodiments, R10 is, at each occurrence, independently H. In some more specific embodiments, R10 is, at each occurrence, independently F. In certain embodiments, R8 is O, R9 is H, and R10 is H. Some embodiments provide a compound having the following structure (IA): (IA) or a stereoisomer, salt or tautomer thereof. Some embodiments provide a compound having the following structure (IB): (IB) or a stereoisomer, salt or tautomer thereof. Compounds of structure (IB) have a gemcitabine moiety incorporated to the polymeric backbone, which can act a therapeutic agent. In some embodiments, at least one occurrence of R6 or R7 is F or H. In certain embodiments, each occurrence of R6 or R7 is F or H. In some more specific embodiments, wherein at least each occurrence of R6 and R7 are F, R8 is O, R9 is H, and R10 is H. Some embodiments provide a compound having the following structure (IC): (IC) or a stereoisomer, salt or tautomer thereof, wherein k is, at each occurrence, an integer of zero or greater. In some embodiments, at least one occurrence of L4 is heteroalkylene. In some embodiments, each occurrence of L4 is heteroalkylene. In some embodiments, at least one occurrence of L4 comprises alkylene oxide. In some more specific embodiments, the alkylene oxide of L4 is ethylene oxide. In some more specific embodiments, the ethylene oxide is polyethylene oxide. In certain embodiments, L4, at each occurrence, has the following structure: , wherein: z is an integer from 1 to 100; and * indicates a bond to the adjacent phosphorous atom. In some embodiments, z is an integer from 3 to 8. In some embodiments, z is 3. In some embodiments, z is 4. In some embodiments, z is 5. In some embodiments, z is 6. In some embodiments, z is 7. In some embodiments, z is 8. In some other embodiments, z is an integer from 22 to 26. In some embodiments, z is 22. In some embodiments, z is 23. In some embodiments, z is 24. In some embodiments, z is 25. In some embodiments, z is 26. In some other embodiments, z ranges from 19 to 28. In certain embodiments, the average z is 23. In some embodiments, the average z is 19, 20, 21, 22, 23, 24, 25, 26, 27, or 28. In other embodiments, at least one occurrence of L4 is an alkylene linker (e.g., methylene). In some more specific embodiments, L4 is an alkylene linker at each occurrence (e.g., methylene). In some embodiments, at least one occurrence of in compounds of structure (I) comprising one of the following structures: ; ; ; or wherein z is an integer from 1 to 100. In some embodiments, z is an integer from 3 to 8. In some other embodiments, z is an integer from 22 to 26. In some other embodiments, z ranges from 19 to 28. In some embodiments, at each occurrence, in compounds of structure (I) comprising one of the following structures: ; ; ; or wherein z is an integer from 1 to 100. In some embodiments, z is an integer from 3 to 8. In some other embodiments, z is an integer from 22 to 26. In some other embodiments, z ranges from 19 to 28. Some embodiments provide a compound having one of the following structures (ID), (IE), or (IF): (ID) (IE) or (IF) or a stereoisomer, salt or tautomer thereof. In some embodiments, L3, L5, and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers. In some embodiments, L3, L5 or L6 are, at each occurrence, independently alkylene. In more embodiments, L3, L5 or L6 are, at each occurrence, independently C1-C6 alkylene, C2-C6 alkenylene or C2-C6 alkynylene. In some more specific embodiments, the alkylene linker of L3 and L5 are C1 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C2 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C3 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C4 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C5 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C6 alkyl linker. In some specific embodiments, the alkylene linker of L6 are C1 alkyl linker. In some specific embodiments, the alkylene linker of L6 are C2 alkyl linker. In some specific embodiments, the alkylene linker of L6 are C3 alkyl linker. In other embodiments, at least one occurrence of L6 is a direct bond. In some other embodiments, each occurrence of L6 is a direct bond. Some embodiments provide a compound having one of the following structures (IG), (IH), or (IJ): (IG) (IH) or (IJ) or a stereoisomer, salt or tautomer thereof, wherein: y1 is, at each occurrence, independently an integer from 0 to 6; and y2 and y3 are, at each occurrence, independently an integer from 1 to 6. In some embodiments, y1 is, at each occurrence, an integer 0 (i.e., L6 is a direct bond). In some embodiments, y1 is, at each occurrence, an integer 1. In some embodiments, y1 is, at each occurrence, an integer 2. In some embodiments, y1 is, at each occurrence, an integer 3. In some embodiments, y1 is, at each occurrence, an integer 4. In some embodiments, y1 is, at each occurrence, an integer 5. In some embodiments, y1 is, at each occurrence, an integer 6. In some embodiments, y2 and y3 are at each occurrence, an integer 1 In some embodiments, y2 and y3 are at each occurrence, an integer 2. In some embodiments, y2 and y3 are at each occurrence, an integer 3. In some embodiments, y2 and y3 are at each occurrence, an integer 4. In some embodiments, y2 and y3 are at each occurrence, an integer 5. In some embodiments, y2 and y3 are at each occurrence, an integer 6. In some specific embodiments, y1 is, at each occurrence, an integer 0 or 1; and y2, and y3 are, at each occurrence, 1. The linkers L1b can be used as a point of attachment of the M1 moiety to the remainder of the compound. For example, in some embodiments a synthetic precursor, which is described as structure (II) hereinbelow, to the compound of structure (I) is prepared, and the M1 moiety is attached to the synthetic precursor using any number of coupling methods known in the art for example methods referred to as “click chemistry.” For this purpose, any reaction which is rapid and substantially irreversible can be used to attach M1 to the synthetic precursor to form a compound of structure (I). Exemplary reactions include the copper catalyzed reaction of an azide and alkyne to form a triazole (Huisgen 1, 3-dipolar cycloaddition), reaction of a diene and dienophile (Diels-Alder), strain-promoted alkyne-nitrone cycloaddition, reaction of a strained alkene with an azide, tetrazine or tetrazole, alkene and azide [3+2] cycloaddition, alkene and tetrazine inverse-demand Diels-Alder, alkene and tetrazole photoreaction and various displacement reactions, such as displacement of a leaving group by nucleophilic attack on an electrophilic atom. Exemplary displacement reactions include reaction of an amine with: an activated ester; an N-hydroxysuccinimide ester; an isocyanate; an isothioscyanate or the like. In some embodiments the reaction to form L1b may be performed in an aqueous environment. Accordingly, in some embodiments L1b is, at each occurrence, a linker comprising a functional group capable of formation by reaction of two complementary reactive groups, for example a functional group which is the product of one of the foregoing “click” reactions. In more embodiments, at least one occurrence of L1b comprises a functional group formed by reaction of an aldehyde, oxime, hydrazone, alkyne, amine, azide, acylazide, acylhalide, nitrile, nitrone, sulfhydryl, disulfide, sulfonyl halide, isothiocyanate, imidoester, activated ester, ketone, ^ ^ ^-unsaturated carbonyl, alkene, maleimide, ^-haloamide, epoxide, aziridine, tetrazine, tetrazole, phosphine, biotin, or thiirane with a complementary reactive group. In more specific embodiments, at least one occurrence of L1b comprises a functional group formed by a reaction of an alkyne and an azide. In certain embodiments, at least one occurrence of L1b is a linker comprising a triazolyl functional group. In some embodiments, at least one occurrence of L1b-M1 comprises one of the following structures: or , wherein Lc and Ld are each independently optional linkers. In some embodiments, Lc or Ld, or both, is absent. In some other embodiments, Lc or Ld, or both, is present. In certain embodiments, Lc and Ld, when present, are each independently alkylene or heteroalkylene. In some more certain embodiments, Lc and Ld independently have one of the following structures: ; ; ; ; ; ; or . In some embodiments, M1-L1b has one of the following structures: ; ; or , wherein a, b, c, and d are each independently an integer ranging from 1-6. In some embodiments, a, b, c, and d are each independently an integer 1. In some embodiments, a, b, c, and d are each independently an integer 2. In some embodiments, a, b, c, and d are each independently an integer 3. In some embodiments, a, b, c, and d are each independently an integer 4. In some embodiments, a, b, c, and d are each independently an integer 5. In some embodiments, a, b, c, and d are each independently an integer 6. In some embodiments, at least one occurrence of M1-L1b has one of the following structures: ; ; ; ; or . In some embodiments, each occurrence of M1-L1b has one of the following structures: ; ; ; ; or . In other embodiments, L1b is, at least one occurrence, H. In this regard, the compound has structure (IB), (IC), (IE), (IF), (IH), or (IJ). In other embodiments, L1b is, at each occurrence, H. In this regard, the compound has structure (IB), (IE), or (IH). Some embodiments provide a compound having structure (III): (III) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; L7 is, at each occurrence, independently a linker; L2, L3, L5 and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; R21 is, at each occurrence, independently a natural or unnatural amino acid side chain; R1 and R2 are each independently H, OH, SH, –NH2, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of structure (III); m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, independently an integer of zero or greater; v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties. Compounds of structure (III) have a peptide-based backbone. The various linkers and substituents (e.g., M2, M3, Q, R1, R2, R21, L1a, L1b, L2, L3, L5, L6, L7 and L') in the compound of structure (III) are optionally substituted with one more substituent. For example, in some embodiments the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (III). In certain embodiments, each alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (III) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether, alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether. In certain embodiments the optional substituent is ˗OP(=R R , wherein R is O or S, R is OH, SH, O-, S-, OR or SR , R is OH, SH, O-, S- a)(Rb) c a b d d c , ORd, SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, and Rd is a counter ion. In certain embodiments, substituents are selected to increase cellular or tissue permeation. In related embodiments, substituents are selected to increase cellular or tissue retention.
In some embodiments, at least one R21 is a neutral amino acid side chain. In some embodiments, at least one R21 is a charged amino acid side chain. In some embodiments, R21 is, at each occurrence, independently H, alkyl, -CH2CO2-, -CH2CH2CO2-, -CH2CH2CH2CH2NH3 +,
-CH2CH2CH2NHC(=NH2 +)NH2 or imidazolyl.
In more specific embodiments, R21, L5 and m are selected such that
Figure imgf000035_0001
has an amino acid sequence of (G)io, (GDGDGDGDGD) or (GKGKGKGKGK).
In another embodiment, R21, L5 and m are selected such that
Figure imgf000035_0002
has an amino acid sequence capable of forming an α-helix or 0- sheet secondary structure. In some of those embodiments, the amino acid sequence is (GGEEFMLVYKFARKHGG) or (GGMSMVVSGG).
In some embodiments, L5 or L3, or both, is present for at least one occurrence. In more specific embodiments, when present, L5 or L3, or both, is a heteroalkylene linker. In some of those embodiments, the heteroalkylene linker comprises a functional group capable of maintaining a positive or negative charge at pH values ranging from 3 to 11 in aqueous solution. In more specific embodiments, at least one occurrence of L5 or L3, or both, has the following structure:
Figure imgf000035_0003
In some embodiments, at least one occurrence of L5 or L3, or both, has the following structure: . In some embodiments, L6 and L2 are independently absent or a heteroalkylene linker. In more specific embodiments, the heteroalkylene linker is a peptidyl linker. Some embodiments provide a compound having structure (IV): (IV) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; L7 is, at each occurrence, independently a linker; L2, L5 and L6 are, at each occurrence, independently a direct bond orindependently an optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker; L8 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker comprising one or more charged moieties, provided at least one charged moiety is not a phosphate ester; R3 is, at each occurrence, independently H, alkyl or alkoxy; R1 and R2 are each independently -H, -OH, -SH, alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl, alkoxycarbonyl, ˗OP(=Ra)(Rb)Rc, Q or a protected form thereof, or L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a solid support or solid support residue, a linker comprising a covalent bond to a solid support or solid support residue or a linker comprising a covalent bond to a further compound of structure (IV); q is, at each occurrence, independently an integer of zero or greater; v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties. The various linkers and substituents (e.g., M2, M3, Q, R1, R2, R3, L1a, L1b, L2, L5, L6, L7, L8, and L') in the compound of structure (IV) are optionally substituted with one more substituent. For example, in some embodiments the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (IV). In certain embodiments, each alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker or alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (IV) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether , alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether. In certain embodiments the optional substituent is ˗OP(=Ra)(Rb)Rc, wherein Ra is O or S, Rb is OH, SH, O-, S-, ORd or SRd, Rc is OH, SH, O-, S-, ORd, SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, and Rd is a counter ion. In certain embodiments, substituents are selected to increase cellular or tissue permeation. In related embodiments, substituents are selected to increase cellular or tissue retention. In some embodiments, the alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl and alkyloxycarbonyl are optionally substituted with hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, or combinations thereof. In some embodiments, the charged moieties are positively charged. In more specific embodiments, the charged moieties comprise a protonated amine or quaternary amine functional group. In more specific embodiments, the charged moieties independently have one of the following structures: ; or wherein: R is, at each occurrence, independently H or C1-C6 alkyl. In other embodiments, the charged moieties are negatively charged. In certain embodiments, the charged moieties comprise a carboxylic acid, phosphate or sulfate functional group. In some embodiments, the charged moieties have the following structure: . In some embodiments, the charged moieties comprise a combination of positively charged and negatively charged moieties. In certain embodiments, the charged moieties are pendant to the backbone of the compound (e.g., attached to a linker via an alkylene or heteroalkylene linker). In some embodiments, the charged moiety is part of the backbone of the compound (e.g., part of the contiguous chain of the linker). In some embodiments, L8 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or ’ wherein: R is, at each occurrence, independently H or C1-C6 alkyl; x is an integer from 0 to 6; and m is an integer of 1 or greater, provided that m is selected such that the compound comprises at least two charged moieties. The linker L7 can be used as a point of attachment of the M2 and M3 moieties to the remainder of the compound. For example, in some embodiments a synthetic precursor to the compound of structure (I) is prepared, and the M2 and M3 moieties are attached to the synthetic precursor using any number of coupling methods known in the art. In more embodiments, at least one occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof. In other embodiments, the functional group comprises an amide or an ester. In more specific embodiments, at least one occurrence of L7 or L1b comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or . In more embodiments, each occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof. In other embodiments, the functional group comprises an amide. In more specific embodiments, each occurrence of L7 or L1b comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or . In some embodiments, at least one occurrence of R3 is H. In some embodiments, at least one occurrence of R3 is alkyl. In some embodiments, at least one occurrence of R3 is alkoxy. In certain embodiments, each occurrence of R3 is H. In some embodiments, at least one occurrence of R5 is oxo (=O). In some embodiments, at least one occurrence of R5 is thioxo (=S). In certain embodiments, each occurrence of R5 is oxo (=O). In some embodiments, at least one occurrence of R4 is OH. In some embodiments, at least one occurrence of R4 is SH. In some embodiments, at least one occurrence of R4 is O-. In some embodiments, at least one occurrence of R4 is S-. In certain embodiments, each occurrence of R4 is O-. In other various embodiments, R1 and R2 are each independently OH or ˗OP(=Ra)(Rb)Rc. In some different embodiments, R1 or R2 is OH or ˗OP(=Ra)(Rb)Rc, and the other of R1 or R2 is Q or a linker comprising a covalent bond to Q. In still more different embodiments of any of the foregoing compounds of structures (I), (III), and (IV), R1 and R2 are each independently ˗OP(=Ra)(Rb)Rc. In some of these embodiments, Rc is OL'. In other embodiments, R1 and R2 are each independently ˗OP(=Ra)(Rb)OL', and L' is an alkylene or heteroalkylene linker to: Q, a targeting moiety, an analyte (e.g., analyte molecule), a solid support, a solid support residue, a nucleoside or a further compound of structure (I), (III), or (IV). The linker L' can be any linker suitable for attaching Q, a targeting moiety, an analyte (e.g., analyte molecule), a solid support, a solid support residue, a nucleoside or a further compound of structure (I), (III), or (IV) to the compound of structure (I), (III), or (IV). Advantageously certain embodiments include use of L' moieties selected to increase or optimize water solubility of the compound. In certain embodiments, L' is a heteroalkylene moiety. In some other certain embodiments, L' comprises an alkylene oxide or phosphodiester moiety, or combinations thereof. In some other embodiments, R2 is L'. For example, in some embodiments, L' is a linker to a targeting moiety. In another example, in some embodiments, R2 is –NH2. In other embodiments, one of R2 or R1 is L' and L' is a linker comprising a covalent bond to a solid support. In certain embodiments, the solid support is a polymeric bead or non-polymeric bead. In some specific embodiments, L' is a linker to a targeting moiety, the linker comprising an alkylene oxide or phosphodiester moiety, or combinations thereof. In other embodiments, L' has one of the following structures: ; ; ; or wherein: x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer from 1 to 10; Rb is H, an electron pair or a counter ion; L'' is the targeting moiety or a linkage to the targeting moiety. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 1. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 2. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 3. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 4. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 5. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 6. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 7. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 8. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 9. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 10. In some embodiments, Rb is H. In some embodiments, Rb is a counter ion. For example, in some embodiments, Rb is Na+. In some embodiments, Rb is K+. In some embodiments, L'' is the targeting moiety. In some other embodiments, L'' is a linkage to the targeting moiety. For example, the targeting moiety is an antibody. In another example, the targeting moiety is cell surface receptor antagonist. In some more specific embodiments, the antibody or cell surface receptor antagonist is an epidermal growth factor receptor (EGFR) inhibitor, a hepatocyte growth factor receptor (HGFR) inhibitor, an insulin-like growth factor receptor (IGFR) inhibitor, a folate, or a MET inhibitor. In other embodiments, the targeting moiety is a monoclonal antibody. For instance, in some embodiments, the monoclonal antibody is Abciximab, Adalimumab, Alemtuzumab, Alirocumab, Avibactam, Basiliximab, Benralizumab, Bezlotoxumab, Blinatumomab, Brodalumab, Burosumab, Canakinumab, Caplacizumab, Certolizumab pegol, Daclizumab, Denosumab, Dupilumab, Eculizumab, Emicizumab, Erenumab, Evolocumab, Fremanezumab, Galcanezumab, Golimumab, Guselkumab, Ibalizumab, Idarucizumab, Infliximab, Itolizumab, Ixekizumab, Lanadelumab, Lokivetmab, Mepolizumab, Natalizumab, Obiltoxaximab, Ocrelizumab, Omalizumab, Palivizumab, Ranibizumab, Raxibacumab, Reslizumab, Rmab, Rovelizumab, Ruplizumab, Sarilumab, Secukinumab, Tildrakizumab, Thiomab, Tocilizumab, Ustekinumab, Vedolizumab, Abrilumab, Actoxumab, Aducanumab, Afasevikumab, Afelimomab, Anifrolumab, Anrukinzumab (IMA-638), Aselizumab, Atorolimumab, Bapineuzumab, BCD-100, Bertilimumab, Besilesomab, Biciromab, Bimagrumab, Bimekizumab, Birtamimab, Bleselumab, Blosozumab, Bococizumab, Brazikumab, Briakinumab, Brolucizumab, Carlumab, Carotuximab, Cedelizumab, Clazakizumab, Clenoliximab, Concizumab, Cosfroviximab, CR6261, Crenezumab, Crizanlizumab, Crotedumab, Depatuxizumab, mafodotin, Derlotuximab biotin, Dezamizumab, Diridavumab, Domagrozumab, Dusigitumab, Ecromeximab, Edobacomab, Efalizumab, Efungumab, Eldelumab, Elezanumab, Enokizumab, Eptinezumab, Erlizumab, Etrolizumab, Evinacumab, Exbivirumab, Fanolesomab, Faralimomab, Faricimab, Fasinumab, Felvizumab, Fezakinumab, Flanvotumab, Fletikumab, Flotetuzumab, Fontolizumab, Foravirumab, Frovocimab, Fulranumab, Gantenerumab, Gavilimomab, Gevokizumab, Gimsilumab, Gomiliximab, Gosuranemab, Ianalumab, Inclacumab, Inolimomab, Iomab-B, Keliximab, Lampalizumab, Landogrozumab, Larcaviximab, Lebrikizumab, Lenvervimab, Lerdelimumab, Letolizumab, Libivirumab, Ligelizumab, Lodelcizumab, Lulizumab pegol, Marstacimab, Mavrilimumab, Metelimumab, Mirikizumab, Motavizumab, Muromonab CD3, Nebacumab, Nemolizumab, NEOD001, Nirsevimab, Odulimomab, Olendalizumab, Olokizumab, OMS721, Opicinumab, Orticumab, Otelixizumab, Otilimab, Oxelumab, Ozanezumab, Ozoralizumab, Pagibaximab, Panobacumab, Pascolizumab, Pateclizumab, PDR001, Perakizumab, Pexelizumab, Placulumab, Plozalizumab, Ponezumab, Porgaviximab, Prasinezumab, Priliximab, PRO 140, Quilizumab, Rafivirumab, Ralpancizumab, Ranevetmab, Ravagalimab, Ravulizumab, Refanezumab, Regavirumab, Relatlimab, Rinucumab, Risankizumab, Roledumab, Romosozumab, Rontalizumab, SA237, Satralizumab, Sevirumab, SHP647, Sifalimumab, Simtuzumab, Siplizumab, Sirukumab, Solanezumab, Sonepcizumab, Spartalizumab, Stamulumab, Sulesomab, Suptavumab, Sutimlimab, Suvizumab, Suvratoxumab, Tadocizumab, Talizumab, Tamtuvetmab, Tanezumab, Tefibazumab, Telimomab aritox, Teneliximab, Teplizumab, Teprotumumab, Tezepelumab, Tibulizumab, Toralizumab, Tralokinumab, Trevogrumab, Tuvirumab, Ulocuplumab, Urtoxazumab, Varisacumab, Vepalimomab, Vesencumab, Visilizumab, Vobarilizumab, Zolimomab aritox, trastuzumab, gemtuzumab, brentuximab, vorsetuzumab, lorvotuzumab, cantuzumab, bivatuzumabor inotuzumab, or vadastuximab. In other more specific embodiments of any of the foregoing compounds of structure (I), R1 or R2 has one of the following structures: ; ; ; ; ; ; ; ; ; ; or , wherein Ra is H or a solid support. In some specific embodiments, R2 has one of the following structures: ; or . . Certain embodiments of compounds of structure (I) can be prepared according to solid- phase synthetic methods analogous to those known in the art for preparation of oligonucleotides. Accordingly, in some embodiments, L' is a linkage to a solid support, a solid support residue or a nucleoside. Solid supports comprising an activated deoxythymidine (dT) group are readily available, and in some embodiments can be employed as starting material for preparation of compounds of structure (I). Accordingly, in some specific embodiments, R1 has the following structure: . One of skill in the art will understand that the dT group depicted above is included for ease of synthesis and economic efficiencies only, and is not required. Other solid supports can be used and would result in a different nucleoside or solid support residue being present on L', or the nucleoside or solid support residue can be removed or modified post synthesis. In some embodiments, the analyte molecule is a nucleic acid, amino acid or a polymer thereof. In some embodiments, the analyte molecule is an enzyme, receptor, receptor ligand, antibody, glycoprotein, aptamer or prion. In some embodiments, the targeting moiety is an antibody or cell surface receptor antagonist. In further some embodiments, the solid support is a polymeric bead or non-polymeric bead. In still other embodiments, Q is, at each occurrence, independently a moiety comprising a reactive group capable of forming a covalent bond with an analyte molecule or a solid support. In other embodiments, Q is, at each occurrence, independently a moiety comprising a reactive group capable of forming a covalent bond with a complementary reactive group Q′. For example, in some embodiments, Q′ is present on a further compound of structure (I) (e.g., in the R1 or R2 position), and Q and Q′ comprise complementary reactive groups such that reaction of the compound of structure (I) and the further compound of structure (I) results in covalently bound dimer of the compound of structure (I). Multimer compounds of structure (I) can also be prepared in an analogous manner and are included within the scope of embodiments of the disclosure. The type of Q group and connectivity of the Q group to the remainder of the compound of structure (I) is not limited, provided that Q comprises a moiety having appropriate reactivity for forming the desired bond. In certain embodiments, Q is a moiety which is not susceptible to hydrolysis under aqueous conditions, but is sufficiently reactive to form a bond with a corresponding group on an analyte molecule or solid support (e.g., an amine, azide or alkyne). Certain embodiments of compounds of structure (I) comprise Q groups commonly employed in the field of bioconjugation. For example in some embodiments, Q comprises a nucleophilic reactive group, an electrophilic reactive group or a cycloaddition reactive group. In some more specific embodiments, Q comprises a sulfhydryl, disulfide, activated ester, isothiocyanate, azide, alkyne, alkene, diene, dienophile, acid halide, sulfonyl halide, phosphine, ^-haloamide, biotin, amino or maleimide functional group. In some embodiments, the activated ester is an N-succinimide ester, imidoester or polyfluorophenyl ester. In other embodiments, the alkyne is an alkyl azide or acyl azide. The Q groups can be conveniently provided in protected form to increase storage stability or other desired properties, and then the protecting group removed at the appropriate time for conjugation with, for example, a targeting moiety or analyte. Accordingly, Q groups include "protected forms" of a reactive group, including any of the reactive groups described above and in the Table 1 below. A "protected form" of Q refers to a moiety having lower reactivity under predetermined reaction conditions relative to Q, but which can be converted to Q under conditions, which preferably do not degrade or react with other portions of the compound of structure (I). One of skill in the art can derive appropriate protected forms of Q based on the particular Q and desired end use and storage conditions. For example, when Q is SH, a protected form of Q includes a disulfide, which can be reduced to reveal the SH moiety using commonly known techniques and reagents. Exemplary Q moieties are provided in Table I below. Table 1. Exemplary Q Moieties
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
It should be noted that in some embodiments, wherein Q is SH, the SH moiety will tend to form disulfide bonds with another sulfhydryl group, for example on another compound of structure (I). Accordingly, some embodiments include compounds of structure (I), which are in the form of disulfide dimers, the disulfide bond being derived from SH Q groups. In other embodiments, the Q moiety is conveniently masked (e.g., protected) as a disulfide moiety, which can later be reduced to provide an activated Q moiety for binding to a desired analyte molecule or targeting moiety. For example, the Q moiety may be masked as a disulfide having the following structure: wherein R is an optionally substituted alkyl group. For example, in some embodiments, Q is provided as a disulfide moiety having the following structure: where n is an integer from 1 to 10. In some embodiments, M2 is, at one or more occurrences, independently a moiety comprising four or more aryl or heteroaryl rings, or combinations thereof. In some embodiments, M2 is, at one or more occurrences, independently fluorescent or colored. In certain embodiments, M2 is, at one or more occurrences, fluorescent. In other embodiments, M2 is, at one or more occurrences, independently comprise a fused-multicyclic aryl or heteroaryl moiety comprising at least four fused rings. In some embodiments, M2 is, at each occurrence, independently selected from the group consisting of phenyl, (bis-fluorophenyl-difluorobora- diaza-indacene)phenyl, quaterphenyl, bi-benzothiazole, ter-benzothiazole, bi-naphthyl, bi- anthracyl, squaraine, squarylium, 9,10-ethynylanthracene, and ter-naphthyl moiety. In some other embodiments, M2 is, at each occurrence, independently selected from the group consisting of p-terphenyl, perylene, azobenzene, phenazine, phenanthroline, acridine, thioxanthrene, chrysene, rubrene, coronene, cyanine, perylene imide, perylene amide, and derivatives thereof. In some other embodiments, M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, resorufin dye, dipyrrometheneboron difluoride dye, ruthenium bipyridyl dye, thiazole orange dye, polymethine, and N-aryl-1,8-naphthalimide dye. Further in some other embodiments, M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, boron-dipyrromethene, rhodamine, cyanine, pyrene, perylene, perylene monoimide, 6-carboxyfluorescein (6-FAM), 5-carboxyfluorescein (5-FAM), 6- fluorescein isothiocyanate (6-FITC), 5-fluorescein isothiocyanate (5-FITC), and derivatives thereof. In certain embodiments, M2, at each occurrence, independently has one of the following structures:
; ; ; ; ; ; or . In some specific embodiments, at least one occurrence of M2 has the following structure: . In some more specific embodiments, each occurrence of M2 has the following structure: . In some embodiments, at least one occurrence of –L7- M2 has one of the following structures: . In some more specific embodiments, each occurrence of –L7- M2 has one of the following structures: . M1 and M3 are selected based on the desired alkylating properties. In some embodiments, M1 and M3 are the same at each occurrence; however, it is important to note that each occurrence of M1 and M3 need not be an identical M1 and M3, and certain embodiments include compounds wherein M1 and M3 are not the same at each occurrence. For example, in some embodiments each M1 and M3 are not the same and the different M1 and M3 moieties are selected to have different alkylating agents. Exemplary M1 and M3 moieties can be appropriately selected by one of ordinary skill in the art based on the desired end use. Further, in some embodiments, alkylating agents are protected by protecting groups such as an allyloxycarbonyl group (–Alloc) and a tert-butyl dimethyl silyl ether (-TBS) in order to survive during the DNA synthesis cycle. Alloc protecting group can be easily cleaved by a palladium catalyst, for example, Pd(PPh3)4 with PhSiH3 to afford the corresponding amine. TBS protecting group can be cleaved by a fluoride source such as tetra-n-butylammonium fluoride (TBAF) to afford the corresponding alcohol. Once the deprotections are complete, PBD moiety becomes active with alkylating capability. In this regard, PBD moiety is protected with protecting groups until the DNA synthesis cycle is completed. Then, the deprotections allow for converting the protected PBD moiety which is inactive in alkylating into the deprotected PBD moiety which is active in alkylating. In some embodiments, at least one occurrence of M1 or M3 moieties are an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic. In more specific embodiments, each occurrence of M1 or M3 is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic. In certain embodiments, at least one occurrence of M1 or M3 is a nitrogen mustard, a nitrosourea, a tetrazine, an aziridine, a cisplatin or cisplatin derivative, or a non-classical alkylating agent. In more specific embodiments, at least one occurrence of M1 or M3 is mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide, busulfan, N-nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU), semustine (MeCCNU), fotemustine, streptozotocin, dacarbazine, mitozolomide, temozolomide, thiotepa, mytomycin, diaziquone (AZQ), cisplatin, carboplatin, oxaliplatin, procarbazine, or hexamethylmelamine. In some embodiments, at least one occurrence of M1 or M3 is an anti-folate, a fluoropyrimidines, a deoxynucleoside analogue, or a thiopurine. In certain embodiments, at least one occurrence of M1 or M3 is methotrexate, pemetrexed, fluorouracil, capecitabine, cytarabine, gemcitabine, decitabine, azacitidine, fludarabine, nelarabine, cladribine, clofarabine, pentostatin, thioguanine, and mercaptopurine. In some specific embodiments, at least one occurrence of M1 or M3 is an auristatin, a Vinca alkaloid, or a taxane. In certain specific embodiments, at least one occurrence of M1 or M3 is auristatin F, auristatin E, vincristine, vinblastine, vinorelbine, vindesine, vinflunine, paclitaxel, docetaxel, etoposide, or teniposide. In some more specific embodiments, at least one occurrence of M1 or M3 is irinotecan, SN 38, topotecan, camptothecin, doxorubicin, mitoxantrone, teniposide. novobiocin, merbarone, or aclarubicin. In certain more specific embodiments, at least one occurrence of M1 or M3 is an anthracycline or a bleomycin. In some embodiments, at least one occurrence of M1 or M3 is doxorubicin, daunorubicin, epirubicin, idarubicin, pirarubicin, aclarubicin, or mitoxantrone. In some embodiments, at least one occurrence of M1 or M3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan. In some embodiments, each occurrence of M1 or M3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan. M1 or M3 may be attached to the remainder of the molecule from any position (i.e., atom) on M1 or M3, respectively. One of skill in the art will recognize means for attaching M1 or M3 to the remainder of molecule. For example, M1 or M3 may be attached to the remainder of the molecule through nitrogen of diazepine, oxygen of diazepine or phenyl ring, or carbon of pyrrolidine ring. In some embodiments, at least one occurrence of M1 or M3 has one of the following structures: ; ; ; ; ; ; ; ; ; or . In some embodiments, each occurrence of M1 or M3 has one of the following structures: ; ; ; ; ; ; ; ; ; or . In some specific embodiments, the compound is a compound selected from Table 2A. The compounds in Table 2A are prepared according to the procedures set forth in the Examples.
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
wherein M3 has the following structure: , wherein M1 has one of the following structures: ; ; ; ; ; ; ; ; ; or . Further in some embodiments, at least one occurrence of M3 is an anti-inflammatory compound. M3 is a steroid which is a biologically active organic compound with four rings arranged in specific molecular configuration as shown below. For example, in some embodiments, at least one occurrence of M3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone. In some more specific embodiments, at least one occurrence of M3 has one of the following structures: or , wherein: R11 is H or halogen; R12 and R13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R14 is H, halogen, OH, or alkyl; R15 is H or OH; R16, R17, and R18, are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond. In some embodiments, at least one occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or . In some specific embodiments, the compound is a compound selected from Table 2B. The compounds in Table 2B are prepared according to the procedures set forth in the Examples.
Figure imgf000088_0001
.o 0 N 3- I
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
wherein M3 has one of the following structures: ; ; ; ; or , wherein or is selected from the following structures: ; ; ; ; ; ; ; ; ; ; ; or , wherein M1 has one of the following structures: ; ; ; ; ; ; ; ; ; or . In some embodiments, at least one occurrence of M3 is a Vitamin. For example, in some embodiments, at least one occurrence of M3 is a Vitamin E. In some more specific embodiments, at least one occurrence of M3 is a tocopherol. In certain embodiments, at least one occurrence of M3 is α-tocopherol, β-tocopherol, γ-tocopherol, δ-tocopherol, α -tocotrienol, β - tocotrienol, γ -tocotrienol, or δ -tocotrienol. In some embodiments, at least one occurrence of M3 has the following structure: , wherein: R19 and R20 are independently H or CH3; and represents a single or double carbon-carbon bond. In some specific embodiments, at least one occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; or . In some specific embodiments, the compound is a compound selected from Table 2C. The compounds in Table 2C are prepared according to the procedures set forth in the Examples.
Figure imgf000102_0001
a T .o 0 N 4- I
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
wherein M3 has one of the following structures: ; ; ; ; or , wherein is selected from the following structures: ; ; ; ; ; ; ; or , wherein M1 has one of the following structures: ; ; ; ; ; ; ; ; ; or . In some specific embodiments, the compounds of structure (III) are selected from Table 2D. The compounds in Table 2D are prepared according to the procedures set forth in the Examples.
Figure imgf000114_0001
.o 1 N -I 2 I I -I I I
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
In some specific embodiments, the compounds of structure (IV) are selected from Table 2E. The compounds in Table 2E are prepared according to the procedures set forth in the Examples.
Figure imgf000119_0001
.o 1- N V I
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
As used in Tables 2D-2E and throughout the application wherein M3 has one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or . The fluorescence intensity or the effectiveness toward alkylation of DNA can also be tuned by selection of different values of n. In some embodiments, n is an integer of one or greater for structure (I). In certain embodiments, n is an integer from 1 to 100 for structure (I). In other embodiments, n is an integer from 1 to 10 for structure (I). In some embodiments, n is 1 for structure (I). In some embodiments, n is 2 for structure (I). In some embodiments, n is 3 for structure (I). In some embodiments, n is 4 for structure (I). In some embodiments, n is 5 for structure (I). In some embodiments, n is 6 for structure (I). In some embodiments, n is 7 for structure (I). In some embodiments, n is 8 for structure (I). In some embodiments, n is 9 for structure (I). In some embodiments, n is 10 for structure (I). In some embodiments of structures (III) and (IV), n is an integer of two or greater. In certain embodiments, n is an integer from 2 to 100 for structures (III) and (IV). In other embodiments, n is an integer from 2 to 10 for structures (III) and (IV). In some embodiments, n is 2 for structures (III) and (IV). In some embodiments, n is 3 for structures (III) and (IV). In some embodiments, n is 4 for structures (III) and (IV). In some embodiments, n is 5 for structures (III) and (IV). In some embodiments, n is 6 for structures (III) and (IV). In some embodiments, n is 7 for structures (III) and (IV). In some embodiments, n is 8 for structures (III) and (IV). In some embodiments, n is 9 for structures (III) and (IV). In some embodiments, n is 10 for structures (III) and (IV). The fluorescence or the effectiveness toward alkylation of DNA may also be tuned by selection of values for m. The value for m has the ability to control the spacing between neighboring M1, M2, or M3. In some embodiments, m is an integer of zero or greater. In certain embodiments, m is an integer from 0 to 100. In certain embodiments, m is an integer from 0 to 10. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments, m is 5. In some embodiments, m is 6. In some embodiments, m is 7. In some embodiments, m is 8. In some embodiments, m is 9. In some embodiments, m is 10. In some certain embodiments, m is 1 or 2. The fluorescence intensity may be tuned how many fluorescent dye moieties are attached to the polymeric backbone. The value for q has the ability to control the brightness of compounds. In some embodiments, q is, at each occurrence, an integer of zero or greater. In some more specific embodiments, q is from 0 to 10. In some embodiments, q is from 0 to 5. For example, in some embodiments, q is 0. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, q is 3. In some embodiments, q is 4. In some embodiments, q is 5. In some other embodiments, q is 0, 1, 2, 3, or 4. In some other embodiments, q is 1 or 2. The effectiveness toward alkylation of DNA may also be tuned by selection of values for p. In some embodiments, p is, at each occurrence, an integer of zero or greater. In some more specific embodiments, p is from 0 to 10. In some embodiments, p is from 0 to 5. For example, in some embodiments, p is 0. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, p is 4. In some embodiments, p is 5. In some other embodiments, p is 1, 2, 3, or 4. In some other embodiments, p is 2 or 3. The effectiveness toward alkylation of DNA may also be tuned by selection of values for w. In some embodiments, w is, at each occurrence, an integer of zero or greater. In some more specific embodiments, w is from 0 to 10. In some embodiments, w is from 0 to 5. In some embodiments, w is from 1 to 5. For example, in some embodiments, w is 1. In some embodiments, w is 2. In some embodiments, w is 3. In some embodiments, w is 4. In some embodiments, w is 5. In some other embodiments, w is 1, 2, 3, or 4. In some other embodiments, w is 1 or 2. In some embodiments of structure (III) or (IV), v is an integer of one or greater. For example, in some embodiments, v is an integer from 1 to 100. Further in some embodiments, v is an integer from 1 to 10. In some embodiments, v is 1. In some embodiments, v is 2. In some embodiments, v is 3. In some embodiments, v is 4. In some embodiments, v is 5. In some embodiments, v is 6. In some embodiments, v is 7. In some embodiments, v is 8. In some embodiments, v is 9. In some embodiments, v is 10. The value n, q, w, p, and m are closely related to provide the ability to control the fluorescence and effectiveness toward alkylation of DNA. In some specific embodiments, n is 1; q is 2; w is 2, p is 2; and m is 1. In some other specific embodiments, n is 1; q is 2; w is 1, p is 3; and m is 1. The ratio of a biological active moiety on M1, a fluorescent dye M2, and a biological active moiety on M3 can be fine-tuned based on a type of disease, progression of the disease, a therapeutic method, and/or diagnostic method used for a treatment. For example, in some embodiments, a ratio of M1:M2:M3 is 1:1:1. In some other embodiments, M1:M2:M3 is 1:2:3. In some more other embodiments, M1:M2:M3 is 2:1:1. In other embodiments, M1:M2:M3 is 1:0:1 when q is 0. In other specific embodiments, M1:M2:M3 is 1:0:2 when q is 0. Further in other embodiments, M1:M2:M3 is 2:0:1 when q is 0. Pharmaceutical Compositions One embodiment provides a composition comprising the compound according to any one of the embodiments disclosed herein (e.g., a compound of Structure (I), (III), or (IV)) and a pharmaceutically acceptable carrier. Other embodiments are directed to pharmaceutical compositions. The pharmaceutical composition comprises any one (or more) of the compounds of Structure (I), (III), or (IV) and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is formulated for oral administration. In other embodiments, the pharmaceutical composition is formulated for injection. In still more embodiments, the pharmaceutical compositions comprise a compound of Structure (I), (III), or (IV) and an additional therapeutic agent (e.g., anticancer agent). Non-limiting examples of such therapeutic agents are described herein below. Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration. In addition, by way of example only, parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections. In certain embodiments, a compound of Structure (I), (III), or (IV) is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation. In specific embodiments, long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Furthermore, in other embodiments, the drug is delivered in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody. In such embodiments, the liposomes are targeted to and taken up selectively by the organ. In yet other embodiments, the compound of Structure (I), (III), or (IV) is provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation. In yet other embodiments, the compound of Structure (I), (III), or (IV) is administered topically. The compounds of Structure (I), (III), or (IV) are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that are used in some embodiments. An exemplary dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician. In some embodiments, a compound of Structure (I), (III), or (IV) is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes are used as appropriate. A single dose of a compound of Structure (I), (III), or (IV) may also be used for treatment of an acute condition. In some embodiments, a compound of Structure (I), (III), or (IV) is administered in multiple doses. In some embodiments, dosing is about once, twice, three times, four times, five times, six times, or more than six times per day. In other embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of Structure (I), (III), or (IV) and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of Structure (I), (III), or (IV) and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary. Administration of the compounds of Structure (I), (III), or (IV) may continue as long as necessary. In some embodiments, a compound of Structure (I) is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, a compound of Structure (I), (III), or (IV) is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, a compound of Structure (I), (III), or (IV) is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects. In some embodiments, the compounds of Structure (I), (III), or (IV) are administered in dosages. It is known in the art that due to inter-subject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the disclosure may be found by routine experimentation in light of the instant disclosure. In some embodiments, the compounds of Structure (I), (III), or (IV) are formulated into pharmaceutical compositions. In specific embodiments, pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are used as suitable to formulate the pharmaceutical compositions described herein: Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins1999). Provided herein are pharmaceutical compositions comprising a compound of Structure (I), (III), or (IV) and a pharmaceutically acceptable diluent(s), excipient(s), or carrier(s). In certain embodiments, the compounds described are administered as pharmaceutical compositions in which compounds of Structure (I), (III), or (IV) are mixed with other active ingredients, as in combination therapy. Encompassed herein are all combinations of actives set forth in the combination therapies section below and throughout this disclosure. In specific embodiments, the pharmaceutical compositions include one or more compounds of Structure (I), (III), or (IV). A pharmaceutical composition, as used herein, refers to a mixture of a compound of Structure (I), (III), or (IV) with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. In certain embodiments, the pharmaceutical composition facilitates administration of the compound to an organism. In some embodiments, practicing the methods of treatment or use provided herein, therapeutically effective amounts of compounds of Structure (I), (III), or (IV) provided herein are administered in a pharmaceutical composition to a mammal having a disease, disorder or medical condition to be treated. In specific embodiments, the mammal is a human. In certain embodiments, therapeutically effective amounts vary depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. The compounds of Structure (I), (III), or (IV) are used singly or in combination with one or more therapeutic agents as components of mixtures. In one embodiment, one or more compounds of Structure (I), (III), or (IV) is formulated in an aqueous solution. In specific embodiments, the aqueous solution is selected from, by way of example only, a physiologically compatible buffer, such as Hank’s solution, Ringer’s solution, or physiological saline buffer. In other embodiments, one or more compound of Structure (I), (III), or (IV) is/are formulated for transmucosal administration. In specific embodiments, transmucosal formulations include penetrants that are appropriate to the barrier to be permeated. In still other embodiments wherein the compounds described herein are formulated for other parenteral injections, appropriate formulations include aqueous or non-aqueous solutions. In specific embodiments, such solutions include physiologically compatible buffers and/or excipients. In another embodiment, compounds described herein are formulated for oral administration. Compounds described herein are formulated by combining the active compounds with, e.g., pharmaceutically acceptable carriers or excipients. In various embodiments, the compounds described herein are formulated in oral dosage forms that include, by way of example only, tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like. In certain embodiments, pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. In specific embodiments, disintegrating agents are optionally added. Disintegrating agents include, by way of example only, cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. In one embodiment, dosage forms, such as dragee cores and tablets, are provided with one or more suitable coating. In specific embodiments, concentrated sugar solutions are used for coating the dosage form. The sugar solutions, optionally contain additional components, such as by way of example only, gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs and/or pigments are also optionally added to the coatings for identification purposes. Additionally, the dyestuffs and/or pigments are optionally utilized to characterize different combinations of active compound doses. In certain embodiments, therapeutically effective amounts of at least one of the compounds described herein are formulated into other oral dosage forms. Oral dosage forms include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. In specific embodiments, push-fit capsules contain the active ingredients in admixture with one or more filler. Fillers include, by way of example only, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In other embodiments, soft capsules, contain one or more active compound that is dissolved or suspended in a suitable liquid. Suitable liquids include, by way of example only, one or more fatty oil, liquid paraffin, or liquid polyethylene glycol. In addition, stabilizers are optionally added. In other embodiments, therapeutically effective amounts of at least one of the compounds described herein are formulated for buccal or sublingual administration. Formulations suitable for buccal or sublingual administration include, by way of example only, tablets, lozenges, or gels. In still other embodiments, the compounds described herein are formulated for parental injection, including formulations suitable for bolus injection or continuous infusion. In specific embodiments, formulations for injection are presented in unit dosage form (e.g., in ampoules) or in multi-dose containers. Preservatives are, optionally, added to the injection formulations. In still other embodiments, the pharmaceutical compositions are formulated in a form suitable for parenteral injection as sterile suspensions, solutions or emulsions in oily or aqueous vehicles. Parenteral injection formulations optionally contain formulatory agents such as suspending, stabilizing and/or dispersing agents. In specific embodiments, pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. In additional embodiments, suspensions of the active compounds (e.g., compounds of Structure (I), (III), or (IV)) are prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles for use in the pharmaceutical compositions described herein include, by way of example only, fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. In certain specific embodiments, aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension contains suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, in other embodiments, the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. In still other embodiments, the compounds of Structure (I), (III), or (IV) are administered topically. The compounds described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments. Such pharmaceutical compositions optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives. In yet other embodiments, the compounds of Structure (I), (III), or (IV) are formulated for transdermal administration. In specific embodiments, transdermal formulations employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive. In various embodiments, such patches are constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. In additional embodiments, the transdermal delivery of the compounds of Structure (I), (III), or (IV) is accomplished by means of iontophoretic patches and the like. In certain embodiments, transdermal patches provide controlled delivery of the compounds of Structure (I), (III), or (IV). In specific embodiments, the rate of absorption is slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel. In alternative embodiments, absorption enhancers are used to increase absorption. Absorption enhancers or carriers include absorbable pharmaceutically acceptable solvents that assist passage through the skin. For example, in one embodiment, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. In other embodiments, the compounds of Structure (I), (III), or (IV) are formulated for administration by inhalation. Various forms suitable for administration by inhalation include, but are not limited to, aerosols, mists or powders. Pharmaceutical compositions of any of compound of Structure (I), (III), or (IV) are 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 specific embodiments, the dosage unit of a pressurized aerosol is determined by providing a valve to deliver a metered amount. In certain embodiments, capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator is formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. In still other embodiments, the compounds of Structure (I), (III), or (IV) are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like. In suppository forms of the compositions, a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with melted cocoa butter. In certain embodiments, pharmaceutical compositions are formulated in any conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are optionally used as suitable. Pharmaceutical compositions comprising a compound of Structure (I), (III), or (IV) are manufactured in a conventional manner, such as, by way of example only, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes. Pharmaceutical compositions include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of Structure (I), (III), or (IV), described herein as an active ingredient. The active ingredient is in free-acid or free-base form, or in a pharmaceutically acceptable salt form. In addition, the methods and pharmaceutical compositions described herein include the use of N-oxides, crystalline forms (also known as polymorphs), as well as active metabolites of these compounds having the same type of activity. All tautomers of the compounds described herein are included within the scope of the compounds presented herein. Additionally, the compounds described herein encompass unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein. In addition, the pharmaceutical compositions optionally include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, buffers, and/or other therapeutically valuable substances. Methods for the preparation of compositions comprising the compounds described herein include formulating the compounds with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid or liquid. Solid compositions include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories. Liquid compositions include solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein. Semi-solid compositions include, but are not limited to, gels, suspensions and creams. The form of the pharmaceutical compositions described herein include liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions also optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth. In some embodiments, pharmaceutical composition comprising at least one compound of Structure (I), (III), or (IV) illustratively takes the form of a liquid where the agents are present in solution, in suspension or both. Typically, when the composition is administered as a solution or suspension a first portion of the agent is present in solution and a second portion of the agent is present in particulate form, in suspension in a liquid matrix. In some embodiments, a liquid composition includes a gel formulation. In other embodiments, the liquid composition is aqueous. In certain embodiments, useful aqueous suspensions contain one or more polymers as suspending agents. Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross- linked carboxyl-containing polymers. Certain pharmaceutical compositions described herein comprise a mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran. Useful pharmaceutical compositions also, optionally, include solubilizing agents to aid in the solubility of a compound of Structure (I), (III), or (IV). The term "solubilizing agent" generally includes agents that result in formation of a micellar solution or a true solution of the agent. Certain acceptable nonionic surfactants, for example polysorbate 80, are useful as solubilizing agents, as can ophthalmically acceptable glycols, polyglycols, e.g., polyethylene glycol 400, and glycol ethers. Furthermore, useful pharmaceutical compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride. Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range. Additionally, useful compositions also, optionally, include one or more salts in an amount required to bring osmolality of the composition into an acceptable range. Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate. Other useful pharmaceutical compositions optionally include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride. Still other useful compositions include one or more surfactants to enhance physical stability or for other purposes. Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40. Still other useful compositions include one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example only, ascorbic acid and sodium metabisulfite. In certain embodiments, aqueous suspension compositions are packaged in single-dose non-reclosable containers. Alternatively, multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition. In alternative embodiments, other delivery systems for hydrophobic pharmaceutical compounds are employed. Liposomes and emulsions are examples of delivery vehicles or carriers useful herein. In certain embodiments, organic solvents such as N-methylpyrrolidone are also employed. In additional embodiments, the compounds described herein are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials are useful herein. In some embodiments, sustained-release capsules release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization are employed. In certain embodiments, the formulations described herein comprise one or more antioxidants, metal chelating agents, thiol containing compounds and/or other general stabilizing agents. Examples of such stabilizing agents, include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1% w/v methionine, (c) about 0.1% to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v. polysorbate 20, (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (l) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof. In some embodiments, the concentration of one or more compounds provided in the pharmaceutical compositions is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%,14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v or v/v. In some embodiments, the concentration of one or more compounds is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%, 4.25%, 4%, 3.75%, 3.50%, 3.25%, 3%, 2.75%, 2.50%, 2.25%, 2%, 1.75%, 1.50%, 125% , 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v, or v/v. In some embodiments, the concentration of one or more compounds is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v. In some embodiments, the concentration of one or more compounds is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v. In some embodiments, the amount of one or more compounds is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.008 g, 0.007 g, 0.006 g, 0.005 g, 0.004 g, 0.003 g, 0.002 g, 0.001 g, 0.0009 g, 0.0008 g, 0.0007 g, 0.0006 g, 0.0005 g, 0.0004 g, 0.0003 g, 0.0002 g, or 0.0001 g. In some embodiments, the amount of one or more compounds is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075 g, 0.08 g, 0.085 g, 0.09 g, 0.095 g, 0.1 g, , 0.15 g, 0.2 g, , 0.25 g, 0.3 g, , 0.35 g, 0.4 g, , 0.45 g, 0.5 g, 0.55 g, 0.6 g, , 0.65 g, 0.7 g, 0.75 g, 0.8 g, 0.85 g, 0.9 g, 0.95 g, 1 g, 1.5 g, 2 g, 2.5, 3 g, 3.5, 4 g, 4.5 g, 5 g, 5.5 g, 6 g, 6.5g, 7 g, 7.5g, 8 g, 8.5 g, 9 g, 9.5 g, or 10 g. In some embodiments, the amount of one or more compounds ranges from 0.0001 to 10 g, 0.0005 to 9 g, 0.001 to 8 g, 0.005 to 7 g, 0.01 to 6 g, 0.05 to 5 g, 0.1 to 4 g, 0.5 to 4 g, or 1 to 3 g. Method of Treatment Certain compounds of the present disclosure are useful for treating disease (i.e., compounds of Structure (I), (III), or (IV)). Those compounds disclosed herein offer a targeted approach to drug delivery strategies. Accordingly, one embodiment provides a method of treating a disease (or the symptoms thereof) comprising administering to a mammal (e.g., a human) in need thereof a therapeutically effective amount of a compound of Structure (I), (III), or (IV). For example, in certain embodiments the disclosure provides a method of treating solid tumors, multiple myeloma, gliomas, clear cell renal cell carcinoma, prostate cancer, ovarian cancer, non-small cell lung cancer, GI malignancies, acute lymphoblastic leukemia, acute myelogenous leukemia, renal cell carcinoma, colorectal carcinoma, epithelial cancers, pancreatic and gastric cancers, renal cell carcinoma, non-Hodgkin’s lymphoma, metastatic renal cell carcinoma, malignant mesothelioma, pancreatic, ovarian, and/or lung adenocarcinoma, B-cell malignancies, breast cancer, melanoma, recurrent multiple myeloma, small cell lung cancer, CD22-positive B cell malignancies, Hodgkin’s lymphoma/anaplastic large cell lymphoma, or HER2-positive breast cancer. In some of the foregoing embodiments, the disease is cancer. For example, in certain embodiments, the cancer is breast cancer, non-Hodgkin's lymphoma, acute myeloid leukemia, multiple myeloma, gastric cancer, renal cell carcinoma, solid tumors, ovarian cancer, prostate cancer, colorectal cancer, pancreatic cancer, small cell lung cancer, diffuse large B-cell lymphoma, a neoplasm, urothelial cancer, ALL, CLL, glioblastoma, Hodgkin's lymphoma, lymphoma, mesothelioma, non-small cell lung cancer, recurrent head and neck cancer, or a combination thereof. Certain embodiments also relate to a method of treating a hyperproliferative disorder in a mammal (e.g., a human) that comprises administering to said mammal a therapeutically effective amount of a compound of Structure (I), (III), or (IV), or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In some embodiments, said method relates to the treatment of cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g., Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin’s lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer, multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic / myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin’s lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T- Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or Viral- Induced cancer. In some embodiments, said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)). Certain particular embodiments provide methods for treatment of lung cancers, the methods comprise administering an effective amount of any of the above described compounds of Structure (I), (III), or (IV) (or a pharmaceutical composition comprising the same) to a subject in need thereof. In certain embodiments the lung cancer is a non-small cell lung carcinoma (NSCLC), for example adenocarcinoma, squamous-cell lung carcinoma or large-cell lung carcinoma. In other embodiments, the lung cancer is a small cell lung carcinoma. Other lung cancers treatable with the disclosed compounds include, but are not limited to, glandular tumors, carcinoid tumors and undifferentiated carcinomas. Accordingly, in some embodiments of Structure (I), (III), or (IV) A is an antibody or a cell surface receptor antagonist. For example, epidermal growth factor receptor (EGFR) inhibitor, a hepatocyte growth factor receptor (HGFR) inhibitor, an insulin-like growth factor receptor (IGFR) inhibitor, a folate, or a MET inhibitor. In even more embodiments, the method further comprises inducing apoptosis. In some embodiments, the method of treatment comprises treating a tumor having tumor cells with tumor cell receptors. In some embodiments, the tumor cells have receptors ranging from 1,000 to 100,000, from 1,000 to 50,000, from 1,000 to 25,000 receptors, 1,000 to 10,000 receptors per cell. For example, in some embodiments the tumor cells have about 1,000, about 10,000, or less than 100,000 receptors per cell. Further therapeutic agents that can be combined with a compound of the disclosure are found in Goodman and Gilman’s "The Pharmacological Basis of Therapeutics" Tenth Edition edited by Hardman, Limbird and Gilman or the Physician’s Desk Reference, both of which are incorporated herein by reference in their entirety. The compounds of Structure (I), (III), or (IV) described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the one or more compounds of the disclosure will be co-administered with other agents as described above. When used in combination therapy, the compounds described herein are administered with the second agent simultaneously or separately. This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the disclosure and any of the agents described above can be simultaneously administered, wherein both the agents are present in separate formulations. In another alternative, a compound of the present disclosure can be administered just followed by and any of the agents described above, or vice versa. In some embodiments of the separate administration protocol, a compound of the disclosure and any of the agents described above are administered a few minutes apart, or a few hours apart, or a few days apart. In some embodiments, the method further comprises administering an additional therapeutic agent selected from the group consisting of an antineoplastic agent, an enediyne antitumor antibiotic, a maytansinoid, a topoisomerase inhibitor, a kinase inhibitor, an anthracycline, and EGFR inhibitor, an alkylating agent and combinations thereof. In some more specific embodiments, the method further comprises administering an additional therapeutic agent selected from the group consisting of an antineoplastic agent, an enediyne antitumor antibiotic, a maytansinoid, a topoisomerase inhibitor, a kinase inhibitor, an anthracycline, and EGFR inhibitor, an alkylating agent and combinations thereof. In certain embodiments, the additional therapeutic agent comprises auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan. In some embodiments, a pharmaceutical composition comprising the compound of any of the forementioned structures, and a pharmaceutically acceptable carrier, diluent, or excipient. In some embodiments, a method of treating a disease or disorder, comprising administering a therapeutically effective amount of a compound of any of the forementioned structures, or the pharmaceutical composition of the same, to a subject in need thereof. In certain embodiments, the disease or disorder is cancer. In some more specific embodiments, the cancer is breast cancer, stomach cancer, lung cancer, ovarian cancer, lymphoma, and bladder cancer. The examples and preparations provided below further illustrate and exemplify the compounds of the present disclosure and methods of preparing such compounds. It is to be understood that the scope of the present disclosure is not limited in any way by the scope of the following examples and preparations. In the following examples, and throughout the specification and claims, molecules and moieties with a single stereocenter, unless otherwise noted, exist as a racemic mixture. Those molecules and moieties with two or more stereocenters, unless otherwise noted, exist as a racemic mixture of diastereomers. Single enantiomers/diastereomers may be obtained by methods known to those skilled in the art. As described in detail above, compounds of structures (I), (IA), (IB), (IC), (ID), (IE), (IF), (IG), (IH), and (IJ), can be prepared by oligomerization using well known phosphoramidite chemistry. Applicants have discovered intermediate compounds useful for synthesis of compounds of structures (I), (IA), (IB), (IC), (ID), (IE), (IF), (IG), (IH), and (IJ). Accordingly, embodiments of the present disclosure provide a compound having one of the following structures (II): (II) or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein R1 and R2 are each independently H, OH, SH, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; R3 is, at each occurrence, independently H, alkyl or alkoxy; R4 is, at each occurrence, independently OH, SH, O-, S-, ORd or SRd; R5 is, at each occurrence, independently oxo or thioxo; R6 and R7 are, at each occurrence, independently H, OH, or halo; L2, L3, L5, and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; L4 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker; L7 is, at each occurrence, independently a linker; M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; G is, at each occurrence, independently a moiety reactive under cycloaddition; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of Structure (I); and m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, an integer of zero or greater; p is, at each occurrence, an integer of zero or greater; w is, at each occurrence, an integer of zero or greater; k is, at each occurrence, an integer of zero or greater; x9 is, at each occurrence, an integer from 1 to 6; and n is an integer of one or greater, wherein the sum of p and w is an integer of two or greater, and the compound comprises at least two different biologically active moieties. In some embodiments, at least one occurrence of L4 is heteroalkylene. In some embodiments, each occurrence of L4 is heteroalkylene. In some embodiments, at least one occurrence of L4 comprises alkylene oxide. In some more specific embodiments, the alkylene oxide of L4 is ethylene oxide. In some more specific embodiments, the ethylene oxide is polyethylene oxide. In certain embodiments, L4, at each occurrence, has the following structure: , wherein: z is an integer from 1 to 100; and * indicates a bond to the adjacent phosphorous atom. In some embodiments, z is an integer from 3 to 8. In some embodiments, z is 3. In some embodiments, z is 4. In some embodiments, z is 5. In some embodiments, z is 6. In some embodiments, z is 7. In some embodiments, z is 8. In some other embodiments, z is an integer from 22 to 26. In some embodiments, z is 22. In some embodiments, z is 23. In some embodiments, z is 24. In some embodiments, z is 25. In some embodiments, z is 26. In some other embodiments, z ranges from 19 to 28. In certain embodiments, the average z is 23. In some embodiments, the average z is 19, 20, 21, 22, 23, 24, 25, 26, 27, or 28. In other embodiments, at least one occurrence of L4 is an alkylene linker (e.g., methylene). In some more specific embodiments, L4 is an alkylene linker at each occurrence (e.g., methylene). In some embodiments, at least one occurrence of in compounds of structure (II) comprising one of the following structures: ; ; ; or wherein z is an integer from 1 to 100. In some embodiments, z is an integer from 3 to 8. In some other embodiments, z is an integer from 22 to 26. In some other embodiments, z ranges from 19 to 28. In some embodiments, at each occurrence, in compounds of structure (II) comprising one of the following structures: ; ; ; or wherein z is an integer from 1 to 100. In some embodiments, z is an integer from 3 to 8. In some other embodiments, z is an integer from 22 to 26. In some other embodiments, z ranges from 19 to 28. Some embodiments provide a compound having the following structure (IIA): (IIA) or a stereoisomer, salt or tautomer thereof. In some embodiments, L3, L5, and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers. In some embodiments, L3, L5 or L6 are, at each occurrence, independently alkylene. In more embodiments, L3, L5 or L6 are, at each occurrence, independently C1-C6 alkylene, C2-C6 alkenylene or C2-C6 alkynylene. In some more specific embodiments, the alkylene linker of L3 and L5 are C1 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C2 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C3 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C4 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C5 alkyl linker. In some more specific embodiments, the alkylene linker of L3 and L5 are C6 alkyl linker. In some specific embodiments, the alkylene linker of L6 are C1 alkyl linker. In some specific embodiments, the alkylene linker of L6 are C2 alkyl linker. In some specific embodiments, the alkylene linker of L6 are C3 alkyl linker. In other embodiments, at least one occurrence of L6 is a direct bond. In some other embodiments, each occurrence of L6 is a direct bond. Some embodiments provide a compound having the following structure (IIB): (IIB) or a stereoisomer, salt or tautomer thereof, wherein: y1 is, at each occurrence, independently an integer from 0 to 6; and y2 and y3 are, at each occurrence, independently an integer from 1 to 6. In some embodiments, y1 is, at each occurrence, an integer 0 (i.e., L6 is a direct bond). In some embodiments, y1 is, at each occurrence, an integer 1. In some embodiments, y1 is, at each occurrence, an integer 2. In some embodiments, y1 is, at each occurrence, an integer 3. In some embodiments, y1 is, at each occurrence, an integer 4. In some embodiments, y1 is, at each occurrence, an integer 5. In some embodiments, y1 is, at each occurrence, an integer 6. In some embodiments, y2 and y3 are at each occurrence, an integer 1 In some embodiments, y2 and y3 are at each occurrence, an integer 2. In some embodiments, y2 and y3 are at each occurrence, an integer 3. In some embodiments, y2 and y3 are at each occurrence, an integer 4. In some embodiments, y2 and y3 are at each occurrence, an integer 5. In some embodiments, y2 and y3 are at each occurrence, an integer 6. In some specific embodiments, y1 is, at each occurrence, an integer 0 or 1; and y2, and y3 are, at each occurrence, 1. As described in detail above, compounds of (III), can be prepared by oligomerization using well known phosphoramidite chemistry. Applicants have discovered intermediate compounds (V) useful for synthesis of compounds of structures (III). Accordingly, some embodiments provide intermediate compounds having structure (V): (V) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: G2 is, at each occurrence, independently a moiety reactive under cycloaddition; G3 is, at each occurrence, independently a moiety reactive under cycloaddition; L1a is, at each occurrence, independently a heteroalkylene or heteroarylene linker; L1b is, at each occurrence, independently either i) absent or ii) a linker; L7 is, at each occurrence, independently a linker; L2, L3, L5 and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; R21 is, at each occurrence, independently a natural or unnatural amino acid side chain; R1 and R2 are each independently H, OH, SH, –NH2, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of structure (V); m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, independently an integer of zero or greater; v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties. Compounds of structure (V) have a peptide-based backbone. The various linkers and substituents (e.g., G2, G3, Q, R1, R2, R21, L2, L3, L5, L6, L7 and L') in the compound of structure (V) are optionally substituted with one more substituent. For example, in some embodiments the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (V). In certain embodiments, each alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (V) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether, alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether. In certain embodiments the optional substituent is ˗OP(=Ra)(Rb)Rc, wherein Ra is O or S, Rb is OH, SH, O-, S-, ORd or SRd, Rc is OH, SH, O-, S-, ORd, SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, and Rd is a counter ion. In certain embodiments, substituents are selected to increase cellular or tissue permeation. In related embodiments, substituents are selected to increase cellular or tissue retention. In some embodiments, at least one R21 is a neutral amino acid side chain. In some embodiments, at least one R21 is a charged amino acid side chain. In some embodiments, R21 is, at each occurrence, independently H, alkyl, ˗CH2CO2-, ˗CH2CH2CO2-, ˗CH2CH2CH2CH2NH3+, ˗CH2CH2CH2NHC(=NH2+)NH2 or imidazolyl. In more specific embodiments, R21, L5 and m are selected such that has an amino acid sequence of (G)10, (GDGDGDGDGD) or (GKGKGKGKGK). In another embodiment, R21, L5 and m are selected such that has an amino acid sequence capable of forming an ^-helix or ^-sheet secondary structure. In some of those embodiments, the amino acid sequence is (GGEEFMLVYKFARKHGG) or (GGMSMVVSGG). In some embodiments, L5 or L3, or both, is present for at least one occurrence. In more specific embodiments, when present, L5 or L3, or both, is a heteroalkylene linker. In some of those embodiments, the heteroalkylene linker comprises a functional group capable of maintaining a positive or negative charge at pH values ranging from 3 to 11 in aqueous solution. In more specific embodiments, at least one occurrence of L5 or L3, or both, has the following structure: . In some embodiments, at least one occurrence of L5 or L3, or both, has the following structure: . In some embodiments, L6 and L2 are independently absent or a heteroalkylene linker. In more specific embodiments, the heteroalkylene linker is a peptidyl linker. As described in detail above, compounds of (IV), can be prepared by oligomerization using well known phosphoramidite chemistry. Applicants have discovered intermediate compounds (VI) useful for synthesis of compounds of structures (IV). Accordingly, some embodiments provide a compound having structure (VI): (VI) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: G2 is, at each occurrence, independently a moiety reactive under cycloaddition; G3 is, at each occurrence, independently a moiety reactive under cycloaddition; L7 is, at each occurrence, independently a linker; L2, L5 and L6 are, at each occurrence, independently a direct bond or independently an optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker; L8 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker comprising one or more charged moieties, provided at least one charged moiety is not a phosphate ester; R3 is, at each occurrence, independently H, alkyl or alkoxy; R1 and R2 are each independently -H, -OH, -SH, alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl, alkoxycarbonyl, ˗OP(=Ra)(Rb)Rc, Q or a protected form thereof, or L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a solid support or solid support residue, a linker comprising a covalent bond to a solid support or solid support residue or a linker comprising a covalent bond to a further compound of structure (VI); q is, at each occurrence, independently an integer of zero or greater; v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties. The various linkers and substituents (e.g., G2, G3, Q, R1, R2, R3, L2, L5, L6, L7, L8, and L') in the compound of structure (VI) are optionally substituted with one more substituent. For example, in some embodiments the optional substituent is selected to optimize the water solubility, permeability, retention, or other property of the compound of structure (VI). In certain embodiments, each alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker or alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl or alkoxycarbonyl in the compound of structure (VI) is optionally substituted with one more substituents selected from the group consisting of hydroxyl, alkoxy, alkylether, alkoxyalkylether, sulfhydryl, amino, alkylamino, carboxyl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether and thiophosphoalkylether. In certain embodiments the optional substituent is ˗OP(=Ra)(Rb)Rc, wherein Ra is O or S, Rb is OH, SH, O- , S-, ORd or SRd, Rc is OH, SH, O-, S-, ORd, SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, and Rd is a counter ion. In certain embodiments, substituents are selected to increase cellular or tissue permeation. In related embodiments, substituents are selected to increase cellular or tissue retention. In some embodiments, the alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl and alkyloxycarbonyl are optionally substituted with hydroxyl, amino, sulfhydryl, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether, or combinations thereof In some embodiments, the charged moieties are positively charged. In more specific embodiments, the charged moieties comprise a protonated amine or quaternary amine functional group. In more specific embodiments, the charged moieties independently have one of the following structures: ; or wherein: R is, at each occurrence, independently H or C1-C6 alkyl. In other embodiments, the charged moieties are negatively charged. In certain embodiments, the charged moieties comprise a carboxylic acid, phosphate or sulfate functional group. In some embodiments, the charged moieties have the following structure: . In some embodiments, the charged moieties comprise a combination of positively charged and negatively charged moieties. In certain embodiments, the charged moieties are pendant to the backbone of the compound (e.g., attached to a linker via an alkylene or heteroalkylene linker). In some embodiments, the charged moiety is part of the backbone of the compound (e.g., part of the contiguous chain of the linker). In some embodiments, L8 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or ’ wherein: R is, at each occurrence, independently H or C1-C6 alkyl; x is an integer from 0 to 6; and m is an integer of 1 or greater, provided that m is selected such that the compound comprises at least two charged moieties. The linker L7 can be used as a point of attachment of the M2 and M3 moieties or G2 and G3 moieties in case of structure (V) and (VI) to the remainder of the compound. For example, in some embodiments a synthetic precursor (II) to the compound of structure (I) is prepared, and the M2 and M3 moieties are attached to the synthetic precursor using any number of coupling methods known in the art. In more embodiments, at least one occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof. In other embodiments, the functional group comprises an amide or an ester. In more specific embodiments, at least one occurrence of L7 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or . In more embodiments, each occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof. In other embodiments, the functional group comprises an amide. In more specific embodiments, each occurrence of L7 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or . In some embodiments, at least one occurrence of R3 is H. In some embodiments, at least one occurrence of R3 is alkyl. In some embodiments, at least one occurrence of R3 is alkoxy. In certain embodiments, each occurrence of R3 is H. In some embodiments, at least one occurrence of R5 is oxo (=O). In some embodiments, at least one occurrence of R5 is thioxo (=S). In certain embodiments, each occurrence of R5 is oxo (=O). In some embodiments, at least one occurrence of R4 is OH. In some embodiments, at least one occurrence of R4 is SH. In some embodiments, at least one occurrence of R4 is O-. In some embodiments, at least one occurrence of R4 is S-. In certain embodiments, each occurrence of R4 is O-. In other various embodiments, R1 and R2 are each independently OH or ˗OP(=Ra)(Rb)Rc. In some different embodiments, R1 or R2 is OH or ˗OP(=Ra)(Rb)Rc, and the other of R1 or R2 is Q or a linker comprising a covalent bond to Q. In still more different embodiments of any of the foregoing compounds of structure (II), (V), or (VI), R1 and R2 are each independently ˗OP(=Ra)(Rb)Rc. In some of these embodiments, Rc is OL'. In other embodiments, R1 and R2 are each independently ˗OP(=Ra)(Rb)OL', and L' is an alkylene or heteroalkylene linker to: Q, a targeting moiety, an analyte (e.g., analyte molecule), a solid support, a solid support residue, a nucleoside or a further compound of structure (II), (V), or (VI). The linker L' can be any linker suitable for attaching Q, a targeting moiety, an analyte (e.g., analyte molecule), a solid support, a solid support residue, a nucleoside or a further compound of structure (II), (V), or (VI) to the compound of structure (II), (V), or (VI). Advantageously certain embodiments include use of L' moieties selected to increase or optimize water solubility of the compound. In certain embodiments, L' is a heteroalkylene moiety. In some other certain embodiments, L' comprises an alkylene oxide or phosphodiester moiety, or combinations thereof. In In some other embodiments, R2 is L'. For example, in some embodiments, L' is a linker to a targeting moiety. In some specific embodiments, L' is a linker to a targeting moiety, the linker comprising an alkylene oxide or phosphodiester moiety, or combinations thereof. In other embodiments, L' has one of the following structures: ; ; ; or wherein: x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer from 1 to 10; Rb is H, an electron pair or a counter ion; L'' is the targeting moiety or a linkage to the targeting moiety. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 1. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 2. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 3. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 4. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 5. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 6. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 7. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 8. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 9. In some embodiments, x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer 10. In some embodiments, Rb is H. In some embodiments, Rb is a counter ion. For example, in some embodiments, Rb is Na+. In some embodiments, Rb is K+. In some embodiments, L'' is the targeting moiety. In some other embodiments, L'' is a linkage to the targeting moiety. For example, the targeting moiety is an antibody. In another example, the targeting moiety is cell surface receptor antagonist. In some more specific embodiments, the antibody or cell surface receptor antagonist is an epidermal growth factor receptor (EGFR) inhibitor, a hepatocyte growth factor receptor (HGFR) inhibitor, an insulin-like growth factor receptor (IGFR) inhibitor, a folate, or a MET inhibitor. In other embodiments, the targeting moiety is a monoclonal antibody. For instance, in some embodiments, the monoclonal antibody is Abciximab, Adalimumab, Alemtuzumab, Alirocumab, Avibactam, Basiliximab, Benralizumab, Bezlotoxumab, Blinatumomab, Brodalumab, Burosumab, Canakinumab, Caplacizumab, Certolizumab pegol, Daclizumab, Denosumab, Dupilumab, Eculizumab, Emicizumab, Erenumab, Evolocumab, Fremanezumab, Galcanezumab, Golimumab, Guselkumab, Ibalizumab, Idarucizumab, Infliximab, Itolizumab, Ixekizumab, Lanadelumab, Lokivetmab, Mepolizumab, Natalizumab, Obiltoxaximab, Ocrelizumab, Omalizumab, Palivizumab, Ranibizumab, Raxibacumab, Reslizumab, Rmab, Rovelizumab, Ruplizumab, Sarilumab, Secukinumab, Tildrakizumab, Thiomab, Tocilizumab, Ustekinumab, Vedolizumab, Abrilumab, Actoxumab, Aducanumab, Afasevikumab, Afelimomab, Anifrolumab, Anrukinzumab (IMA-638), Aselizumab, Atorolimumab, Bapineuzumab, BCD-100, Bertilimumab, Besilesomab, Biciromab, Bimagrumab, Bimekizumab, Birtamimab, Bleselumab, Blosozumab, Bococizumab, Brazikumab, Briakinumab, Brolucizumab, Carlumab, Carotuximab, Cedelizumab, Clazakizumab, Clenoliximab, Concizumab, Cosfroviximab, CR6261, Crenezumab, Crizanlizumab, Crotedumab, Depatuxizumab, mafodotin, Derlotuximab biotin, Dezamizumab, Diridavumab, Domagrozumab, Dusigitumab, Ecromeximab, Edobacomab, Efalizumab, Efungumab, Eldelumab, Elezanumab, Enokizumab, Eptinezumab, Erlizumab, Etrolizumab, Evinacumab, Exbivirumab, Fanolesomab, Faralimomab, Faricimab, Fasinumab, Felvizumab, Fezakinumab, Flanvotumab, Fletikumab, Flotetuzumab, Fontolizumab, Foravirumab, Frovocimab, Fulranumab, Gantenerumab, Gavilimomab, Gevokizumab, Gimsilumab, Gomiliximab, Gosuranemab, Ianalumab, Inclacumab, Inolimomab, Iomab-B, Keliximab, Lampalizumab, Landogrozumab, Larcaviximab, Lebrikizumab, Lenvervimab, Lerdelimumab, Letolizumab, Libivirumab, Ligelizumab, Lodelcizumab, Lulizumab pegol, Marstacimab, Mavrilimumab, Metelimumab, Mirikizumab, Motavizumab, Muromonab CD3, Nebacumab, Nemolizumab, NEOD001, Nirsevimab, Odulimomab, Olendalizumab, Olokizumab, OMS721, Opicinumab, Orticumab, Otelixizumab, Otilimab, Oxelumab, Ozanezumab, Ozoralizumab, Pagibaximab, Panobacumab, Pascolizumab, Pateclizumab, PDR001, Perakizumab, Pexelizumab, Placulumab, Plozalizumab, Ponezumab, Porgaviximab, Prasinezumab, Priliximab, PRO 140, Quilizumab, Rafivirumab, Ralpancizumab, Ranevetmab, Ravagalimab, Ravulizumab, Refanezumab, Regavirumab, Relatlimab, Rinucumab, Risankizumab, Roledumab, Romosozumab, Rontalizumab, SA237, Satralizumab, Sevirumab, SHP647, Sifalimumab, Simtuzumab, Siplizumab, Sirukumab, Solanezumab, Sonepcizumab, Spartalizumab, Stamulumab, Sulesomab, Suptavumab, Sutimlimab, Suvizumab, Suvratoxumab, Tadocizumab, Talizumab, Tamtuvetmab, Tanezumab, Tefibazumab, Telimomab aritox, Teneliximab, Teplizumab, Teprotumumab, Tezepelumab, Tibulizumab, Toralizumab, Tralokinumab, Trevogrumab, Tuvirumab, Ulocuplumab, Urtoxazumab, Varisacumab, Vepalimomab, Vesencumab, Visilizumab, Vobarilizumab, Zolimomab aritox, trastuzumab, gemtuzumab, brentuximab, vorsetuzumab, lorvotuzumab, cantuzumab, bivatuzumabor inotuzumab, or vadastuximab. In other more specific embodiments of any of the foregoing compounds of structure (II), (V), or (VI), R1 or R2 has one of the following structures: ; ; ; ; ; ; ; ; ; ; or , wherein Ra is H or a solid support. In some specific embodiments, R2 has one of the following structures: ; or . . Certain embodiments of compounds of structure (II), (V), or (VI) can be prepared according to solid-phase synthetic methods analogous to those known in the art for preparation of oligonucleotides. Accordingly, in some embodiments, L' is a linkage to a solid support, a solid support residue or a nucleoside. Solid supports comprising an activated deoxythymidine (dT) group are readily available, and in some embodiments can be employed as starting material for preparation of compounds of structure (II), (V), or (VI). Accordingly, in some specific embodiments, R1 has the following structure: . One of skill in the art will understand that the dT group depicted above is included for ease of synthesis and economic efficiencies only, and is not required. Other solid supports can be used and would result in a different nucleoside or solid support residue being present on L', or the nucleoside or solid support residue can be removed or modified post synthesis. In some embodiments, the analyte molecule is a nucleic acid, amino acid or a polymer thereof. In some embodiments, the analyte molecule is an enzyme, receptor, receptor ligand, antibody, glycoprotein, aptamer or prion. In some embodiments, the targeting moiety is an antibody or cell surface receptor antagonist. In further some embodiments, the solid support is a polymeric bead or non-polymeric bead. In some embodiments, M2 is, at one or more occurrences, independently a moiety comprising four or more aryl or heteroaryl rings, or combinations thereof. In some embodiments, M2 is, at one or more occurrences, independently fluorescent or colored. In certain embodiments, M2 is, at one or more occurrences, fluorescent. In other embodiments, M2 is, at one or more occurrences, independently comprise a fused-multicyclic aryl or heteroaryl moiety comprising at least four fused rings. In some embodiments, M2 is, at each occurrence, independently selected from the group consisting of phenyl, (bis-fluorophenyl-difluorobora- diaza-indacene)phenyl, quaterphenyl, bi-benzothiazole, ter-benzothiazole, bi-naphthyl, bi- anthracyl, squaraine, squarylium, 9,10-ethynylanthracene, and ter-naphthyl moiety. In some other embodiments, M2 is, at each occurrence, independently selected from the group consisting of p-terphenyl, perylene, azobenzene, phenazine, phenanthroline, acridine, thioxanthrene, chrysene, rubrene, coronene, cyanine, perylene imide, perylene amide, and derivatives thereof. In some other embodiments, M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, resorufin dye, dipyrrometheneboron difluoride dye, ruthenium bipyridyl dye, thiazole orange dye, polymethine, and N-aryl-1,8-naphthalimide dye. Further in some other embodiments, M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, boron-dipyrromethene, rhodamine, cyanine, pyrene, perylene, perylene monoimide, 6-carboxyfluorescein (6-FAM), 5-carboxyfluorescein (5-FAM), 6- fluorescein isothiocyanate (6-FITC), 5-fluorescein isothiocyanate (5-FITC), and derivatives thereof. In certain embodiments, M2, at each occurrence, independently has one of the following structures: ; ; ; ; ; ; or . In some specific embodiments, at least one occurrence of M2 has the following structure: . In some more specific embodiments, each occurrence of M2 has the following structure: . In some embodiments, at least one occurrence of –L7- M2 has one of the following structures: . In some more specific embodiments, each occurrence of –L7- M2 has one of the following structures: . M3 is selected based on the desired alkylating properties. Exemplary M3 moiety can be appropriately selected by one of ordinary skill in the art based on the desired end use. In some embodiments, at least one occurrence of M3 moiety is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic. In more specific embodiments, each occurrence of M3 is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic. In certain embodiments, at least one occurrence of M1 or M3 is a nitrogen mustard, a nitrosourea, a tetrazine, an aziridine, a cisplatin or cisplatin derivative, or a non-classical alkylating agent. In more specific embodiments, at least one occurrence of M3 is mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide, busulfan, N-nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU), semustine (MeCCNU), fotemustine, streptozotocin, dacarbazine, mitozolomide, temozolomide, thiotepa, mytomycin, diaziquone (AZQ), cisplatin, carboplatin, oxaliplatin, procarbazine, or hexamethylmelamine. In some embodiments, at least one occurrence of M3 is an anti-folate, a fluoropyrimidines, a deoxynucleoside analogue, or a thiopurine. In certain embodiments, at least one occurrence of M3 is methotrexate, pemetrexed, fluorouracil, capecitabine, cytarabine, gemcitabine, decitabine, azacitidine, fludarabine, nelarabine, cladribine, clofarabine, pentostatin, thioguanine, and mercaptopurine. In some specific embodiments, at least one occurrence of M3 is an auristatin, a Vinca alkaloid, or a taxane. In certain specific embodiments, at least one occurrence of M3 is auristatin F, auristatin E, vincristine, vinblastine, vinorelbine, vindesine, vinflunine, paclitaxel, docetaxel, etoposide, or teniposide. In some more specific embodiments, at least one occurrence of M3 is irinotecan, SN 38, topotecan, camptothecin, doxorubicin, mitoxantrone, teniposide. novobiocin, merbarone, or aclarubicin. In certain more specific embodiments, at least one occurrence of M3 is an anthracycline or a bleomycin. In some embodiments, at least one occurrence of M3 is doxorubicin, daunorubicin, epirubicin, idarubicin, pirarubicin, aclarubicin, or mitoxantrone. In some embodiments, at least one occurrence of M3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan. In some embodiments, each occurrence of M3 is auristatin F, monomethyl auristatin F, monomethyl auristatin E, paciltaxol, SN-38, calicheamicin, anthramycin, abbeymycin, chicamycin, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin prothracarcin, sibanomicin, sibiromycin, tomamycin, mertansine, emtansine, irinotecan, camptothecin, topotecan, silatecan, cositecan, Exatecan, Lurtotecan, gimatecan, Belotecan, and Rubitecan. M3 may be attached to the remainder of the molecule from any position (i.e., atom) on M3. One of skill in the art will recognize means for attaching M3 to the remainder of molecule. For example, M3 may be attached to the remainder of the molecule through nitrogen of diazepine, oxygen of diazepine or phenyl ring, or carbon of pyrrolidine ring. In some embodiments, at least one occurrence of M3 is an anti-inflammatory compound. M3 is a steroid which is a biologically active organic compound with four rings arranged in specific molecular configuration as shown below. For example, in some embodiments, at least one occurrence of M3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone. In some more specific embodiments, at least one occurrence of M3 has one of the following structures: or , wherein: R11 is H or halogen; R12 and R13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R14 is H, halogen, OH, or alkyl; R15 is H or OH; R16, R17, and R18, are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond. In some embodiments, at least one occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or . In some embodiments, each occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or . In some embodiments, G, G2, or G3 is a reactive moiety that undergoes the cycloaddition. In some specific embodiments, the cycloaddition is click reaction between an azide and an alkyne. In some embodiments, G, G2, or G3 has one of the following structures: ; or . In certain embodiments, G, G2, or G3, at each occurrence, has the structure of . In some other certain embodiments, G, G2, or G3, at each occurrence, has the structure of . The fluorescence intensity or the effectiveness toward alkylation of DNA can also be tuned by selection of different values of n. In some embodiments, n is an integer of one or greater. In certain embodiments, n is an integer from 1 to 100. In other embodiments, n is an integer from 1 to 10. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, n is 5. In some embodiments, n is 6. In some embodiments, n is 7. In some embodiments, n is 8. In some embodiments, n is 9. In some embodiments, n is 10. The fluorescence or the effectiveness toward alkylation of DNA may also be tuned by selection of values for m. The value for m has the ability to control the spacing between neighboring M1, M2, or M3 (or G, G2, or G3). In some embodiments, m is an integer of zero or greater. In certain embodiments, m is an integer from 0 to 100. In certain embodiments, m is an integer from 0 to 10. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments, m is 5. In some embodiments, m is 6. In some embodiments, m is 7. In some embodiments, m is 8. In some embodiments, m is 9. In some embodiments, m is 10. The fluorescence intensity may be tuned how many fluorescent dye moieties are attached to the polymeric backbone. The value for q has the ability to control the brightness of compounds. In some embodiments, q is, at each occurrence, an integer of zero or greater. In some more specific embodiments, q is from 0 to 10. In some embodiments, q is from 0 to 5. For example, in some embodiments, q is 0. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, q is 3. In some embodiments, q is 4. In some embodiments, q is 5. In some other embodiments, q is 0, 1, 2, 3, or 4. In some other embodiments, q is 1 or 2. The effectiveness toward alkylation of DNA may also be tuned by selection of values for p. In some embodiments, p is, at each occurrence, an integer of zero or greater. In some more specific embodiments, p is from 0 to 10. In some embodiments, p is from 0 to 5. For example, in some embodiments, p is 0. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, p is 4. In some embodiments, p is 5. In some other embodiments, p is 1, 2, 3, or 4. In some other embodiments, p is 2 or 3. The effectiveness toward alkylation of DNA may also be tuned by selection of values for w. In some embodiments, w is, at each occurrence, an integer of zero or greater. In some more specific embodiments, w is from 0 to 10. In some embodiments, w is from 0 to 5. In some embodiments, w is from 1 to 5. For example, in some embodiments, w is 1. In some embodiments, w is 2. In some embodiments, w is 3. In some embodiments, w is 4. In some embodiments, w is 5. In some other embodiments, w is 1, 2, 3, or 4. In some other embodiments, w is 1 or 2. The effectiveness toward alkylation of DNA may also be tuned by selection of values for k. In some embodiments, k is, at each occurrence, an integer of zero or greater. In some more specific embodiments, k is from 0 to 10. In some embodiments, k is from 0 to 5. For example, in some embodiments, k is 0. In some embodiments, k is 1. In some embodiments, k is 2. In some embodiments, k is 3. In some embodiments, k is 4. In some embodiments, k is 5. In other embodiments, k is 0, 1, 2, 3, or 4. In some other embodiments, k is 0 or 1. In some embodiments of structure (V) or (VI), v is an integer of one or greater. For example, in some embodiments, v is an integer from 1 to 100. Further in some embodiments, v is an integer from 1 to 10. In some embodiments, v is 1. In some embodiments, v is 2. In some embodiments, v is 3. In some embodiments, v is 4. In some embodiments, v is 5. In some embodiments, v is 6. In some embodiments, v is 7. In some embodiments, v is 8. In some embodiments, v is 9. In some embodiments, v is 10. The value n, q, w, p, k, and m are closely related to provide the ability to control the fluorescence and effectiveness toward alkylation of DNA. In some specific embodiments, n is 1; q is 2; w is 2, k is 0, p is 2; and m is 1. In some other specific embodiments, n is 1; q is 2; w is 1, k is 0, p is 3; and m is 1. In some other specific embodiments, n is 3; q is 1 for the first occurrence, q is 0 for the second occurrence, q is 0 for the third occurrence; w is 0 for the first occurrence, w is 0 for the second occurrence, and w is 1 for the third occurrence; k is 0 for the first occurrence, k is 1 for the second occurrence, and k is 0 for the third occurrence; p is 1 for the first occurrence, p is 0 for the second occurrence, and p is 0 for the third occurrence; and m is 1 for all occurrences. In some embodiments, the sum of p and w is an integer of two or greater, and the compound comprises at least two different biologically active moieties. In some embodiments, at least one occurrence of R6 or R7 is F or H. In other embodiments, at least each occurrence of R6 and R7 are F or H. In certain embodiments, at least each occurrence of R6 and R7 are F, R8 is O, R9 is H, and R10 is H. In some other embodiments, at least each occurrence of R6 and R7 are H, R8 is O, R9 is H, and R10 is H. In some embodiments, x9 is, at each occurrence, an integer from 1 to 6. For example, in some embodiments, each occurrence of x9 is 1. In some embodiments, each occurrence of x9 is 2. In some embodiments, each occurrence of x9 is 3. In some embodiments, each occurrence of x9 is 4. In some embodiments, each occurrence of x9 is 5. In some embodiments, each occurrence of x9 is 6. In some other embodiments, each occurrence of x9 is 2,4, or 6. In some other embodiments, each occurrence of x9 is 2 or 4. In some specific embodiments, the compound is a compound selected from Table 3A. The compounds in Table 3A are prepared according to the procedures set forth in the Examples.
Figure imgf000173_0001
.o 1 N -I I
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
wherein M3 has the following structure: . Further in some embodiments, at least one occurrence of M3 is an anti-inflammatory compound. M3 is a steroid which is a biologically active organic compound with four rings arranged in specific molecular configuration as shown below. For example, in some embodiments, at least one occurrence of M3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone. In some more specific embodiments, at least one occurrence of M3 has one of the following structures: or , wherein: R11 is H or halogen; R12 and R13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R14 is H, halogen, OH, or alkyl; R15 is H or OH; R16, R17, and R18, are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond. In some embodiments, at least one occurrence of M3 has one of the following structures:
; ; ; ; ; ; ; ; ; ; ; or . In some specific embodiments, the compound is a compound selected from Table 3B. The compounds in Table 3B are prepared according to the procedures set forth in the Examples
Figure imgf000185_0001
.o 1 N 1-I I
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
wherein M3 has one of the following structures: ; ; ; ; or , wherein or is selected from the following structures: ; ; ; ; ; ; ; ; ; ; ; or . In some embodiments, at least one occurrence of M3 is a Vitamin. For example, in some embodiments, at least one occurrence of M3 is a Vitamin E. In some more specific embodiments, at least one occurrence of M3 is a tocopherol. In certain embodiments, at least one occurrence of M3 is α-tocopherol, β-tocopherol, γ-tocopherol, δ-tocopherol, α -tocotrienol, β - tocotrienol, γ -tocotrienol, or δ -tocotrienol. In some embodiments, at least one occurrence of M3 has the following structure: , wherein R19 and R20 are independently H or CH3; and represents a single or double carbon-carbon bond. In some specific embodiments, at least one occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; or . In some specific embodiments, the compound is a compound selected from Table 3C. The compounds in Table 3C are prepared according to the procedures set forth in the Examples.
Figure imgf000197_0001
.o 5 N 1-I I
Figure imgf000198_0001
Figure imgf000199_0001
wherein M3 has one of the following structures: ; ; ; ; or , wherein is selected from the following structures: ; ; ; ; ; ; ; or . In some specific embodiments, the compounds of structure (V) are selected from Table 3D. The compounds in Table 3D are prepared according to the procedures set forth in the Examples.
Figure imgf000202_0001
.o 1 N - 2- V V
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
In some specific embodiments, the compounds of structure (VI) are selected from Table 3E. The compounds in Table 3E are prepared according to the procedures set forth in the Examples.
Figure imgf000207_0001
.o 1 N -I V
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
A REPRESENTATIVE DNA SYNTHESIS CYCLE Oligomerization is initiated, typically, through the removal of a protecting group (e.g., a dimethoxytrityl group, DMTr) to reveal a free –OH (hydroxyl) group (Step 1, DETRITYLATION). In a subsequent coupling step, a phosphoramidite monomer is introduced that reacts with the free OH group making a new covalent bond to phosphorus, with concomitant loss of the diisopropyl amine group (Step 2, COUPLING). The resultant, phosphite triester is oxidized (e.g., with I2 and pyridine) to the more stable phosphate ester (Step 3, OXIDATION) and a capping step renders unreactive any remaining free OH groups (Step 4, CAPPING). The new product, phosphate oligomer, contains a DMTr protected OH group that can be deprotected to reinitiate the synthetic cycle so another phosphoramidite monomer can be appended to the oligomer. Customization occurs at step 2 through the choice of phosphoramidite monomer. The nature of L (i.e., a linker group) and M (i.e., a chemotherapeutic agent) in the scheme above are selected such that a desired compound of Structure (I), (III), or (IV) is synthesized. M can be optionally absent to incorporate desired spacing between M moieties. A person of ordinary skill in the art can select multiple monomer types to arrive at compounds of the disclosure containing multiple therapeutic agents and/or other moieties (e.g., fluorophores or chromophores) with concurrent variability in linker groups. Several steroid phosphoramidites and vitamin phosphoramidites are commercially available and can be used in the above DNA synthesis cycle. This includes cholesterol phosphoramidite and tocopherol/tocotrienol phosphoramidite as shown below: and . GENERAL REACTION SCHEME 1 (PHOSPHORAMIDITE) Reaction Scheme I illustrates a method for preparation of phosphoramidite intermediates useful for preparation of compounds of Structure (I). Referring to Reaction Scheme I, G1 represents a desired moiety containing a carboxylic acid functional group (e.g., a drug moiety such as Auristatin F or SN 38), L represents a bivalent linker moiety (e.g., an alkylene, or alkylene ether), X represents a leaving group (e.g., halo such as Cl), and PG represents a protecting group (e.g., 4,4'-dimethoxytriphenylmethyl). Step 1 of Reaction Scheme I starts with an activation of the carboxylic acid functional group of the first compound shown using known reagents under basic conditions (e.g., HATU and DIPEA in DMF). The activated acid is then reacted with an amine to provide the reaction product of Step 1. The resulting diol is then protected under standard conditions (e.g., 4,4'-dimethoxytriphenylmethyl chloride and pyridine). The protected product is then reacted with 3- ((chloro(diisopropylamino)phosphaneyl)oxy)propanenitrile (or other appropriate reagent) to yield a desired compound of Structure (VII) as shown above. The resultant compound of Structure (VII) can then be used to synthesize a desired compound of Structure (I) by reaction under well-known (automated) DNA synthesis conditions. In addition to compounds of Structure (VII), additional repeat units may be incorporated to achieve a final compound of Structure (I). Generally, compounds having the following structure may be used: wherein: L is a desired linker moiety (e.g., including PEG or dye-containing moiety). In some specific embodiments, the following compound may be used in the synthesis of a compound of Structure (I): . EXAMPLES General Methods Mass spectral analysis is performed on a Waters/Micromass Quattro micro MS/MS system (in MS only mode) using MassLynx 4.1 acquisition software. Mobile phase used for LC/MS on dyes is 100 mM 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP), 8.6 mM triethylamine (TEA), pH 8. Phosphoramidites and precursor molecules are also analyzed using a Waters Acquity UHPLC system with a 2.1 mm × 50 mm Acquity BEH-C18 column held at 45 °C, employing an acetonitrile / water mobile phase gradient. Molecular weights for monomer intermediates are obtained using tropylium cation infusion enhanced ionization on a Waters/Micromass Quattro micro MS/MS system (in MS only mode). Excitation and emission profiles experiments are recorded on a Cary Eclipse spectra photometer. All reactions are carried out in oven dried glassware under a nitrogen atmosphere unless otherwise stated. Commercially available DNA synthesis reagents are purchased from Glen Research (Sterling, VA). Anhydrous pyridine, toluene, dichloromethane, diisopropylethyl amine, triethylamine, acetic acid, pyridine, and THF are purchased from Aldrich. All other chemicals are purchase from Aldrich or TCI and are used as is with no additional purification.
EXAMPLE 1 SYNTHESIS OF DMT PROTECTED GEMCITABINE COMPOUND Gencitabine (0.5057 mmole) is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of pyridine, anhydrous (5.06 mL). The reaction flask is then transferred to an ice water bath (0°C) and allowed to cool with mixing until thermally equalized (approximately 10 minutes). Then, 4,4′-Dimethoxytrityl chloride (0.257 g, 0.759 mmole) is added to the cooled mixture with continuous mixing under inert gas. The reaction mixture is allowed to warm to room temperature then sampled for TLC analysis. When reaction completion verified, the remaining unreacted 4,4′-Dimethoxytrityl chloride is quenched by addition of methanol to the reaction mixture (0.160 g, 5.06 mmole). The solvent is stripped off by rotary evaporation, under vacuum (10 mbar), with heating (55°C). The concentrated residue is then suspended in toluene (5.06 mL) and toluene stripped off by rotary evaporation, under vacuum (10 mbar), with heating (55°C); repeated two time. The crude produce is dissolved in dichloromethane (5.06 mL) and washed with sodium bicarbonate (5.06 mL, saturate aq.) and separated. This process is repeated one time. The separated organic phase is washed with sodium chloride (5.06 mL, saturated aq.) and separated. The separated organic phase is dried over sodium sulfate, anhydrous and the sodium sulfate filtered off. The product containing organic phase is sampled for TLC and LC-UV/MS analysis. Solvent is removed by rotary evaporation, resulting in crude DMT protected gemcitabine. This crude material is then combined with crude material from a small-scale test reaction. The combined crude material is purified by silica gel flash chromatography, dichloromethane / methanol mobile phase, product containing fractions are pooled and solvent is removed by rotary evaporation, and then placed on vacuum line for at least 24 hours to yield DMT protected gemcitabine. EXAMPLE 2 SYNTHESIS OF DMT PROTECTED GEMCITABINE PHOSPHORAMIDITE The purified DMT protected gemcitabine (0.226 mmole), dried under vacuum for at least 24 hours is dissolved in dichloromethane (2.26 mL), under inert gas blanket, with magnetic stir bar, followed by addition of DIPEA (0.117 g), and then addition of Cl-Phos. (0.107 g). The reaction is allowed to mix for approximately 15 minutes and then sampled for TLC analysis (TLC showed reaction completion). When reaction completion is verified, the reaction mixture is washed by adding directly to sodium bicarbonate (2.26 mL, saturated aq.) and organic phase separated, repeated one time. The organic phases are combined and dried over sodium sulfate, anhydrous, and then the sodium sulfate filtered off. The product containing organic phase is sampled for TLC and LC-UV/MS analysis. Then, dichloromethane is stripped off by rotary evaporation and proceeded to purification without crude weight. This crude material is then combined with crude material from a small-scale test reaction. The combined crude material is purified by silica gel solid phase extraction, dichloromethane/ methanol / triethylamine mobile phase, product containing fractions are pooled. The mobile phase is stripped off by rotary evaporation, and then placed on vacuum line for at least 24 hours to yield DMT protected gemcitabine phosphoramidite. Alternatively, the DMT protected gemcitabine phosphoramidite can be purchased from Glen Research and used as is with no additional purification. EXAMPLE 3 SYNTHESIS OF AURISTATIN F PHOSPHORAMIDITE Auristatin F (0.501 g, 0.671 mmole) was added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF (6.10 mL). The Auristatin F was allowed to dissolve completely under inert gas at room temperature. Then, to the mixture was added DIPEA (0.351 g, 2.013 mmole), followed by addition of HATU (0.278 g, 0.732 mmole).6,7-Dihydroxy-4-oxaheptylamine (0.091 g, 0.610 mmole) was added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF (6.10 mL), and dissolve completely at room temperature. Then the Auristatin F reaction mixture was added to the solution containing 6,7-dihydroxy-4-oxaheptylamine; the resultant mixture was allowed to mix at under inert gas, at room temperature, until reaction completion verified by TLC and LC-UV/MS analysis (analytical LC-UV248nm chromatogram showed 12% target product by total peak area or approx.60% related peak area, identified by MS). At reaction completion the solvents were stripped off by rotary evaporation, under vacuum (10 mbar), with heating (55 °C). The concentrated residue was placed under full vacuum, at room temperature, for several hours resulting in 1.09g of crude Compound by weight (theoretical, 0.535 g, 0.610 mmole). The crude compound (a portion of crude material from previous step, 0.444 g, 0.5057 mmole theoretical) was added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of pyridine, anhydrous (5.06 mL). The reaction flask was then transferred to an ice water bath (0°C) and allowed to cool with mixing until thermally equalized (approximately 10 minutes). Then, 4,4′-Dimethoxytrityl chloride (0.257 g, 0.759 mmole) was added to the cooled mixture with continuous mixing under inert gas. The reaction mixture was allowed to warm to room temperature then sampled for TLC analysis. When reaction completion verified, the remaining unreacted 4,4′-Dimethoxytrityl chloride was quenched by addition of methanol to the reaction mixture (0.160 g, 5.06 mmole). The solvent was stripped off by rotary evaporation, under vacuum (10 mbar), with heating (55°C). The concentrated residue was then suspended in toluene (5.06 mL) and toluene stripped off by rotary evaporation, under vacuum (10 mbar), with heating (55°C); repeated two time. The crude produce was dissolved in dichloromethane (5.06 mL) and washed with sodium bicarbonate (5.06 mL, saturate aq.) and separated. This process was repeated one time. The separated organic phase was washed with sodium chloride (5.06 mL, saturated aq.) and separated. The separated organic phase was dried over sodium sulfate, anhydrous and the sodium sulfate filtered off. The product containing organic phase was sampled for TLC and LC-UV/MS analysis (analytical LC- UV248nm chromatogram showed ~24% target product by total peak area, identified by MS). Solvent was removed by rotary evaporation, resulting in 0.769 g of crude DMT protected compound. This crude material was then combined with crude material from a small-scale test reaction (0.135 g), for a combined crude yield of 0.931 g. The combined crude material was purified by silica gel flash chromatography, dichloromethane / methanol mobile phase, product containing fractions were pooled and solvent was removed by rotary evaporation, and then placed on vacuum line for at least 24 hours to yield 0.399 g (analytical LC-UV248nm chromatogram showed 82% target product by total peak area, identified by MS).
The purified DMT protected compound (portion of the material 0.267, 0.226 mmole), dried under vacuum for at least 24 hours was dissolved in dichloromethane (2.26 mL), under inert gas blanket, with magnetic stir bar, followed by addition of DIPEA (0.117 g), and then addition of Cl-Phos. (0.107 g). The reaction was allowed to mix for approximately 15 minutes and then sampled for TLC analysis (TLC showed reaction completion). When reaction completion was verified, the reaction mixture was washed by adding directly to sodium bicarbonate (2.26 mL, saturated aq.) and organic phase separated, repeated one time. The organic phases were combined and dried over sodium sulfate, anhydrous, and then the sodium sulfate filtered off. The product containing organic phase was sampled for TLC and LC-UV/MS analysis (analytical LC-UV248nm chromatogram showed two product peaks (diastereomers) ~64% by total peak area, identified by MS). Then, dichloromethane stripped off by rotary evaporation and proceeded to purification without crude weight. This crude material was then combined with crude material from a small-scale test reaction. The combined crude material was purified by silica gel solid phase extraction, dichloromethane/ methanol / triethylamine mobile phase, product containing fractions were pooled (determined by TLC) sampled for TLC and LC- UV/MS analysis. The mobile phase striped off by rotary evaporation, and then placed on vacuum line for at least 24 hours to yield 0.363 g of Auristatin F phosphoramidite. (Analytical LC-UV248nm chromatogram showed 68% target product by total peak area, identified by MS). Similarly, SN38 phosphoramidites, ibuprofen phosphoramidite, and naproxen phosphoramidite are synthesized according to the above procedure. The SN38 phosphoramidites, ibuprofen phosphoramidite, and naproxen phosphoramidite include the following:
Figure imgf000226_0001
EXAMPLE 4
SYNTHESIS OF ALANINE LINKED SN38
Step 5 Piperidine
Dimethylformamide
Step 1 Into a 500 mL round bottomed flask was placed Boc-Ala-Ala-OH (4.0g, 15.4mmol, Chem-Impex Cat#04505), DMF (150mL) and a magnetic stir bar. HATU coupling agent (7.0g, 18.4mmol) was added and the mixture was stirred for 5min before adding Fmoc-1,2- diaminoethane-HCl (4.9g, 15.4mmol) and diisopropylethylamine (8.0mL, 46.1mmol). After stirring overnight, TLC (silica plats with F254 and dichloromethane:methanol elution 9:1) indicated the reaction was complete. The reaction mixture as concentrated on the rotovap and then partitioned between dichloromethane and water. The solvent layers were separated with an extraction funnel and the aqueous layer extracted three additional times with dichloromethane. The organic layers were combined, dried over sodium sulfate and concentrated under reduced pressure. The product was used in the next step without additional purification. Step 2 In a 250 mL round bottomed flask with magnetic stir bar was placed the Boc-Ala-Ala derivative prepared in the previous step (2.5g, 4.8mmol). Dichloromethane (30mL) and DMF (10mL) were added and the mixture stirred. To this was added 4M hydrochloric acid in dioxane (30mL, Sigma Cat#345547) and the mixture was stirred. After 1h, TLC (silica plates with F254, elution with 9:1 dichloromethane:methanol) indicated the reaction was complete. The solvents were removed on the rotovap. Acetonitrile (50mL) was added and the mixture shaken for a few minutes. The heterogeneous mixture was cooled to 4 °C for 1h and the solids then collected by filtration (2.0g). The solids were confirmed to be product by LC-MS. Step 3 Into a 250 mL round bottomed flask was placed the amine from the previous step (1.5 g, 3.5 mmol), DMF (35mL) and a magnetic stir bar. Succinic anhydride (1.8g, 17.7mmol) was added in a single portion followed by triethylamine (6.6 mL, 47.2 mmol). The mixture was stirred for 2h at which point TLC indicated the reaction was complete (TLC elution 4:1 DCM:MeOH). The mixture was concentrated on the rotovap and then treated with potassium carbonate solution (1M, 50 mL). The mixture was allowed to stir for 60min. The mixture was acidified with hydrochloric acid (20%, 50 mL) and the product precipitated. The mixture was cooled on ice for 30 min and the solids were collected medium frit glass filter. Solid weight 1.32g. The product was confirmed by LC-MS. Step 4 In a small 20 mL glass vial was placed 6,7-dihydroxy-4-oxaheptylamine (443 mg, 3.0 mmol, Berry & Associates Cat#LK4010) and DMF (5 mL). The sample was warmed on a heat plate set a 60 °C for 10 min prior to assembling the other components of the reaction. In a 200 mL round bottomed flask was placed the Succinylated Ala-Ala derivative prepared in the prior step (1.3 g, 2.5 mmol), a magnetic stir bar and DMF (18 mL) . HATU (1.1 g, 3.0 mmol, Anaspec) was added and the mixture was stirred for 5 min. To the reaction flask was added with the DMF-amino diol solution in the 20 mL glass vial. The vial was rinsed with 2 mL of DMF and this was added to the reaction vessel. Diisopropylethylamine (1.3 mL, 7.44 mmol) was added, the flask was capped and allowed to stir overnight at room temperature. The magnetic stir bar was removed and the mixture was concentrated on the rotovap and placed on the high vacuum overnight. The reaction was purified on a silica column (Teledyne-Isco) and elution with a dichloromethane:methanol gradient (yield 1.3 g). The product was confirmed by LC-MS. Step 5 Fmoc protecting group is cleaved with a base such as piperidine, 4-methylpiperidine, piperazine, 1,8-diazabicyclo[5.4.0]undec-7-ene, or morpholine. A chose of the base depends on how acidic a substrate is resistant towards. The Fmoc protected diol is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF. In a separate dried round bottom flask with a magnetic star bar, 20% piperidine in 80% DMF is prepared. To the Fmoc protected diol containing DMF solution, 20% piperidine in 80% DMF is added and the resulting mixture is allowed to mix at under inert gas, at room temperature, until reaction completion verified by TLC and LC-UV/MS analysis. At reaction completion the solvents are stripped off by rotary evaporation, under vacuum (10 mbar), with heating (55 °C). The concentrated residue is placed under full vacuum, at room temperature, for several hours resulting in crude Compound. The product is isolated by flash chromatography 24 g silica column (Teledyne-ISCO) and eluted with a dichloromethane:methanol gradient. Step 6 Carbonylated Camptothecin derivative prepared according to the above carbonylation is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF (6.10 mL). The carbonylated Camptothecin derivative is allowed to dissolve completely under inert gas at room temperature. Then, to the mixture is added DIPEA (0.351 g, 2.013 mmole), followed by addition of HATU (0.278 g, 0.732 mmole). The amine diol (0.610 mmole) is added to a dried round bottom flask, under inert gas blanket, with magnetic stir bar, followed by addition of DMF (6.10 mL), and dissolve completely at room temperature. Then the carbonylated Camptothecin derivative reaction mixture is added to the solution containing the amine diol; the resultant mixture is allowed to mix at under inert gas, at room temperature, until reaction completion verified by TLC and LC-UV/MS analysis. At reaction completion the solvents are stripped off by rotary evaporation, under vacuum (10 mbar), with heating (55 °C). The concentrated residue is placed under full vacuum, at room temperature, for several hours resulting in crude alanine linked SN38. Alanine linked camptonthecin can also be prepared similarly with the camptonthecin derivative with a hydroxyl group at 11 position instead of 10 position. Step 7 In a 200 mL round bottomed flask with magnetic stir bar is placed the diol prepared in the prior step (1.9 mmol) and pyridine (38 mL). 4,4-dimethoxytrityl chloride is added to the solution in a single portion. The flask is capped and allowed to stir overnight. The mixture is concentrated under reduced pressure and the product is isolated by flash chromatography 24 g silica column (Teledyne-ISCO) and eluted with a dichloromethane:methanol gradient resulting in crude product. Step 8 The tritylated product is placed in 50 mL RB flask with magnetic stir bar (0.28 mmol). Dichloromethane (5.5 mL) and 4 to 5.4 Å molecular sieves are added. 2-Cyanoethyl N,N- diisopropylchlorophosphoramidite (192 μL, 0.6 mmol) and diisopropylethylamine (192 μL, 1.1 mmol) are added via syringe. The reaction is stirred for 2h. The reaction is monitored by TLC (100% DCM with 5% TEA - prewashed TLC plates). The reaction is concentrated and taken up on silica gel (12 g column – Teledyne-ISCO) and eluted with a hexane:ethylacetate gradient with 2.5% trimethylamine resulting in alanine linked SN38. EXAMPLE 5 SYNTHESIS OF DOXORUBICIN-PEG-AZIDE Doxorubicin-PEG-azide was synthesized according to the reaction sequence shown above on a 50 mg scale. Azide 7-1 was reacted with dihydrofuran-2,5-dione to afford intermediate 7-2, which was reacted with perfluorophenol to afford intermediate 7-3. Intermediate 7-3 was coupled to doxorubicin to afford the desired product, doxorubicin-PEG- azide in good yield (74%). The presence of the desired product was confirmed by LC-MS. Similarly, SN38 azide, alanine linked SN38, and monomethyl auristatin E (MMAE) azide were prepared and their structures are shown below:
Figure imgf000232_0001
EXAMPLE 6 SYNTHESIS OF DOXORUBICIN POLYMER An exemplary alkynyl-containing polymer is coupled to doxorubicin-PEG-azide. Reaction conditions include CuSO4, tris(3-hydroxypropyltriazolylmethyl)amine (THPTA), and sodium ascorbate. The reaction is carried out in phosphate buffered aqueous solvent with 60% DMS at a pH of 7.6. The reaction is run at room temperature and the presence of the desired product is confirmed by LC-MS. D’ indicates another biologically active moiety attached to the backbone structure (I)-(VI) different from doxorubicin. Other moieties in a compound such as a fluorescent dye/another alkyne moiety which may be present and attached to the backbone structure (I)-(VI) are omitted for the clarity purposes in the reaction scheme shown above. In some embodiments, SN38 azide, alanine linked SN38, and monomethyl auristatin E (MMAE) azide were also coupled to the compound of structure (II), (V), or (VI) via click reaction as shown above. EXAMPLE 7 SYNTHESIS OF COMPOUND I-1 Stock solution preparation Borate buffer prepared at 250 mM, pH 10 Fluorscein-NHS solution prepared at 350 mM (300 mg in 1.35 mL DMSO:acetonitrile at 25:75) Solid Phase Synthesis Compound I-1 is prepared on the DNA synthesizer via solid support using standard DNA synthesis techniques (i.e., DMT protected 2-cyanoethyl phosphoramidite). The polymer is removed from the solid support with ammonium hydroxide and lyophilized to a paste. 250 mg aliquots are reconstituted in water. A small aliquot is removed and serial dilutions are prepared in 100 mM NaCO3 at pH 9 to determine concentration (A 263 ε = 10,000). Final stock concentration is found to be 14.5 mM. Dye Coupling Reaction In 50 mL centrifuge tube equipped with magnetic stir bar is placed water (1.110 µL), borate buffer (1.800 µL), Compound I-1 polymer solution (466 µL), acetonitrile (137.5 µL), triethylamine (313 µL) and fluorescein-NHS solution (675 µL). The tube is wrapped in aluminum foil and the mixture stirred overnight at room temperature. Size Exclusion Filtration To an Amicon Ultra-15 Centrifugal filter (Millipore UFC900324, MW cutoff = 3000) is added 1 mL of water. The crude reaction from the dye coupling reaction (4.5 mL) is added to the filtration setup. The reaction vessel is rinsed 2 × with 4 mL of 100 mM NaOH and the rinseates are transferred to the filtration setup. The filtration setup is centrifuged at max speed (3220 g, swing bucket, 30 minutes). The filtrate is removed and the retentate treated with an additional 10 mL of 100 mM NaOH. The filtration setup is centrifuged as before. Again, the filtrate is removed and a third 10 mL 100 mM NaOH aliquot is added to the retentate. The setup is centrifuged as before and the filtrate removed. A fourth 10 mL 100 mM NaOH aliquot is added to the retentate and centrifuged as before. The filtrate is removed and 10 mL of water are added to the filtration setup. The mixture is centrifuged as before. The retentate is removed, the filtration vessel is washed with water and the rinesates are added to the final volume (3.5 mL). The desired product is confirmed by LC-MS and absorbance is used to determine concentration. EXAMPLE 8 ACTIVATION AND ANTIBODY CONJUGATION OF COMPOUND I-1 The maleimide functionalized Compound I-1 is prepared according to the method described in Example 1. In parallel, an UCHT-1 antibody is treated with bis-maleimidoethane ("BMOE") to reduce disulfide bonds. The reduced antibody is reacted with Compound I-1 in a 5:1 molar ratio of polymer to antibody. The reaction results in a final product having a polymer to antibody ratio of 1:1 as detected by size exclusion chromatography. In some embodiments, anti-CD33, anti-CD70, or anti-CD123 may be used with bis-maleimidoethane ("BMOE") to reduce disulfide bonds.
Figure imgf000236_0001
A mixture of CuSO4 (2mM), sodium ascorbate (10mM), tris- hydroxypropyltriazolylmethylamine (THPTA) (4mM) in water was stirred vigorously at room temperature for 15 minutes. To the mixture compound II-10 (5mM)) containing alkyne moiety and doborubicin azide (25mM) were added sequentially and the resultant mixture was stirred for 8 hours at room temperature. The reaction mixture was diluted by adding EtOAc and aqueous NH4Cl solution. The mixture was stirred for an additional 30 minutes, and two layers were separated. The aqueous layer was extracted with EtOAc twice. The combined organic layers were dried over Na2SO4 and evaporated by a rotovap. The crude product obtained was optionally further purified by recrystallization, but the crude product had enough purity to use without further purification. Presence of compound I-29 was confirmed by a mass spec. The mass spectra of the crude product compound I-29 after 4 hours is included hereinbelow. At 4 hours the reaction was a 1:1 mixture between conjugated and unconjugated polymer. Theoretical MW of I-29 is 5307 and observed MW is 5307.3.
EXAMPLE 10 GENERAL SYNTHESIS OF PEPTIDE BACKBONES VIA SOLID PHASE SYNTHESIS The reaction scheme above illustrates an exemplary method for preparing an intermediate useful for preparation of compounds of structure (III), where PG is a suitable protecting group, X is a functional unit that a peptide chain can be built on, the shaded circle is a suitable solid support, and "BAM" is a biologically active moiety, or fragment thereof, a prodrug of a biologically active moiety, or fragment thereof, a fluorescent dye, an imaging agent, or a radioisotope binding site, provided at least one occurrence of M is not a fluorescent dye. Small porous beads are initially treated with functional units, which bind to the surface of the porous beads. Peptide chains are built upon the functional units sites and remain covalently bonded to the bead until they are cleaved. When attached, a peptide chain is immobilized on the solid phase and retained during a filtration process, wherein liquid reagents and by-products of the synthesis are washed away. The general cycle of solid phase synthesis is one of repeated cycles of deprotection-wash- coupling-wash. A free N-terminal amine of a peptide, attached to a solid support, is coupled to an N-protected amino acid group (e.g., with Fmoc or Boc). The newly introduced amino acid unit is deprotected to reveal a new N-terminal amine, which is further reacted with additional amino acids. The process is repeated and the peptide chain is elongated. When the peptide chain has incorporated all desired amino acid and monomer units, it is cleaved from the bead. Cleaving reagents such as anhydrous hydrogen fluoride or trifluoroacetic acid can be used to cleave peptide chains from beads. The peptide chain is then collected, purified and characterized. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, including U.S. Provisional Patent Application No.63/409,065, filed on September 22, 2022, are incorporated herein by reference, in their entirety to the extent not inconsistent with the present description. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments. From the foregoing it will be appreciated that, although specific embodiments of the disclosure have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the disclosure. Accordingly, the disclosure is not limited except as by the appended claims.

Claims

CLAIMS What is claimed is: 1. A compound having the following Structure (I): (I) or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein: R1 and R2 are each independently H, OH, SH, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; R3 is, at each occurrence, independently H, alkyl or alkoxy; R4 is, at each occurrence, independently OH, SH, O-, S-, ORd or SRd; R5 is, at each occurrence, independently oxo or thioxo; R6 and R7 are, at each occurrence, independently H, OH, or halo; L1a is, at each occurrence, independently a heteroalkylene or heteroarylene linker; L1b is, at each occurrence, independently either i) H or ii) a linker; L2, L3, L5, and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; L4 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker; L7 is, at each occurrence, independently a linker; M1 is, at each occurrence, independently either i) absent or ii) a moiety comprising a biologically active moiety, provided that M1 is absent when L1b is H; M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of Structure (I); and m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, an integer of zero or greater; p is, at each occurrence, an integer of zero or greater; w is, at each occurrence, an integer of zero or greater; and n is an integer of one or greater, wherein the sum of p and w is an integer of two or greater, and the compound comprises at least two different biologically active moieties.
2. The compound of claim 1, wherein L1a is, at each occurrence independently an optionally substituted 5-7 membered heteroarylene linker.
3. The compound of claim 1 or 2, wherein L1a has one of the following structures: or .
4. The compound of claim 1 or 2, wherein L1a has the following structure when L1b and M1 are absent: ; wherein: R8 is, at each occurrence, independently O, NH, or NRe; R9 is, at each occurrence, independently H, alkyl, or optionally substituted alkyl; R10 is, at each occurrence, independently H or F; and Re is, at each occurrence, independently alkyl or optionally substituted alkyl.
5. The compound of any one of claims 1-3 having the following structure (IA): (IA) or a stereoisomer, salt or tautomer thereof.
6. The compound of any one of claims 1-2 and 4 having the following structure (IB): (IB) or a stereoisomer, salt or tautomer thereof.
7. The compound of any one of claims 1-4 having the following structure (IC): v(IC) or a stereoisomer, salt or tautomer thereof, wherein k is, at each occurrence, an integer of zero or greater.
8. The compound of any one of claims 1-7, wherein at least one occurrence of L4 is heteroalkylene, or wherein each occurrence of L4 is heteroalkylene.
9. The compound of claim 8, wherein the heteroalkylene comprises alkylene oxide.
10. The compound of claim 9, wherein the heteroalkylene comprises ethylene oxide.
11. The compound of claim 10, wherein the ethylene oxide is polyethylene oxide.
12. The compound of any one of claims 1-11, wherein L4, at each occurrence, has the following structure: , wherein: z is an integer from 1 to 100; and * indicates a bond to the adjacent phosphorous atom.
13. The compound of claim 12, wherein z is an integer from 3 to 8 or an integer from 22 to 26.
14. The compound of any one of claims 12-13 having one of the following structures (ID), (IE), or (IF): (ID) (IE) or (IF) or a stereoisomer, salt or tautomer thereof.
15. The compound of any one of claims 1-14, wherein at least one occurrence of L5 is alkylene.
16. The compound of any one of claims 1-15, wherein each occurrence of L5 is alkylene.
17. The compound of any one of claims 1-16, wherein at least one occurrence of L3 is alkylene.
18. The compound of any one of claims 1-17, wherein each occurrence of L3 is alkylene.
19. The compound of any one of claims 1-18, wherein at least one occurrence of L6 is a direct bond.
20. The compound of any one of claims 17-19 having one of the following structures (IG), (IH), or (IJ): (IG) (IH) or (IJ) or a stereoisomer, salt or tautomer thereof, wherein: y1 is, at each occurrence, independently an integer from 0 to 6; and y2 and y3 are, at each occurrence, independently an integer from 1 to 6.
21 The compound of any one of claims 1-20, wherein at least one occurrence of L1b comprises a functional group formed by reaction of an aldehyde, oxime, hydrazone, alkyne, amine, azide, acylazide, acylhalide, nitrile, nitrone, sulfhydryl, disulfide, sulfonyl halide, isothiocyanate, imidoester, activated ester, ketone, α , β-unsaturated carbonyl, alkene, maleimide, α-haloimide, epoxide, aziridine, tetrazine, tetrazole, phosphine, biotin, or thiirane with a complementary reactive group.
22. The compound of claim 21, wherein at least one occurrence of L1b comprises a functional group formed by a reaction of an alkyne and an azide.
23. The compound of claim 22, wherein at least one occurrence of L1b is a linker comprising a triazolyl functional group.
24. The compound of any one of claims 1-23, wherein at least one occurrence of L1b- M1 comprises one of the following structures: or , wherein Lc and Ld are each independently optional linkers.
25. The compound of claim 24, wherein Lc or Ld, or both, is absent.
26. The compound of claim 24, wherein Lc or Ld, or both, is present.
27. The compound of claim 26, wherein Lc and Ld, when present, are each independently alkylene or heteroalkylene.
28. The compound of claim 26, wherein Lc and Ld independently have one of the following structures: ; ; ; ; ; ; or .
29. The compound of any one of claims 1-28, wherein M1-L1b has one of the following structures: ; ; or , wherein a, b, c, and d are each independently an integer ranging from 1-6.
30. The compound of any one of claims 1-29, wherein at least one occurrence of M1- L1b has one of the following structures: ; ; ; ; or .
31. The compound of any one of claims 1-30, wherein each occurrence of M1-L1b has one of the following structures: ; ; ; ; or .
32. A compound having the following structure (III): (III) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; L2, L3, L5 and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; L7 is, at each occurrence, independently a linker; R21 is, at each occurrence, independently a natural or unnatural amino acid side chain; R1 and R2 are each independently H, OH, SH, –NH2, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of structure (III); m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, independently an integer of zero or greater; v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties.
33. The compound of claim 32, wherein at least one R21 is a neutral amino acid side chain.
34. The compound of any one of claims 32 or 33, wherein at least one R21 is a charged amino acid side chain.
35. The compound of any one of claims 32-34, wherein R21 is, at each occurrence, independently H, alkyl, ˗CH2CO2-, ˗CH2CH2CO2-, ˗CH2CH2CH2CH2NH3 +, ˗CH2CH2CH2NHC(=NH2 +)NH2 or imidazolyl.
36. The compound of any one of claims 32-35, wherein R21, L5 and m are selected such that has an amino acid sequence of (G)10, (GDGDGDGDGD) or (GKGKGKGKGK).
37. The compound of claim 32, wherein R21, L5 and m are selected such that has an amino acid sequence capable of forming an α-helix or β-sheet secondary structure.
38. The compound of claim 37, wherein the amino acid sequence is (GGEEFMLVYKFARKHGG) or (GGMSMVVSGG).
39. The compound of any one of claims 32-38, wherein L5 or L3, or both, is present for at least one occurrence.
40. The compound of claim 39, wherein, when present, L5 or L3, or both, is a heteroalkylene linker.
41. The compound of claim 40, wherein the heteroalkylene linker comprises a functional group capable of maintaining a positive or negative charge at pH values ranging from 3 to 11 in aqueous solution.
42. The compound of claim 41, wherein at least one occurrence of L5 or L3, or both, has the following structure: .
43. The compound of claim 41, wherein at least one occurrence of L5 or L3, or both, has the following structure: .
44. The compound of any one of claims 32-43, wherein L6 and L2 are independently absent or a heteroalkylene linker.
45. The compound of claim 44, wherein the heteroalkylene linker is a peptidyl linker.
46. A compound having the following structure (IV): (IV) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; L7 is, at each occurrence, independently a linker; L2, L5 and L6 are, at each occurrence, independently a direct bond or independently an optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker; L8 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker comprising one or more charged moieties, provided at least one charged moiety is not a phosphate ester; R3 is, at each occurrence, independently H, alkyl or alkoxy; R1 and R2 are each independently -H, -OH, -SH, alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl, alkoxycarbonyl, ˗OP(=Ra)(Rb)Rc, Q or a protected form thereof, or L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a solid support or solid support residue, a linker comprising a covalent bond to a solid support or solid support residue or a linker comprising a covalent bond to a further compound of structure (IV); q is, at each occurrence, independently an integer of zero or greater; v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties.
47. The compound of claim 46, wherein the charged moieties are positively charged.
48. The compound of claim 47, wherein the charged moieties independently have one of the following structures: ; or wherein: R is, at each occurrence, independently H or C1-C6 alkyl.
49. The compound of claim 46, wherein the charged moieties are negatively charged.
50. The compound of claim 49, wherein the charged moieties have the following structure: .
51. The compound of any one of claims 46-50, wherein L8 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or ’ wherein: R is, at each occurrence, independently H or C1-C6 alkyl; x is an integer from 0 to 6; and m is an integer of 1 or greater.
52. The compound of any one of claims 1-51, wherein at least one occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof.
53. The compound of claim 52, wherein L7 comprises an amide or ester functional group.
54. The compound of any one of claims 1-53, wherein at least one occurrence of L7 or L1b comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or .
55. The compound of any one of claims 1-54, wherein each occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence, a ketone, a diol, a cyano, a nitro or combinations thereof.
56. The compound of any one of claims 1-55, wherein each occurrence of L7 or L1b comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or .
57. The compound of any one of claims 1-56, wherein at least one occurrence of R3 is H.
58. The compound of any one of claims 1-57, wherein R5 is, at each occurrence, oxo.
59. The compound of any one of claims 1-58, wherein R4 is, at each occurrence, O-.
60. The compound of any one of claims 1-59, wherein R1 and R2 are each independently OH or -OP(=Ra)(Rb)Rc.
61. The compound of any one of claims 1-60, wherein one of R1 or R2 is OH or - OP(=Ra)(Rb)Rc, and the other of R1 or R2 is Q or a linker comprising a covalent bond to Q.
62. The compound of any one of claims 1-61, wherein R1 and R2 are each independently -OP(=Ra)(Rb)Rc.
63. The compound of any one of claims 1-62, wherein R2 is L'.
64. The compound of any one of claims 1-63, wherein R2 is –NH2.
65. The compound of any one of claims 1-64, wherein one of R2 or R1 is L' and L' is a linker comprising a covalent bond to a solid support.
66. The compound of claim 65, wherein the solid support is a polymeric bead or non- polymeric bead.
67. The compound of any one of claims 1-66, wherein L' is a linker to a targeting moiety.
68. The compound of any one of claims 1-67, wherein L' is a linker to a targeting moiety, the linker comprising an alkylene oxide or phosphodiester moiety, or combinations thereof.
69. The compound of any one of claims 1-68, wherein L' has one of the following structures: ; ; ; or wherein: x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer from 1 to 10; Rb is H, an electron pair or a counter ion; and L'' is the targeting moiety or a linkage to the targeting moiety.
70. The compound of any one of claims 1-69, wherein the targeting moiety is an antibody or cell surface receptor antagonist.
71. The compound of claim 70, wherein the antibody or cell surface receptor antagonist is an epidermal growth factor receptor (EGFR) inhibitor, a hepatocyte growth factor receptor (HGFR) inhibitor, an insulin-like growth factor receptor (IGFR) inhibitor, a folate, or a MET inhibitor.
72. The compound of claim 1-71, wherein the targeting moiety is a monoclonal antibody.
73. The compound of claim 72, wherein the monoclonal antibody is Abciximab, Adalimumab, Alemtuzumab, Alirocumab, Avibactam, Basiliximab, Benralizumab, Bezlotoxumab, Blinatumomab, Brodalumab, Burosumab, Canakinumab, Caplacizumab, Certolizumab pegol, Daclizumab, Denosumab, Dupilumab, Eculizumab, Emicizumab, Erenumab, Evolocumab, Fremanezumab, Galcanezumab, Golimumab, Guselkumab, Ibalizumab, Idarucizumab, Infliximab, Itolizumab, Ixekizumab, Lanadelumab, Lokivetmab, Mepolizumab, Natalizumab, Obiltoxaximab, Ocrelizumab, Omalizumab, Palivizumab, Ranibizumab, Raxibacumab, Reslizumab, Rmab, Rovelizumab, Ruplizumab, Sarilumab, Secukinumab, Tildrakizumab, Thiomab, Tocilizumab, Ustekinumab, Vedolizumab, Abrilumab, Actoxumab, Aducanumab, Afasevikumab, Afelimomab, Anifrolumab, Anrukinzumab (IMA-638), Aselizumab, Atorolimumab, Bapineuzumab, BCD-100, Bertilimumab, Besilesomab, Biciromab, Bimagrumab, Bimekizumab, Birtamimab, Bleselumab, Blosozumab, Bococizumab, Brazikumab, Briakinumab, Brolucizumab, Carlumab, Carotuximab, Cedelizumab, Clazakizumab, Clenoliximab, Concizumab, Cosfroviximab, CR6261, Crenezumab, Crizanlizumab, Crotedumab, Depatuxizumab, mafodotin, Derlotuximab biotin, Dezamizumab, Diridavumab, Domagrozumab, Dusigitumab, Ecromeximab, Edobacomab, Efalizumab, Efungumab, Eldelumab, Elezanumab, Enokizumab, Eptinezumab, Erlizumab, Etrolizumab, Evinacumab, Exbivirumab, Fanolesomab, Faralimomab, Faricimab, Fasinumab, Felvizumab, Fezakinumab, Flanvotumab, Fletikumab, Flotetuzumab, Fontolizumab, Foravirumab, Frovocimab, Fulranumab, Gantenerumab, Gavilimomab, Gevokizumab, Gimsilumab, Gomiliximab, Gosuranemab, Ianalumab, Inclacumab, Inolimomab, Iomab-B, Keliximab, Lampalizumab, Landogrozumab, Larcaviximab, Lebrikizumab, Lenvervimab, Lerdelimumab, Letolizumab, Libivirumab, Ligelizumab, Lodelcizumab, Lulizumab pegol, Marstacimab, Mavrilimumab, Metelimumab, Mirikizumab, Motavizumab, Muromonab CD3, Nebacumab, Nemolizumab, NEOD001, Nirsevimab, Odulimomab, Olendalizumab, Olokizumab, OMS721, Opicinumab, Orticumab, Otelixizumab, Otilimab, Oxelumab, Ozanezumab, Ozoralizumab, Pagibaximab, Panobacumab, Pascolizumab, Pateclizumab, PDR001, Perakizumab, Pexelizumab, Placulumab, Plozalizumab, Ponezumab, Porgaviximab, Prasinezumab, Priliximab, PRO 140, Quilizumab, Rafivirumab, Ralpancizumab, Ranevetmab, Ravagalimab, Ravulizumab, Refanezumab, Regavirumab, Relatlimab, Rinucumab, Risankizumab, Roledumab, Romosozumab, Rontalizumab, SA237, Satralizumab, Sevirumab, SHP647, Sifalimumab, Simtuzumab, Siplizumab, Sirukumab, Solanezumab, Sonepcizumab, Spartalizumab, Stamulumab, Sulesomab, Suptavumab, Sutimlimab, Suvizumab, Suvratoxumab, Tadocizumab, Talizumab, Tamtuvetmab, Tanezumab, Tefibazumab, Telimomab aritox, Teneliximab, Teplizumab, Teprotumumab, Tezepelumab, Tibulizumab, Toralizumab, Tralokinumab, Trevogrumab, Tuvirumab, Ulocuplumab, Urtoxazumab, Varisacumab, Vepalimomab, Vesencumab, Visilizumab, Vobarilizumab, Zolimomab aritox, trastuzumab, gemtuzumab, brentuximab, vorsetuzumab, lorvotuzumab, cantuzumab, bivatuzumabor inotuzumab, or vadastuximab.
74. The compound of claim 1-73, wherein the targeting moiety is an antibody specific for a tumor cell antigen.
75. The compound of claim 74, wherein the tumor cell antigen is an EGFR, a HER 2, a folate receptor, CD 20 or CD 33.
76. The compound of any one of claims 1-75, wherein R1 or R2 has one of the following structures: ; ; ; ; ; ; ; ; ; ; or , wherein Ra is H or a solid support.
77. The compound of any one of claims 1-76, wherein R2 has one of the following structures: ; or .
78. The compound of any one of claims 1-77, wherein R1 has the following structure: .
79. The compound of any one of claims 1-78, wherein M2 is, at one or more occurrences, independently a moiety comprising four or more aryl or heteroaryl rings, or combinations thereof.
80. The compound of any one of claims 1-79, wherein M2 is, at one or more occurrences, independently fluorescent or colored.
81. The compound of claim 80, wherein M2 is fluorescent.
82. The compound of any one of claims 1-81, wherein M2 is, at one or more occurrences, independently comprise a fused-multicyclic aryl or heteroaryl moiety comprising at least four fused rings.
83. The compound of any one of claims 1-82, wherein M2 is, at each occurrence, independently selected from the group consisting of phenyl, (bis-fluorophenyl-difluorobora- diaza-indacene)phenyl, quaterphenyl, bi-benzothiazole, ter-benzothiazole, bi-naphthyl, bi- anthracyl, squaraine, squarylium, 9,10-ethynylanthracene, and ter-naphthyl moiety.
84. The compound of any one of claims 1-83, wherein M2 is, at each occurrence, independently selected from the group consisting of p-terphenyl, perylene, azobenzene, phenazine, phenanthroline, acridine, thioxanthrene, chrysene, rubrene, coronene, cyanine, perylene imide, perylene amide, and derivatives thereof.
85. The compound of any one of claims 1-84, wherein M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, resorufin dye, dipyrrometheneboron difluoride dye, ruthenium bipyridyl dye, thiazole orange dye, polymethine, and N-aryl-1,8-naphthalimide dye.
86. The compound of any one of claims 1-85, wherein M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, boron-dipyrromethene, rhodamine, cyanine, pyrene, perylene, perylene monoimide, 6-FAM, 5-FAM, 6-FITC, 5-FITC, and derivatives thereof.
87. The compound of any one of claims 1-86, wherein M2, at each occurrence, independently has one of the following structures: ; ; ; ; ; ; or .
88. The compound of any one of claims 1-87, wherein at least one occurrence of M2 has the following structure:
.
89. The compound of any one of claims 1-88, wherein each occurrence of M2 has the following structure: .
90. The compound of any one of claims 1-89, wherein at least one occurrence of –L7- M2 has one of the following structures: .
91. The compound of any one of claims 1-90, wherein each occurrence of – L7- M2 has one of the following structures: .
92. The compound of any one of claims 1-91, wherein at least one occurrence of M1 or M3 are an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
93. The compound of any one of claims 1-92, wherein each occurrence of M1 or M3 are an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
94. The compound of any one of claims 1-93, wherein at least one occurrence of M1 or M3 are a nitrogen mustard, a nitrosourea, a tetrazine, an aziridine, a cisplatin or cisplatin derivative, or a non-classical alkylating agent.
95. The compound of any one of claims 1-94, wherein at least one occurrence of M1 or M3 are mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide, busulfan, N-nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU), semustine (MeCCNU), fotemustine, streptozotocin, dacarbazine, mitozolomide, temozolomide, thiotepa, mytomycin, diaziquone (AZQ), cisplatin, carboplatin, oxaliplatin, procarbazine, or hexamethylmelamine.
96. The compound of any one of claims 1-95, wherein at least one occurrence of M1 or M3 are an anti-folate, a fluoropyrimidines, a deoxynucleoside analogue, or a thiopurine.
97. The compound of any one of claims 1-96, wherein at least one occurrence M1 or M3 are methotrexate, pemetrexed, fluorouracil, capecitabine, cytarabine, gemcitabine, decitabine, azacitidine, fludarabine, nelarabine, cladribine, clofarabine, pentostatin, thioguanine, and mercaptopurine.
98. The compound of any one of claims 1-97, wherein at least one occurrence of M1 or M3 are an auristatin, a Vinca alkaloid, or a taxane.
99. The compound of any one of claims 1-98, wherein at least one occurrence of M1 or M3 are auristatin F, auristatin E, vincristine, vinblastine, vinorelbine, vindesine, vinflunine, paclitaxel, docetaxel, etoposide, or teniposide.
100. The compound of any one of claims 1-99, wherein at least one occurrence of M1 or M3 are irinotecan, SN 38, topotecan, camptothecin, doxorubicin, mitoxantrone, teniposide. novobiocin, merbarone, or aclarubicin.
101. The compound of any one of claims 1-100, wherein at least one occurrence of M1 or M3 are an anthracycline or a bleomycin.
102. The compound of any one of claims 1-101, wherein at least one occurrence of M1 or M3 are doxorubicin, daunorubicin, epirubicin, idarubicin, pirarubicin, aclarubicin, or mitoxantrone.
103. The compound of any one of claims 1-102, wherein at least one occurrence of M1 or M3 has one of the following structures: ; ; ; ; ; ; ; ; ; or .
104. The compound of any one of claims 1-103, wherein each occurrence of M1 or M3 has one of the following structures: ; ; ; ; ; ; ; ; ; or .
105. The compound of any one of claims 1-104, wherein at least one occurrence of M3 is an anti-inflammatory compound.
106. The compound of claim 105, wherein at least one occurrence of M3 is a steroid.
107. The compound of claim 105 or 106, wherein at least one occurrence of M3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone.
108. The compound of any of one of claims 105-107, wherein at least one occurrence of M3 has one of the following structures: or , wherein: R11 is H or halogen; R12 and R13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R14 is H, halogen, OH, or alkyl; R15 is H or OH; R16, R17, and R18, are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond.
109. The compound of any of one of claims 1-108, wherein at least one occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or .
110. The compound of any one of claims 1-109, wherein at least one occurrence of M3 is a Vitamin.
111. The compound of claim 110, wherein at least one occurrence of M3 is a Vitamin E.
112. The compound of claim 110 or 111, wherein at least one occurrence of M3 is α- tocopherol, β-tocopherol, γ-tocopherol, δ-tocopherol, α -tocotrienol, β -tocotrienol, γ -tocotrienol, or δ -tocotrienol.
113. The compound of any of one of claims 110-112, wherein at least one occurrence of M3 has the following structure: , wherein: R19 and R20 are independently H or CH3; and represents a single or double carbon-carbon bond.
114. The compound of any of one of claims 1-113, wherein at least one occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; or .
115. The compound of any one of claims 1-114, wherein Q has the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or –NH2.
116. The compound of any one of claims 1-115, wherein m is from 0 to 10.
117. The compound of any one of claims 1-116, wherein m is 0, 1, 2, 3, 4, or 5.
118. The compound of any one of claims 1-116, wherein m is 1 or 2.
119. The compound of any one of claims 1-118, wherein n is 1, 2, 3, or 4.
120. The compound of any one of claims 1-119, wherein n is 1 or 2.
121. The compound of any one of claims 1-120, wherein q is 0, 1, 2, 3, or 4.
122. The compound of any one of claims 1-120, wherein q is 1 or 2.
123. The compound of any one of claims 1-122, wherein p is 1, 2, 3, or 4.
124. The compound of any one of claims 1-122, wherein p is 2 or 3.
125. The compound of any one of claims 1-124, wherein w is 1, 2, 3, or 4.
126. The compound of any one of claims 1-124, wherein w is 1 or 2.
127. The compound of any one of claims 1-126, wherein v is an integer from 1 to 100.
128. The compound of any one of claims 1-126, wherein v is an integer from 1 to 10.
129. The compound of any one of claims 1-128, wherein y1 is, at each occurrence, an integer 0 or 1; and y2, and y3 are, at each occurrence, 1.
130. The compound of any one of claims 1-129, wherein n is 1; q is 2; w is 2, p is 2; and m is 1.
131. The compound of any one of claims 1-129, wherein n is 1; q is 2; w is 1, p is 3; and m is 1.
132. The compound of any one of claims 1-131, wherein a ratio of M1:M2:M3 is 1:1:1.
133. The compound of any one of claims 1-131, wherein a ratio of M1:M2:M3 is 1:2:3.
134. The compound of any one of claims 1-131, wherein a ratio of M1:M2:M3 is 2:1:1.
135. The compound of any one of claims 1-134, wherein at least one occurrence of R6 or R7 is F or H.
136. The compound of any one of claims 1-135, wherein at least each occurrence of R6 and R7 are F or H.
137. The compound of any one of claims 1-136, wherein at least each occurrence of R6 and R7 are F, R8 is O, R9 is H, and R10 is H.
138. The compound of any one of claims 1-137, wherein the compound has one of the structures of Table 2A, 2B, 2C, 2D, or 2E or a salt or tautomer thereof.
139. A pharmaceutical composition comprising the compound of any one of claims 1- 138, and a pharmaceutically acceptable carrier, diluent, or excipient.
140. A method of treating a disease or disorder, comprising administering a therapeutically effective amount of a compound of any one of claims 1-138, or the pharmaceutical composition of claim 139, to a subject in need thereof.
141. The method of claim 140, wherein the disease or disorder is cancer.
142. The method of claim 140 or 141, wherein the cancer is breast cancer, stomach cancer, lung cancer, ovarian cancer, lymphoma, or bladder cancer.
143. A compound having the following structure (II): (II) or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein: R1 and R2 are each independently H, OH, SH, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; R3 is, at each occurrence, independently H, alkyl or alkoxy; R4 is, at each occurrence, independently OH, SH, O-, S-, ORd or SRd; R5 is, at each occurrence, independently oxo or thioxo; R6 and R7 are, at each occurrence, independently H, OH, or halo; R8 is, at each occurrence, independently O, NH, or NRe; R9 is, at each occurrence, independently H, alkyl, or optionally substituted alkyl; R10 is, at each occurrence, independently H or F; Re is, at each occurrence, independently alkyl or optionally substituted alkyl; L2, L3, L5, and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; L4 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker; L7 is, at each occurrence, independently a linker; M2 is, at each occurrence, independently a moiety comprising a fluorescent dye; M3 is, at each occurrence, independently a moiety comprising a biologically active moiety; G is, at each occurrence, independently a moiety reactive under cycloaddition; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of Structure (I); and m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, an integer of zero or greater; p is, at each occurrence, an integer of zero or greater; w is, at each occurrence, an integer of zero or greater; k is, at each occurrence, an integer of zero or greater; x9 is, at each occurrence, an integer from 1 to 6; and n is an integer of one or greater, wherein the sum of p and w is an integer of two or greater, and the compound comprises at least two different biologically active moieties.
144. The compound of claim 143, wherein at least one occurrence of L4 is heteroalkylene, or wherein each occurrence of L4 is heteroalkylene.
145. The compound of claim 144, wherein the heteroalkylene comprises alkylene oxide.
146. The compound of claim 145, wherein the heteroalkylene comprises ethylene oxide.
147. The compound of claim 146, wherein the ethylene oxide is polyethylene oxide.
148. The compound of any one of claims 143-147, wherein L4, at each occurrence, has the following structure: , wherein: z is an integer from 1 to 100; and * indicates a bond to the adjacent phosphorous atom.
149. The compound of claim 148, wherein z is an integer from 3 to 8 or an integer from 22 to 26.
150. The compound of any one of claims 143-149 having the following structure (IIA): (IIA) or a stereoisomer, salt or tautomer thereof.
151. The compound of any one of claims 143-150, wherein at least one occurrence of L5 is alkylene.
152. The compound of any one of claims 143-151, wherein each occurrence of L5 is alkylene.
153. The compound of any one of claims 143-152, wherein at least one occurrence of L3 is alkylene.
154. The compound of any one of claims 143-153, wherein each occurrence of L3 is alkylene.
155. The compound of any one of claims 143-154, wherein at least one occurrence of L6 is a direct bond.
156. The compound of any one of claims 143-155 having the following structure (IIB): (IIB) or a stereoisomer, salt or tautomer thereof, wherein: y1 is, at each occurrence, independently an integer from 0 to 6; and y2, and y3 are, at each occurrence, independently an integer from 1 to 6.
157. A compound having the following structure (V): (V) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: G2 is, at each occurrence, independently a moiety reactive under cycloaddition; G3 is, at each occurrence, independently a moiety reactive under cycloaddition; L7 is, at each occurrence, independently a linker; L2, L3, L5 and L6 are, at each occurrence, independently a direct bond or independently optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linkers; R21 is, at each occurrence, independently a natural or unnatural amino acid side chain; R1 and R2 are each independently H, OH, SH, –NH2, alkyl, alkoxy, alkylether, heteroalkyl, ˗OP(=Ra)(Rb)Rc, Q, or a protected form thereof, L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a linker comprising a covalent bond to a solid support, a linker comprising a covalent bond to a solid support residue, a solid support residue, a linker comprising a covalent bond to a nucleoside, or a linker comprising a covalent bond to a further compound of structure (V); m is, at each occurrence, independently an integer of zero or greater; q is, at each occurrence, independently an integer of zero or greater; and v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties.
158. The compound of claim 157, wherein at least one R21 is a neutral amino acid side chain.
159. The compound of any one of claims 157 or 158, wherein at least one R21 is a charged amino acid side chain.
160. The compound of any one of claims 157-159, wherein R21 is, at each occurrence, independently H, alkyl, ˗CH2CO2-, ˗CH2CH2CO2-, ˗CH2CH2CH2CH2NH3 +, ˗CH2CH2CH2NHC(=NH2 +)NH2 or imidazolyl.
161. The compound of any one of claims 157-160, wherein R21, L5 and m are selected such that has an amino acid sequence of (G)10, (GDGDGDGDGD) or (GKGKGKGKGK).
162. The compound of claim 157, wherein R21, L5 and m are selected such that has an amino acid sequence capable of forming an α-helix or β-sheet secondary structure.
163. The compound of claim 162, wherein the amino acid sequence is (GGEEFMLVYKFARKHGG) or (GGMSMVVSGG).
164. The compound of any one of claims 157-163, wherein L5 or L3, or both, is present for at least one occurrence.
165. The compound of claim 164, wherein, when present, L5 or L3, or both, is a heteroalkylene linker.
166. The compound of claim 165, wherein the heteroalkylene linker comprises a functional group capable of maintaining a positive or negative charge at pH values ranging from 3 to 11 in aqueous solution.
167. The compound of claim 166, wherein at least one occurrence of L5 or L3, or both, has the following structure: .
168. The compound of claim 166, wherein at least one occurrence of L5 or L3, or both, has the following structure: .
169. The compound of any one of claims 157-168, wherein L6 and L2 are independently absent or a heteroalkylene linker.
170. The compound of claim 169, wherein the heteroalkylene linker is a peptidyl linker.
171. A compound having the following structure (VI): (VI) or a stereoisomer, pharmaceutically acceptable salt or tautomer thereof, wherein: G2 is, at each occurrence, independently a moiety reactive under cycloaddition; G3 is, at each occurrence, independently a moiety reactive under cycloaddition; L7 is, at each occurrence, independently a linker; L2, L5 and L6 are, at each occurrence, independently a direct bond or independently an optional alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, heteroalkynylene or heteroatomic linker; L8 is, at each occurrence, independently an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene linker comprising one or more charged moieties, provided at least one charged moiety is not a phosphate ester; R3 is, at each occurrence, independently H, alkyl or alkoxy; R1 and R2 are each independently -H, -OH, -SH, alkyl, alkoxy, alkylether, heteroalkyl, alkylaminyl, alkylcarbonyl, alkoxycarbonyl, ˗OP(=Ra)(Rb)Rc, Q or a protected form thereof, or L'; Ra is O or S; Rb is OH, SH, O-, S-, ORd or SRd; Rc is OH, SH, O-, S-, ORd, OL', SRd, alkyl, alkoxy, heteroalkyl, heteroalkoxy, alkylether, alkoxyalkylether, phosphate, thiophosphate, phosphoalkyl, thiophosphoalkyl, phosphoalkylether or thiophosphoalkylether; Rd is a counter ion; Q is, at each occurrence, independently a moiety comprising a reactive group, or protected form thereof, capable of forming a covalent bond with a complementary reactive group Q′ on a targeting moiety; L' is, at each occurrence, independently a linker comprising a covalent bond to Q, a targeting moiety, a linker comprising a covalent bond to a targeting moiety, a solid support or solid support residue, a linker comprising a covalent bond to a solid support or solid support residue or a linker comprising a covalent bond to a further compound of structure (VI); q is, at each occurrence, independently an integer of zero or greater; v is, at each occurrence, independently an integer of one or greater; and n is an integer of two or greater, wherein the compound comprises at least two different biologically active moieties.
172. The compound of claim 171, wherein the charged moieties are positively charged.
173. The compound of claim 172, wherein the charged moieties independently have one of the following structures: ; or wherein: R is, at each occurrence, independently H or C1-C6 alkyl.
174. The compound of claim 171, wherein the charged moieties are negatively charged.
175. The compound of claim 174, wherein the charged moieties have the following structure: .
176. The compound of any one of claims 171-175, wherein L8 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or ’ wherein: R is, at each occurrence, independently H or C1-C6 alkyl; x is an integer from 0 to 6; and m is an integer of 1 or greater.
177. The compound of any one of claims 171-176, wherein at least one occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence comprising one or more amino acid residues, a ketone, a diol, a cyano, a nitro, or combinations thereof.
178. The compound of claim 177, wherein L7 comprises an amide or ester functional group.
179. The compound of any one of claims 171-178, wherein at least one occurrence of L7 comprises one of the following structures: ; ; ; ; ; ; ; ; ; ; ; or .
180. The compound of any one of claims 171-179, wherein each occurrence of L7 comprises an amide bond, an ester bond, a phosphodiester bond, a disulfide bond, a double bond, a triple bond, an ether bond, a hydrazone, an amino acid sequence, a ketone, a diol, a cyano, a nitro or combinations thereof.
181. The compound of any one of claims 171-180, wherein each occurrence of L7 comprises one of the following structures: c ; ; ; ; ; ; ; ; ; ; ; or .
182. The compound of any one of claims 171-181, wherein at least one occurrence of R3 is H.
183. The compound of any one of claims 171-182, wherein R5 is, at each occurrence, O-.
184. The compound of any one of claims 171-183, wherein R4 is, at each occurrence, oxo.
185. The compound of any one of claims 171-184, wherein R1 and R2 are each independently OH or -OP(=Ra)(Rb)Rc.
186. The compound of any one of claims 171-185, wherein one of R1 or R2 is OH or - OP(=Ra)(Rb)Rc, and the other of R1 or R2 is Q or a linker comprising a covalent bond to Q.
187. The compound of any one of claims 171-186, wherein R1 and R2 are each independently -OP(=Ra)(Rb)Rc.
188. The compound of any one of claims 171-187, wherein R2 is L'.
189. The compound of any one of claims 171-188, wherein L' is a linker to a targeting moiety.
190. The compound of any one of claims 171-189, wherein L' is a linker to a targeting moiety, the linker comprising an alkylene oxide or phosphodiester moiety, or combinations thereof.
191. The compound of any one of claims 171-190, wherein L' has one of the following structures: ; ; ; or wherein: x1, x2, x3, x4, x5, x6, x7 and x8 are independently an integer from 1 to 10; Rb is H, an electron pair or a counter ion; and L'' is the targeting moiety or a linkage to the targeting moiety.
192. The compound of any one of claims 171-191, wherein the targeting moiety is an antibody or cell surface receptor antagonist.
193. The compound of claim 192, wherein the antibody or cell surface receptor antagonist is an epidermal growth factor receptor (EGFR) inhibitor, a hepatocyte growth factor receptor (HGFR) inhibitor, an insulin-like growth factor receptor (IGFR) inhibitor, a folate, or a MET inhibitor.
194. The compound of any one of claims 171-193, wherein the targeting moiety is a monoclonal antibody.
195. The compound of claim 194, wherein the monoclonal antibody is Abciximab, Adalimumab, Alemtuzumab, Alirocumab, Avibactam, Basiliximab, Benralizumab, Bezlotoxumab, Blinatumomab, Brodalumab, Burosumab, Canakinumab, Caplacizumab, Certolizumab pegol, Daclizumab, Denosumab, Dupilumab, Eculizumab, Emicizumab, Erenumab, Evolocumab, Fremanezumab, Galcanezumab, Golimumab, Guselkumab, Ibalizumab, Idarucizumab, Infliximab, Itolizumab, Ixekizumab, Lanadelumab, Lokivetmab, Mepolizumab, Natalizumab, Obiltoxaximab, Ocrelizumab, Omalizumab, Palivizumab, Ranibizumab, Raxibacumab, Reslizumab, Rmab, Rovelizumab, Ruplizumab, Sarilumab, Secukinumab, Tildrakizumab, Thiomab, Tocilizumab, Ustekinumab, Vedolizumab, Abrilumab, Actoxumab, Aducanumab, Afasevikumab, Afelimomab, Anifrolumab, Anrukinzumab (IMA-638), Aselizumab, Atorolimumab, Bapineuzumab, BCD-100, Bertilimumab, Besilesomab, Biciromab, Bimagrumab, Bimekizumab, Birtamimab, Bleselumab, Blosozumab, Bococizumab, Brazikumab, Briakinumab, Brolucizumab, Carlumab, Carotuximab, Cedelizumab, Clazakizumab, Clenoliximab, Concizumab, Cosfroviximab, CR6261, Crenezumab, Crizanlizumab, Crotedumab, Depatuxizumab, mafodotin, Derlotuximab biotin, Dezamizumab, Diridavumab, Domagrozumab, Dusigitumab, Ecromeximab, Edobacomab, Efalizumab, Efungumab, Eldelumab, Elezanumab, Enokizumab, Eptinezumab, Erlizumab, Etrolizumab, Evinacumab, Exbivirumab, Fanolesomab, Faralimomab, Faricimab, Fasinumab, Felvizumab, Fezakinumab, Flanvotumab, Fletikumab, Flotetuzumab, Fontolizumab, Foravirumab, Frovocimab, Fulranumab, Gantenerumab, Gavilimomab, Gevokizumab, Gimsilumab, Gomiliximab, Gosuranemab, Ianalumab, Inclacumab, Inolimomab, Iomab-B, Keliximab, Lampalizumab, Landogrozumab, Larcaviximab, Lebrikizumab, Lenvervimab, Lerdelimumab, Letolizumab, Libivirumab, Ligelizumab, Lodelcizumab, Lulizumab pegol, Marstacimab, Mavrilimumab, Metelimumab, Mirikizumab, Motavizumab, Muromonab CD3, Nebacumab, Nemolizumab, NEOD001, Nirsevimab, Odulimomab, Olendalizumab, Olokizumab, OMS721, Opicinumab, Orticumab, Otelixizumab, Otilimab, Oxelumab, Ozanezumab, Ozoralizumab, Pagibaximab, Panobacumab, Pascolizumab, Pateclizumab, PDR001, Perakizumab, Pexelizumab, Placulumab, Plozalizumab, Ponezumab, Porgaviximab, Prasinezumab, Priliximab, PRO 140, Quilizumab, Rafivirumab, Ralpancizumab, Ranevetmab, Ravagalimab, Ravulizumab, Refanezumab, Regavirumab, Relatlimab, Rinucumab, Risankizumab, Roledumab, Romosozumab, Rontalizumab, SA237, Satralizumab, Sevirumab, SHP647, Sifalimumab, Simtuzumab, Siplizumab, Sirukumab, Solanezumab, Sonepcizumab, Spartalizumab, Stamulumab, Sulesomab, Suptavumab, Sutimlimab, Suvizumab, Suvratoxumab, Tadocizumab, Talizumab, Tamtuvetmab, Tanezumab, Tefibazumab, Telimomab aritox, Teneliximab, Teplizumab, Teprotumumab, Tezepelumab, Tibulizumab, Toralizumab, Tralokinumab, Trevogrumab, Tuvirumab, Ulocuplumab, Urtoxazumab, Varisacumab, Vepalimomab, Vesencumab, Visilizumab, Vobarilizumab, Zolimomab aritox, trastuzumab, gemtuzumab, brentuximab, vorsetuzumab, lorvotuzumab, cantuzumab, bivatuzumabor inotuzumab, or vadastuximab.
196. The compound of any one of claims 171-195, wherein R1 or R2 has one of the following structures: ; ; ; ; ; ; ; ; ; ; or , wherein Ra is H or a solid support.
197. The compound of any one of claims 171-196, wherein R2 has one of the following structures: ; or .
198. The compound of any one of claims 171-197, wherein R1 has the following structure: .
199. The compound of any one of claims 171-198, wherein M2 is, at one or more occurrences, independently a moiety comprising four or more aryl or heteroaryl rings, or combinations thereof.
200. The compound of any one of claims 171-199, wherein M2 is, at one or more occurrences, independently fluorescent or colored.
201. The compound of claim 200, wherein M2 is fluorescent.
202. The compound of any one of claims 171-201, wherein M2 is, at one or more occurrences, independently comprise a fused-multicyclic aryl or heteroaryl moiety comprising at least four fused rings.
203. The compound of any one of claims 171-202, wherein M2 is, at each occurrence, independently selected from the group consisting of phenyl, (bis-fluorophenyl-difluorobora- diaza-indacene)phenyl, quaterphenyl, bi-benzothiazole, ter-benzothiazole, bi-naphthyl, bi- anthracyl, squaraine, squarylium, 9,10-ethynylanthracene, and ter-naphthyl moiety.
204. The compound of any one of claims 171-203, wherein M2 is, at each occurrence, independently selected from the group consisting of p-terphenyl, perylene, azobenzene, phenazine, phenanthroline, acridine, thioxanthrene, chrysene, rubrene, coronene, cyanine, perylene imide, perylene amide, and derivatives thereof.
205. The compound of any one of claims 171-204, wherein M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, resorufin dye, dipyrrometheneboron difluoride dye, ruthenium bipyridyl dye, thiazole orange dye, polymethine, and N-aryl-1,8-naphthalimide dye.
206. The compound of any one of claims 171-205, wherein M2 is, at each occurrence, independently selected from the group consisting of a coumarin dye, boron-dipyrromethene, rhodamine, cyanine, pyrene, perylene, perylene monoimide, 6-FAM, 5-FAM, 6-FITC, 5-FITC, and derivatives thereof.
207. The compound of any one of claims 171-206, wherein M2, at each occurrence, independently has one of the following structures: ; ; ; ; ; ; or .
208. The compound of any one of claims 171-207, wherein at least one occurrence of M2 has the following structure: .
209. The compound of any one of claims 171-208, wherein each occurrence of M2 has the following structure: .
210. The compound of any one of claims 171-209, wherein at least one occurrence of – L7- M2 has one of the following structures: .
211. The compound of any one of claims 171-210, wherein each occurrence of – L7- M2 has one of the following structures: .
212. The compound of any one of claims 171-211, wherein at least one occurrence of M3 is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
213. The compound of any one of claims 171-212, wherein each occurrence of M3 is an alkylating agent, an antimetabolite, a microtubule inhibitor, a topoisomerase inhibitor, or a cytotoxic antibiotic.
214. The compound of any one of claims 171-213, wherein at least one occurrence of M3 is a nitrogen mustard, a nitrosourea, a tetrazine, an aziridine, a cisplatin or cisplatin derivative, or a non-classical alkylating agent.
215. The compound of any one of claims 171-214, wherein at least one occurrence of M3 is mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide, busulfan, N- nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine (CCNU), semustine (MeCCNU), fotemustine, streptozotocin, dacarbazine, mitozolomide, temozolomide, thiotepa, mytomycin, diaziquone (AZQ), cisplatin, carboplatin, oxaliplatin, procarbazine, or hexamethylmelamine.
216. The compound of any one of claims 171-215, wherein at least one occurrence of M3 is an anti-folate, a fluoropyrimidines, a deoxynucleoside analogue, or a thiopurine.
217. The compound of any one of claims 171-216, wherein at least one occurrence M3 is methotrexate, pemetrexed, fluorouracil, capecitabine, cytarabine, gemcitabine, decitabine, azacitidine, fludarabine, nelarabine, cladribine, clofarabine, pentostatin, thioguanine, and mercaptopurine.
218. The compound of any one of claims 171-217, wherein at least one occurrence of M3 is an auristatin, a Vinca alkaloid, or a taxane.
219. The compound of any one of claims 171-218, wherein at least one occurrence of M3 is auristatin F, auristatin E, vincristine, vinblastine, vinorelbine, vindesine, vinflunine, paclitaxel, docetaxel, etoposide, or teniposide.
220. The compound of any one of claims 171-219, wherein at least one occurrence of M3 is irinotecan, SN 38, topotecan, camptothecin, doxorubicin, mitoxantrone, teniposide, novobiocin, merbarone, or aclarubicin.
221. The compound of any one of claims 171-220, wherein at least one occurrence of M3 is an anthracycline or a bleomycin.
222. The compound of any one of claims 171-221, wherein at least one occurrence of M3 is doxorubicin, daunorubicin, epirubicin, idarubicin, pirarubicin, aclarubicin, or mitoxantrone.
223. The compound of any one of claims 171-222, wherein at least one occurrence of M3 is an anti-inflammatory compound.
224. The compound of claim 223, wherein at least one occurrence of M3 is a steroid.
225. The compound of claim 223 or 224, wherein at least one occurrence of M3 is cholesterol, cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, or beclomethasone.
226. The compound of any of one of claims 223-225, wherein at least one occurrence of M3 has one of the following structures: or , wherein: R11 is H or halogen; R12 and R13 are independently OH, H, alkyl, substituted alkyl, or heteroalkyl; R14 is H, halogen, OH, or alkyl; R15 is H or OH; R16, R17, and R18, are H, alkyl or substituted alkyl; and represents a single or double carbon-carbon bond.
227. The compound of any one of claims 171-226, wherein at least one occurrence of M3 is one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or .
228. The compound of any one of claims 171-227, wherein each occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or .
229. The compound of any one of claims 171-228, wherein at least one occurrence of M3 is a Vitamin.
230. The compound of claim 229, wherein at least one occurrence of M3 is a Vitamin E.
231. The compound of claim 229 or 230, wherein at least one occurrence of M3 is α- tocopherol, β-tocopherol, γ-tocopherol, δ-tocopherol, α -tocotrienol, β -tocotrienol, γ -tocotrienol, or δ -tocotrienol.
232. The compound of any of one of claims 229-231, wherein at least one occurrence of M3 has the following structure: , wherein: R19 and R20 are independently H or CH3; and represents a single or double carbon-carbon bond.
233. The compound of any of one of claims 171-232, wherein at least one occurrence of M3 has one of the following structures: ; ; ; ; ; ; ; or
.
234. The compound of any one of claims 171-233, wherein Q has the following structures: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; or –NH2.
235. The compound of any one of claims 171-234, wherein the cycloaddition is click reaction.
236. The compound of any one of claims 171-235, wherein G, G2, or G3 has one of the following structures: ; or .
237. The compound of any one of claims 171-236, wherein G, G2, or G3, at each occurrence, has the following structure: .
238. The compound of any of claims 171-237, wherein x9 is an integer of 4, 5, or 6.
239. The compound of claim 238, wherein x9 is an integer of 4.
240. The compound of any one of claims 171-239, wherein m is 0, 1, 2, 3, 4, or 5.
241. The compound of any one of claims 171-240, wherein m is 1 or 2.
242. The compound of any one of claims 171-241, wherein n is 1, 2, 3, or 4.
243. The compound of any one of claims 171-242, wherein n is 1 or 2.
244. The compound of any one of claims 171-243, wherein q is 0, 1, 2, 3, or 4.
245. The compound of any one of claims 171-244, wherein q is 1 or 2.
246. The compound of any one of claims 171-245, wherein p is 1, 2, 3, or 4.
247. The compound of any one of claims 171-246, wherein p is 2 or 3.
248. The compound of any one of claims 171-247, wherein w is 1, 2, 3, or 4.
249. The compound of any one of claims 171-248, wherein w is 1 or 2.
250. The compound of any one of claims 171-249, wherein k is 0, 1, 2, 3, or 4.
251. The compound of any one of claims 171-250, wherein k is 0 or 1.
252. The compound of any one of claims 171-251, wherein y1 is, at each occurrence, an integer 0 or 1; and y2, and y3 are, at each occurrence, 1.
253. The compound of any one of claims 171-252, wherein n is 1; q is 2; w is 2, p is 2; and m is 1.
254. The compound of any one of claims 171-252, wherein n is 1; q is 2; w is 1, p is 3; and m is 1.
255. The compound of any one of claims 171-254, wherein at least one occurrence of R6 or R7 is F or H.
256. The compound of any one of claims 171-255, wherein at least each occurrence of R6 and R7 are F or H.
257. The compound of any one of claims 171-256, wherein at least each occurrence of R6 and R7 are F, R8 is O, R9 is H, and R10 is H.
258. The compound of any one of claims 171-256, wherein at least each occurrence of R6 and R7 are H, R8 is O, R9 is H, and R10 is H.
259. The compound of any one of claims 171-258, wherein each occurrence of x9 is 2, 4, or 6.
260. The compound of any one of claims 171-259, wherein each occurrence of x9 is 2 or 4.
261. The compound of any one of claims 171-260, wherein the compound has one of the structures of Table 3 or a salt or tautomer thereof.
PCT/IB2023/058834 2022-09-22 2023-09-07 Polymers with dual therapeutic agents WO2024062319A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263409065P 2022-09-22 2022-09-22
US63/409,065 2022-09-22

Publications (1)

Publication Number Publication Date
WO2024062319A1 true WO2024062319A1 (en) 2024-03-28

Family

ID=88287441

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2023/058834 WO2024062319A1 (en) 2022-09-22 2023-09-07 Polymers with dual therapeutic agents

Country Status (1)

Country Link
WO (1) WO2024062319A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019140227A1 (en) * 2018-01-12 2019-07-18 Sony Corporation Phosphoalkyl ribose polymers comprising biologically active compounds

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019140227A1 (en) * 2018-01-12 2019-07-18 Sony Corporation Phosphoalkyl ribose polymers comprising biologically active compounds

Similar Documents

Publication Publication Date Title
JP7403744B2 (en) programmable dendritic drug
US11312736B1 (en) Programmable polymeric drugs
EP3737419B1 (en) Phosphoalkyl polymers comprising biologically active compounds
US11931419B2 (en) Programmable polymeric drugs
EP3737417A1 (en) Phosphoalkyl ribose polymers comprising biologically active compounds
US20200353089A1 (en) Polymers with rigid spacing groups comprising biologically active compounds
EP3724200A1 (en) Ionic polymers comprising biologically active compounds
WO2020210689A1 (en) Programmable polymeric drugs
WO2020210692A1 (en) Programmable polymeric drugs
WO2020210694A1 (en) Programmable polymeric drugs
WO2024062319A1 (en) Polymers with dual therapeutic agents
WO2023052971A1 (en) Automated synthesis of polymeric dual drugs
WO2023052970A1 (en) Pyrrolobenzodiazepine conjugates for cancer treatment
WO2024062318A1 (en) Polymers with steroids or vitamins
KR20240063130A (en) Automated synthesis of polymeric dual drugs
KR20240062140A (en) Pyrrolobenzodiazepine conjugates for cancer treatment
WO2023002431A1 (en) Automated synthesis of polymeric drugs
CN117750977A (en) Automatic synthesis of polymeric drugs