WO2017210246A2 - Conjugués de pénicillamine et particules et formulations associées - Google Patents

Conjugués de pénicillamine et particules et formulations associées Download PDF

Info

Publication number
WO2017210246A2
WO2017210246A2 PCT/US2017/035107 US2017035107W WO2017210246A2 WO 2017210246 A2 WO2017210246 A2 WO 2017210246A2 US 2017035107 W US2017035107 W US 2017035107W WO 2017210246 A2 WO2017210246 A2 WO 2017210246A2
Authority
WO
WIPO (PCT)
Prior art keywords
conjugate
cancer
particles
poly
targeting moiety
Prior art date
Application number
PCT/US2017/035107
Other languages
English (en)
Other versions
WO2017210246A3 (fr
Inventor
Brian H. White
Benoît MOREAU
Patrick Rosaire BAZINET
Mark T. Bilodeau
Original Assignee
Tarveda Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tarveda Therapeutics, Inc. filed Critical Tarveda Therapeutics, Inc.
Publication of WO2017210246A2 publication Critical patent/WO2017210246A2/fr
Publication of WO2017210246A3 publication Critical patent/WO2017210246A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/655Somatostatins
    • C07K14/6555Somatostatins at least 1 amino acid in D-form
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5431IL-11
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • C07K7/083Neurotensin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention generally relates to the field of targeting ligands, conjugates thereof, and particles for drug delivery. More particularly, the invention relates to the use conjugates comprising penicillamine linkers.
  • Nanoparticulate drug delivery systems are attractive for systemic drug delivery because they may be able to prolong the half-life of a drug in circulation, reduce nonspecific uptake of a drug, and improve accumulation of a drug at tumors, e.g., through an enhanced permeation and retention (EPR) effect.
  • EPR enhanced permeation and retention
  • therapeutics formulated for delivery as nanoparticles which include DOXIL®
  • ABRAXANE® albumin bound paclitaxel nanoparticles
  • Applicants have created molecules that are conjugates of a targeting moiety attached to an active agent, e.g., a cancer therapeutic agent with a linker, wherein the linker comprises a penicillamine group or derivative thereof.
  • Linkers comprising a penicillamine group or derivative are hereinafter referred to as penicillamine linkers.
  • conjugates can be encapsulated into particles. The conjugates and particles are useful for delivering active agents such as tumor cytotoxic agents to cells.
  • the conjugates include a targeting ligand and an active agent connected by a linker, wherein the conjugate in some embodiments has the formula:
  • X is a targeting moiety
  • Y is a penicillamine linker
  • Z is an active agent
  • a method of reducing proliferation, increasing apoptosis, or increasing arrest of cells comprises administering a conjugate to the cells, wherein the conjugate comprises an active agent coupled to a targeting moiety by a penicillamine linker.
  • a method of treating a tumor, reducing volume of a tumor or delivering an active agent to a tumor in a subject comprises administering a conjugate to the subject, wherein the conjugate comprises an active agent coupled to a targeting moiety by a penicillamine linker.
  • a method of treating neuroendocrine cancers wherein the neuroendocrine cancer is selected from small cell lung cancer (SCLC), pheochromocytoma, neuroblastoma, ganglioneuroma, paraganglioma, carcinoids, gastrinoma, glucagonoma, vasoactive intestinal polypeptide-secreting tumor, pancreatic polypeptide-secreting tumor, nonfunctioning gastroenteropancreatic tumors, meduallary thyroid cancer, Merkel cell tumor of the skin, pituitary adenoma, and pancreatic cancer.
  • the method comprises administering a conjugate to the cells, wherein the conjugate comprises an active agent coupled to a targeting moiety by a penicillamine linker.
  • Applicants have designed conjugates comprising a targeting moiety attached to an active agent with a penicillamine linker to deliver the active agent to a disease tissue target.
  • Penicillamine is an a-amino acid metabolite of penicillin and has no antibiotic properties.
  • Penicillamine as used herein may be JJ-penicillamine shown below, or J-penicillamine. In WO 2007/022493, the contents of which are
  • Leamon et al. designed a compound comprising folate connected to vinca alkaloid with penicillamine. JJ-penicillamine
  • the active agent may be attached through a disulfide bond incorporating the sulfur atom on the penicillamine linker.
  • the targeting moiety may be attached to the N-terminus or C-terminus of the penicillamine linker, or the penicillamine residue may be part of the targeting ligand.
  • conjugate (B) comprising [0012]
  • the penicillamine linker may be substituted at any position.
  • the -OH group of conjugate (A) may be substituted with group Rl :
  • Rl may be any suitable chemical group, such as - H2 or substituted - H2, an alkyl group (e.g., -CH3 group) or a substituted alkyl group (e.g., an alkoxy group), a cyclic group or a substituted cyclic group, a
  • heterocyclic group e.g. a piperidine group
  • substituted heterocyclic group e.g. a substituted heterocyclic group.
  • the - H2 group of conjugate (B) may be substituted with group
  • R2 may be any suitable chemical group, such as an alkyl group (e.g., -CH3 group) or a substituted alkyl group (e.g., an oxyalkyl group), a cyclic group or a substituted cyclic group, a heterocyclic group or a substituted heterocyclic group.
  • alkyl group e.g., -CH3 group
  • substituted alkyl group e.g., an oxyalkyl group
  • penicillamine as an agent to create hindered disulfide bonds can yield advantages over other reagents used to create hindered disulfide bonds such as N-succinimidyl 4-methyl-4-(2-pyridyldithio)pentanoate (SMPP) (see Kellogg, et. al., Bioconj. Chem., 22:717-727, 2011), used to conjugate a hindered disulfide bond at lysine residues.
  • the penicillamine residue can be incorporated in a peptide sequence, either at the N-terminus, C-terminus, or within the targeting sequence as necessary.
  • Including a targeting moiety in the conjugates can, for example, improve the amount of active agent at a site and decrease active agent toxicity to the subject.
  • toxicity refers to the capacity of a substance or composition to be harmful or poisonous to a cell, tissue organism or cellular environment.
  • Low toxicity refers to a reduced capacity of a substance or composition to be harmful or poisonous to a cell, tissue organism or cellular environment. Such reduced or low toxicity may be relative to a standard measure, relative to a treatment or relative to the absence of a treatment.
  • Toxicity may further be measured relative to a subject's weight loss where weight loss over 15%, over 20% or over 30% of the body weight is indicative of toxicity.
  • Other metrics of toxicity may also be measured such as patient presentation metrics including lethargy and general malaiase.
  • Neutropenia or thrombopenia may also be metrics of toxicity.
  • Pharmacologic indicators of toxicity include elevated AST/ ALT levels, neurotoxicity, kidney damage, GI damage and the like.
  • the toxicity of a conjugate containing a targeting moiety linked to an active agent for cells that do not bind to the targeting moiety is predicted to be decreased compared to the toxicity of the active agent alone. Without committing to any particular theory, applicants believe that this feature is because the ability of the conjugated active agent to enter a cell is decreased compared the ability to enter a cell of the active agent alone. Accordingly, the conjugates comprising an active agent and particles containing the conjugates as described herein generally have decreased toxicity for cells that do not bind to the targeting moiety and at least the same or increased toxicity for cells that bind to the targeting moiety compared to the active agent alone.
  • Conjugates include an active agent or prodrug thereof attached to a targeting moiety by a penicillamine linker.
  • the conjugates can be a conjugate between a single active agent and a single targeting moiety, e.g., a conjugate having the structure X-Y-Z, where X is the targeting moiety, Y is the penicillamine linker, and Z is the active agent.
  • the conjugate contains more than one targeting moiety, more than one linker, more than one active agent, or any combination thereof, wherein at least one linker is a penicillamine linker.
  • the conjugate can have any number of targeting moieties, linkers, and active agents, wherein at least one linker is a penicillamine linker.
  • the conjugate can have the structure X-Y-Z-Y-X, (X-Y)n-Z, X-(Y-Z)n, X-Y-Zn, (X-Y-Z)n, (X-Y-Z-Y)n-Z, Xn-Y-Z where X is a targeting moiety, Y is a linker, Z is an active agent, and n is an integer between 1 and 50, between 2 and 20, for example, between 1 and 5, wherein at least one linker is a penicillamine linker.
  • Each occurrence of X, Y, and Z can be the same or different, e.g., the conjugate can contain more than one type of targeting moiety, more than one type of linker, and/or more than one type of active agent, wherein at least one linker is a penicillamine linker.
  • the conjugate can contain more than one targeting moiety attached to a single active agent.
  • the conjugate can include an active agent with multiple targeting moieties each attached via a different linker.
  • the conjugate can have the structure X-Y-Z-Y-X where each X is a targeting moiety that may be the same or different, each Y is a linker that may be the same or different, and Z is the active agent, wherein at least one linker is a penicillamine linker.
  • the conjugate can contain more than one active agent attached to a single targeting moiety.
  • the conjugate can include a targeting moiety with multiple active agents each attached via a different linker, wherein at least one linker is a penicillamine linker.
  • the conjugate can have the structure Z-Y-X-Y-Z where X is the targeting moiety, each Y is a linker that may be the same or different, and each Z is an active agent that may be the same or different, wherein at least one linker is a penicillamine linker.
  • the conjugate has a structure of Al :
  • X is a targeting moiety and Z is an active agent.
  • the linker moiety of Al may be substituted with any chemical group.
  • the conjugate may have a structure of A2:
  • Rl may be any suitable chemical group, such as - H2 or substituted - H2, an alkyl group (e.g., -CH3 group) or a substituted alkyl group (e.g., an alkoxy group), a cyclic group or a substituted
  • cyclic group e.g. a piperidine group ⁇ /
  • a heterocyclic group e.g. a piperidine group ⁇ /
  • a substituted heterocyclic group e.g. a piperidine group ⁇ /
  • the conjugate has a structure of Bl :
  • X is a targeting moiety and Z is an active agent.
  • the linker moiety of Bl may be substituted with any chemical group.
  • the conjugate may have a structure of B2:
  • X is a targeting moiety
  • Z is an active agent
  • R2 may be any suitable chemical group, such as an alkyl group (e.g., -CH3 group) or a substituted alkyl group (e.g., an oxyalkyl group), a cyclic group or a substituted cyclic group, a heterocyclic group or a substituted heterocyclic group.
  • alkyl group e.g., -CH3 group
  • R2 may be any suitable chemical group, such as an alkyl group (e.g., -CH3 group) or a substituted alkyl group (e.g., an oxyalkyl group), a cyclic group or a substituted cyclic group, a heterocyclic group or a substituted heterocyclic group.
  • a conjugate as described herein contains at least one active agent (a first active agent).
  • the conjugate can contain more than one active agent, that can be the same or different from the first active agent.
  • the active agent can be a therapeutic, prophylactic, diagnostic, or nutritional agent.
  • a variety of active agents are known in the art and may be used in the conjugates described herein.
  • the active agent can be a protein or peptide, small molecule, nucleic acid or nucleic acid molecule, lipid, sugar, glycolipid, glycoprotein, lipoprotein, or combination thereof.
  • the active agent is an antigen, an adjuvant, radioactive, an imaging agent (e.g., a fluorescent moiety) or a polynucleotide.
  • the active agent is an organometallic compound.
  • the active agent is selected from a maytansinoid or derivative such as mertansine (DM1) or DM4, cabazitaxel, SN-38, or doxorubicin.
  • the active agent can be a cancer therapeutic.
  • Cancer therapeutics include, for example, death receptor agonists such as the TNF-related apoptosis-inducing ligand (TRAIL) or Fas ligand or any ligand or antibody that binds or activates a death receptor or otherwise induces apoptosis.
  • TRAIL TNF-related apoptosis-inducing ligand
  • Suitable death receptors include, but are not limited to, TNFR1, Fas, DR3, DR4, DR5, DR6, LTpR and combinations thereof.
  • Cancer therapeutics such as chemotherapeutic agents, cytokines, chemokines, and radiation therapy agents can be used as active agents.
  • Chemotherapeutic agents include, for example, alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumor agents. Such agents typically affect cell division or DNA synthesis and function. Additional examples of therapeutics that can be used as active agents include monoclonal antibodies and the tyrosine kinase inhibitors e.g. imatinib mesylate, which directly targets a molecular abnormality in certain types of cancer (e.g., chronic myelogenous leukemia, gastrointestinal stromal tumors).
  • tyrosine kinase inhibitors e.g. imatinib mesylate
  • Chemotherapeutic agents include, but are not limited to cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, vincristine, vinblastine, vinorelbine, vindesine, taxol and derivatives thereof, irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide, epipodophyllotoxins, trastuzumab, cetuximab, and rituximab, bevacizumab, and combinations thereof. Any of these may be used as an active agent in a conjugate.
  • the active agent is a small molecule having a molecular weight preferably ⁇ about 5 kDa, more preferably ⁇ about 4 kDa, more preferably about 3 kDa, most preferably ⁇ about 1.5 kDa or ⁇ about 1 kDa.
  • antiproliferative agents include cytotoxic compounds (e.g., broad spectrum), angiogenesis inhibitors, cell cycle progression inhibitors, PBK/m- TOR/AKT pathway inhibitors, MAPK signaling pathway inhibitors, kinase inhibitors, protein chaperones inhibitors, HDAC inhibitors, PARP inhibitors, Wnt/Hedgehog signaling pathway inhibitors, RNA polymerase inhibitors and proteasome inhibitors.
  • cytotoxic compounds e.g., broad spectrum
  • angiogenesis inhibitors e.g., cell cycle progression inhibitors, PBK/m- TOR/AKT pathway inhibitors
  • MAPK signaling pathway inhibitors e.g., kinase inhibitors
  • protein chaperones inhibitors e.g., HDAC inhibitors
  • PARP inhibitors e.g., IL-2-activator-like TOR inhibitors
  • Wnt/Hedgehog signaling pathway inhibitors RNA polymerase inhibitors
  • proteasome inhibitors RNA polymerase inhibitor
  • Broad spectrum cytotoxins include, but are not limited to, DNA-binding or alkylating drugs, microtubule stabilizing and destabilizing agents, platinum
  • Exemplary DNA-binding or alkylating drugs include, CC-1065 and its analogs, anthracyclines (doxorubicin, epirubicin, idarubicin, daunorubicin) and its analogs, alkylating agents, such as calicheamicins, dactinomycins, mitomycins, pyrrolobenzodiazepines, trioxacarcins and the like.
  • anthracyclines doxorubicin, epirubicin, idarubicin, daunorubicin
  • alkylating agents such as calicheamicins, dactinomycins, mitomycins, pyrrolobenzodiazepines, trioxacarcins and the like.
  • trioxacarcins include Trioxacarcins DC-45-A2, DC-45-A1, A, D, C7"-epi-C, and C disclosed in Nicolaou et al., J ACS, vol.138:3118 (2016), and trioxacarcin A, DC-45- Al and structural analogues disclosed in Fig. 1 of Magauer et al., Nature Chemistry, vol.5:886 (2013), the contents of each of which are incorporated herein by reference in their entirety.
  • doxorubicin analogs include nemorubicin metabolite or analog drug moiety disclosed in US 20140227299 to Cohen et al., the contents of which are incorporated herein by reference in their entirety.
  • Exemplary CC-1065 analogs include duocarmycin SA, duocarmycin CI, duocarmycin C2, duocarmycin B2, DU-86, KW-2189, bizelesin, seco-adozelesin, and those described in U.S. Patent Nos. 5,475,092; 5,595,499; 5,846,545; 6,534,660; 6,586,618; 6,756,397 and 7,049,316.
  • Doxorubicin and its analogs include PNU- 159682 and those described in U.S. Patent No.6,630,579 and nemorubicin metabolite or analog drugs disclosed in US 20140227299 to Cohen et al., the contents of which are incorporated herein by reference in their entirety.
  • Calicheamicins include those described in U.S. Patent Nos. 5,714,586 and 5,739,116.
  • Duocarmycins include those described in U.S. Patent Nos.5, 070,092; 5, 101,038; 5, 187,186; 6,548,530; 6,660,742; and 7,553,816 B2; and Li et al., Tet Letts., 50:2932 - 2935 (2009).
  • Pyrrolobenzodiazepines include SG2057 and those described in Denny, Exp. Opin. Ther.
  • microtubule stabilizing and destabilizing agents include taxane compounds, such as paclitaxel, docetaxel, cabazitaxel; maytansinoids, auristatins and analogs thereof, tubulysin A and B derivatives, vinca alkaloid derivatives, epothilones, PM060184 and cryptophycins.
  • Exemplary maytansinoids or maytansinoid analogs include maytansinol and maytansinol analogs, maytansine or DM1 and DM4 are those described in U.S. Patent Nos. 5,208,020; 5,416,064; 6,333.410; 6,441, 163; 6,716,821; RE39, 151 and
  • the cytotoxic agent is a maytansinoid, another group of anti-tubulin agents (ImmunoGen, Inc.; see also Chari et al., 1992, Cancer Res. 52: 127-131), maytansinoids or maytansinoid analogs.
  • suitable maytansinoids include maytansinol and maytansinol analogs. Suitable maytansinoids are disclosed in U.S. Patent Nos. 4,424,219; 4,256,746; 4,294,757; 4,307,016;
  • Exemplary auristatins include auristatin E (also known as a derivative of dolastatin-10), auristatin EB (AEB), auristatin EFP (AEFP), monomethyl auristatin E (MMAE), monomethyl auristatin F (MMAF), auristatin F and dolastatin.
  • auristatin E also known as a derivative of dolastatin-10
  • AEB auristatin EB
  • AEFP auristatin EFP
  • MMAE monomethyl auristatin E
  • MMAF monomethyl auristatin F
  • dolastatin dolastatin.
  • Suitable auristatins are also described in U.S. Publication Nos. 2003/0083263, 2011/0020343, and 2011/0070248; PCT Application Publication Nos. WO 09/117531, WO
  • Exemplary tubulysin compounds include compounds described in U.S. Patent Nos. 7,816,377; 7,776,814; 7,754,885; U.S. Publication Nos. 2011/0021568; 2010/004784; 2010/0048490; 2010/00240701; 2008/0176958; and PCT Application Nos. WO 98/13375; WO 2004/005269; WO 2008/138561; WO 2009/002993; WO 2009/055562; WO 2009/012958; WO 2009/026177; WO 2009/134279; WO
  • Exemplary vinca alkaloids include vincristine, vinblastine, vindesine, and navelbine (vinorelbine). Suitable Vinca alkaloids that can be used in the present invention are also disclosed in U.S. Publication Nos. 2002/0103136 and
  • Exemplary epothilone compounds include epothilone A, B, C, D, E and F, and derivatives thereof. Suitable epothilone compounds and derivatives thereof are described, for example, in U.S. Patent Nos. 6,956,036; 6,989,450; 6,121,029;
  • Exemplary platinum compounds include cisplatin (PLATINOL®), carboplatin (PARAPLATIN®), oxaliplatin (ELOX ATINE®), iproplatin, ormaplatin, and tetraplatin.
  • topoisomerase I inhibitors include camptothecin, camptothecin, derivatives, camptothecin analogs and non-natural camptothecins, such as, for example, CPT-11 (irinotecan), SN-38, topotecan, 9-aminocamptothecin, rubitecan, gimatecan, karenitecin, silatecan, lurtotecan, exatecan, diflomotecan, belotecan, lurtotecan and S39625.
  • camptothecin compounds that can be used in the present invention include those described in, for example, J. Med. Chem., 29:2358-2363 (1986); J. Med. Chem., 23 :554 (1980); J. Med. Chem., 30: 1774 (1987).
  • Exemplary topoisomerase II inhibitors include azonafide and etoposide.
  • Lurbinectedin PM01183
  • Trabectedin also known as ecteinascidin 743 or ET-743
  • analogs as described in WO 200107711, WO 2003014127.
  • Angiogenesis inhibitors include, but are not limited to, MetAP2 inhibitors.
  • Exemplary MetAP2 inhibitors include fumagillol analogs, meaning any compound that includes the fumagillin core structure, including fumagillamine, that inhibits the ability of MetAP-2 to remove NFh-terminal methionines from proteins as described in Rodeschini et al., /. Org. Chem., 69, 357-373, 2004 and Liu, et al., Science 282, 1324-1327, 1998.
  • Non-limiting examples of "fumagillol analogs” are disclosed in /. Org. Chem., 69, 357, 2004; J.Org. Chem., 70, 6870, 2005; European Patent Application 0 354 787; /. Med. Chem., 49, 5645, 2006; Bioorg. Med. Chem., 11, 5051, 2003; Bioorg. Med. Chem., 14, 91, 2004; Tet. Lett. 40, 4797, 1999;
  • Exemplary cell cycle progression inhibitors include CDK inhibitors such as BMS-387032 and PD0332991; Rho-kinase inhibitors such as GSK429286;
  • checkpoint kinase inhibitors such as AZD7762; aurora kinase inhibitors such as AZD1152, MLN8054 and MLN8237; PLK inhibitors such as BI 2536, BI6727 (Volasertib), GSK461364, ON-01910 (Estybon); and KSP inhibitors such as SB 743921, SB 715992 (ispinesib), MK-0731, AZD8477, AZ3146 and ARRY-520.
  • PLK inhibitors such as BI 2536, BI6727 (Volasertib), GSK461364, ON-01910 (Estybon)
  • KSP inhibitors such as SB 743921, SB 715992 (ispinesib), MK-0731, AZD8477, AZ3146 and ARRY-520.
  • Exemplary PI3K/m-TOR/AKT signaling pathway inhibitors include phosphoinositide 3 -kinase (PI3K) inhibitors, GSK-3 inhibitors, ATM inhibitors, DNA-PK inhibitors and PDK-1 inhibitors.
  • PI3K phosphoinositide 3 -kinase
  • Exemplary PI3 kinase inhibitors are disclosed in U.S. Patent No. 6,608,053, and include BEZ235, BGT226, BKM120, CALIOI, CAL263, demethoxyviridin, GDC-0941, GSK615, IC87114, LY294002, Palomid 529, perifosine, PF-04691502, PX-866, SAR245408, SAR245409, SF1126, Wortmannin, XL147, XL765,
  • GSK2126458 (Omipalisib), GDC-0326, GDC-0032 (Taselisib, RG7604), PF- 05212384 (Gedatolisib, PKI-587), BAY 80-6946 (copanlisib), PF-04691502, PF- 04989216, PI-103, PKI-402 VS-5584 (SB2343), GDC-0941, NVP-BEZ235
  • Exemplary AKT inhibitors include, but are not limited to, AT7867, MK- 2206, Perifosine, GSK690693, Ipatasertib, AZD5363, TIC10, Afuresertib, SC79, AT13148, PHT-427, A-674563, and CCT128930.
  • Exemplary MAPK signaling pathway inhibitors include MEK, Ras, J K, B-Raf and p38 MAPK inhibitors.
  • MEK inhibitors are disclosed in U.S. Patent No. 7,517,994 and include GDC-0973, GSK1120212, MSC1936369B, AS703026, R05126766 and
  • Exemplary B-raf inhibitors include CDC-0879, PLX-4032, and SB590885.
  • Exemplary B p38 MAPK inhibitors include BIRB 796, LY2228820 and
  • RTK Receptor tyrosine kinases
  • Exemplary inhibitors of ErbB2 receptor include but not limited to AEE788 (NVP-AEE 788), BIBW2992 (Afatinib), Lapatinib, Erlotinib (Tarceva), and Gefitinib (Iressa).
  • Exemplary RTK inhibitors targeting more than one signaling pathway include AP24534 (Ponatinib) that targets FGFR, FLT-3, VEGFR-PDGFR and Bcr-Abl receptors; ABT-869 (Linifanib) that targets FLT-3 and VEGFR- PDGFR receptors; AZD2171 that targets VEGFR-PDGFR, Flt-1 and VEGF receptors; CHR-258 (Dovitinib) that targets VEGFR-PDGFR, FGFR, Flt- 3, and c-Kit receptors.
  • AP24534 Panatinib
  • ABT-869 Liifanib
  • AZD2171 that targets VEGFR-PDGFR, Flt-1 and VEGF receptors
  • CHR-258 Dovitinib
  • Exemplary kinase inhibitors include inhibitors of the kinases ATM, ATR, CHK1, CHK2, WEE1, and RSK.
  • Exemplary protein chaperone inhibitors include HSP90 inhibitors.
  • HSP90 inhibitors include Ganetespib, 17AAG derivatives, BIIB021, BIIB028, SNX-5422, NVP-AUY-922, and KW-2478.
  • HDAC inhibitors include Belinostat (PXD101), CUDC-101, Doxinostat, ITF2357 (Givinostat, Gavinostat), JNJ-26481585, LAQ824 (NVP- LAQ824, Dacinostat), LBH-589 (Panobinostat), MC1568, MGCD0103
  • Exemplary PARP inhibitors include neratinib (HKI-272), iniparib (BSI 201), olaparib (AZD-2281), ABT-888 (Veliparib), rucaparib (AG014699, CEP 9722, niraparib (MK-4827), KU-0059436 (AZD2281), talazoparib (BMN-673), 3- aminobenzamide, A-966492, E7016, BGB-290 and AZD2461
  • Exemplary Wnt/Hedgehog signaling pathway inhibitors include
  • vismodegib RG3616/GDC-0449
  • cyclopamine 11-deoxojervine
  • XAV-939 Wnt pathway inhibitor
  • Exemplary RNA polymerase inhibitors include amatoxins.
  • Exemplary amatoxins include a- amanitins, ⁇ - amanitins, ⁇ - amanitins, ⁇ -amanitins, amanullin, amanullic acid, amaninamide, amanin, and proamanullin.
  • Other amanitin compounds that can be used in the present invention include those described in, for example WO2014135282, WO2016142049, and EP2872479 the contents of each of which are incorporated herein by reference in their entirety.
  • Exemplary proteasome inhibitors include bortezomib, carfilzomib, ONX 0912, CEP- 18770, and MLN9708.
  • the drug of the invention is a non-natural camptothecin compound, vinca alkaloid, kinase inhibitor (e.g., PI3 kinase inhibitor (GDC-0941 and PI- 103)), MEK inhibitor, KSP inhibitor, RNA polymerse inhibitor, PARP inhibitor, docetaxel, paclitaxel, doxorubicin, duocarmycin, tubulysin, auristatin or a platinum compound.
  • kinase inhibitor e.g., PI3 kinase inhibitor (GDC-0941 and PI- 103)
  • MEK inhibitor e.g., PI3 kinase inhibitor (GDC-0941 and PI- 103)
  • MEK inhibitor e.g., PI3 kinase inhibitor (GDC-0941 and PI- 103)
  • MEK inhibitor e.g., PI3 kinase inhibitor (GDC-0941 and PI- 103)
  • KSP inhibitor
  • the drug is a derivative of SN-38, vindesine, vinblastine, PI- 103, AZD 8330, auristatin E, auristatin F, a duocarmycin compound, tubulysin compound, or ARRY-520.
  • the drug used in the invention is a combination of two or more drugs, such as, for example, PI3 kinases and MEK inhibitors; broad spectrum cytotoxic compounds and platinum compounds; PARP inhibitors and platinum compounds; broad spectrum cytotoxic compounds and PARP inhibitors.
  • the active agent can be a cancer therapeutic.
  • the cancer therapeutics may include death receptor agonists such as the TNF-related apoptosis-inducing ligand (TRAIL) or Fas ligand or any ligand or antibody that binds or activates a death receptor or otherwise induces apoptosis.
  • TRAIL TNF-related apoptosis-inducing ligand
  • Fas ligand any ligand or antibody that binds or activates a death receptor or otherwise induces apoptosis.
  • Suitable death receptors include, but are not limited to, TNFRl, Fas, DR3, DR4, DR5, DR6, LTpR and combinations thereof.
  • the active agent can be a DNA minor groove binders such as lurbectidin and trabectidin.
  • the active agent can be E3 ubiquitin ligase inhibitors, adeubiquitinase inhibitors or an NFkB pathway inhibitor.
  • the active agent can be a phopsphatase inhibitors including inhibitors of PTP1B, SHP2, LYP, FAP-1, CD45, STEP, MKP-1, PRL, LMWPTP or CDC25.
  • the active agent can be an inhibitor of tumor metabolism, such as an inhibitor of GAPDH, GLUT1, HK II, PFK, GAPDH, PK, LDH orMCTs
  • the active agent can target epigenetic targets including EZH2, MIX, DOT 1 -like protein (DOT1L), bromodomain-containing protein 4 (BRD4), BRD2, BRD3, NUT, ATAD2, or SMYD2.
  • EZH2, MIX DOT 1 -like protein
  • BRD4 bromodomain-containing protein 4
  • BRD2, BRD3, NUT ATAD2, or SMYD2.
  • the active agent can target the body's immune system to help fight cancer, including moecules targeting IDOl, ID02, TDO, CD39, CD73, A2A antagonists, STING activators, TLR agonists (TLR 1-13), ALK5, CBP/EP300 bromodomain, ARGl, ARG2, iNOS, PDE5, P2X7, P2Y11, COX2, EP2 Receptor, or EP4 receptor.
  • the active agent can target Bcl-2, IAP, or fatty acid synthase.
  • the active agent can be 20-epi-l,25
  • bicalutamide bisantrene, bisantrene hydrochloride, bisaziridinylspermine, bisnafide, bisnafide dimesylate, bistratene A, bizelesin, bleomycin, bleomycin sulfate, BRC/ ABL antagonists, breflate, brequinar sodium, bropirimine, budotitane, busulfan, buthionine sulfoximine, cabazitaxel, cactinomycin, calcipotriol, calphostin C, calusterone, camptothecin, camptothecin derivatives, canarypox IL-2, capecitabine, caracemide, carbetimer, carboplatin, carboxamide-amino-triazole,
  • carboxyamidotriazole carest M3, carmustine, earn 700, cartilage derived inhibitor, carubicin hydrochloride, carzelesin, casein kinase inhibitors, castano spermine, cecropin B, cedefingol, cetrorelix, chlorambucil, chlorins, chloroquinoxaline sulfonamide, cicaprost, cirolemycin, cisplatin, cis-porphyrin, cladribine, clomifene analogs, clotrimazole, collismycin A, collismycin B, combretastatin A4,
  • combretastatin analog conagenin, crambescidin 816, crisnatol, crisnatol mesylate, cryptophycin 8, cryptophycin A derivatives, curacin A, cyclopentanthraquinones, cyclophosphamide, cycloplatam, cypemycin, cytarabine, cytarabine ocfosfate, cytolytic factor, cytostatin, dacarbazine, dacliximab, dactinomycin, daunorubicin hydrochloride, decitabine, dehydrodidemnin B, deslorelin, dexifosfamide,
  • dexormaplatin dexrazoxane, dexverapamil, dezaguanine, dezaguanine mesylate, diaziquone, didemnin B, didox, diethylnorspermine, dihydro-5-azacytidine, dioxamycin, diphenyl spiromustine, docetaxel, docosanol, dolasetron, doxifluridine, doxorubicin, doxorubicin hydrochloride, droloxifene, droloxifene citrate,
  • hydrochloride elemene, elsamitrucin, emitefur, enloplatin, enpromate, epipropidine, epirubicin, epirubicin hydrochloride, epristeride, erbulozole, erythrocyte gene therapy vector system, esorubicin hydrochloride, estramustine, estramustine analog, estramustine phosphate sodium, estrogen agonists, estrogen antagonists, etanidazole, etoposide, etoposide phosphate, etoprine, exemestane, fadrozole, fadrozole hydrochloride, camrabine, fenretinide, filgrastim, finasteride, flavopiridol,
  • fluorodaunorunicin hydrochloride fluorouracil, flurocitabine, forfenimex, formestane, fosquidone, fostriecin, fostriecin sodium, fotemustine, gadolinium texaphyrin, gallium nitrate, galocitabine, ganirelix, gelatinase inhibitors, gemcitabine, gemcitabine hydrochloride, glutathione inhibitors, hepsulfam, heregulin, hexamethylene bisacetamide, hydroxyurea, hypericin, ibandronic acid, idarubicin, idarubicin hydrochloride, idoxifene, idramantone, ifosfamide, ilmofosine, ilomastat,
  • imidazoacridones imiquimod, immunostimulant peptides, insulin-like growth factor- 1 receptor inhibitor, interferon agonists, interferon alpha-2A, interferon alpha-2B, interferon alpha-Nl, interferon alpha-N3, interferon beta-IA, interferon gamma-IB, interferons, interleukins, iobenguane, iododoxorubicin, iproplatin, irinotecan, irinotecan hydrochloride, iroplact, irsogladine, isobengazole, isohomohalicondrin B, itasetron, jasplakinolide, kahalalide F, lamellarin-N triacetate, lanreotide, larotaxel, lanreotide acetate, leinamycin, lenograstim, lentinan sulfate, leptol
  • leuprolide/estrogen/progesterone leuprorelin, levamisole, liarozole, liarozole hydrochloride, linear polyamine analog, lipophilic disaccharide peptide, lipophilic platinum compounds, lissoclinamide 7, lobaplatin, lombricine, lometrexol, lometrexol sodium, lomustine, lonidamine, losoxantrone, losoxantrone hydrochloride, lovastatin, loxoribine, lurtotecan, lutetium texaphyrin, lysofylline, lytic peptides, maitansine, mannostatin A, marimastat, masoprocol, maspin, matrilysin inhibitors, matrix metalloproteinase inhibitors, maytansine, maytansinoid, mertansine (DM1), mechlorethamine hydrochloride, megestrol acetate,
  • modulators nitroxide antioxidant, nitrullyn, nocodazole, nogalamycin, n-substituted benzamides, 06-benzylguanine, octreotide, okicenone, oligonucleotides, onapristone, ondansetron, oracin, oral cytokine inducer, ormaplatin, osaterone, oxaliplatin, oxaunomycin, oxisuran, paclitaxel, paclitaxel analogs, paclitaxel derivatives, palauamine, palmitoylrhizoxin, pamidronic acid, panaxytriol, panomifene, parabactin, pazelliptine, pegaspargase, peldesine, peliomycin, pentamustine, pentosan polysulfate sodium, pentostatin, pentrozole, peplomycin sulfate, perflubron, perfosfamide
  • hydrochloride propyl bis-acridone, prostaglandin J2, prostatic carcinoma
  • the active agent can be an inorganic or organometallic compound containing one or more metal centers.
  • the compound contains one metal center.
  • the active agent can be, for example, a platinum compound, a ruthenium compound (e.g., trans-[KuC (DMSO)4], or tram , -[RuCl4(imidazole) 2, etc.), cobalt compound, copper compound, or iron compounds.
  • the active agent is a small molecule. In some embodiments, the active agent is a small molecule cytotoxin. In one embodiment, the active agent is cabazitaxel, or an analog, derivative, prodrug, or pharmaceutically acceptable salt thereof. In another embodiment, the active agent is mertansine (DM1) or DM4, or an analog, derivative, prodrug, or pharmaceutically acceptable salt thereof. DM1 or DM4 inhibits the assembly of microtubules by binding to tubulin. Structure of DM1 is shown below:
  • the active agent Z is Monomethyl auristatin E (MMAE), or an analog, derivative, prodrug, or pharmaceutically acceptable salt thereof. Structure of MMAE is shown below:
  • the active agent Z is a sequence-selective DNA minor-groove binding crosslinking agent.
  • Z may be
  • pyrrolobenzodiazepine PBD
  • PBD dimer a PBD dimer, or an analog, derivative, prodrug, or pharmaceutically acceptable salt thereof.
  • Pyrrol Strukturzodiazepines or pyrrolo[2, l- c][l, ⁇ benzodiazepines are a family of sequence-selective DNA minor-groove binding agents.
  • the first example of a PBD monomer is the natural product anthramycin.
  • Synthetic PBDs have been developed by attaching non-covalent minor- groove binding components to the C8-position of the PBD aromatic-ring. Monomeric PBD units have been joined together to afford PBD dimers.
  • An example of PBD dimer is SJG-136).
  • PBD analogs and dimers include, but not limited to, any PBD- based payload disclosed in Mantaj et al., Angew. Chem. Int. Ed, vol.56:462 (2017), the contents of which are incorporated herein by reference in their entirety, such as GWL-78, KMR-28-39, DSB-120, SJG-136 (also known as SG2000, NSC 694501 or BN2629) in Fig. 1 of Mantaj et al. Structures of PBD and a PBD dimer are shown below:
  • the active agent Z is a topoisom erase I inhibitor, such as camptothecin, irinotecan, SN-38, or an analog, derivative, prodrug, or pharmaceutically acceptable salt thereof.
  • WO2015184246 the contents of each of which are incorporated herein by reference in their entirety, such as bendamustine, VDA, doxorubicin, pemetrexed, vorinostat, lenalidomide, docetaxel, 17-AAG, 5-FU, abiraterone, crizotinib, KW-2189, BUMB2, DC1, CC-1065, adozelesin, or derivatives/analogs thereof, may be used as an active agent in conjugates of the present invention.
  • bendamustine, VDA, doxorubicin, pemetrexed, vorinostat, lenalidomide, docetaxel, 17-AAG, 5-FU, abiraterone, crizotinib, KW-2189, BUMB2, DC1, CC-1065, adozelesin, or derivatives/analogs thereof may be used as an active agent in conjugates of the present invention.
  • the active agent of the conjugate comprises a predetermined molar weight percentage from about 1% to about 10%, or about 10% to about 20%, or about 20% to about 30%, or about 30% to about 40%, or about 40% to about 50%, or about 50% to about 60%, or about 60% to about 70%, or about 70% to about 80%, or about 80% to about 90%, or about 90% to about 99% such that the sum of the molar weight percentages of the components of the conjugate is 100%.
  • the amount of active agent(s) of the conjugate may also be expressed in terms of proportion to the targeting ligand(s). For example, the present teachings provide a ratio of active agent to ligand of about 10: 1, 9: 1, 8: 1, 7: 1, 6: 1, 5: 1, 4: 1, 3 : 1, 2: 1, 1 : 1,
  • the conjugates contain one or more targeting moieties and/or targeting ligands.
  • Targeting ligands or moieties can be peptides, antibody mimetics, nucleic acids (e.g., aptamers), polypeptides (e.g., antibodies), glycoproteins, small molecules, carbohydrates, or lipids.
  • the targeting moiety, X can be a peptide such as
  • somatostatin e.g., somatostatin, octreotide, LHRH, an EGFR-binding peptide, RGD-containing peptides, a protein scaffold such as a fibronectin domain, an aptide or bipodal peptide, a single domain antibody, a stable scFv, or a bispecific T-cell engagers, nucleic acid (e.g., aptamer), polypeptide (e.g., antibody or its fragment), glycoprotein, small molecule, carbohydrate, or lipid.
  • nucleic acid e.g., aptamer
  • polypeptide e.g., antibody or its fragment
  • the targeting moiety, X can be an aptamer being either RNA or DNA or an artificial nucleic acid; small molecules; carbohydrates such as mannose, galactose and arabinose; vitamins such as ascorbic acid, niacin, pantothenic acid, carnitine, inositol, pyridoxal, lipoic acid, folic acid (folate), riboflavin, biotin, vitamin B12, vitamin A, E, and K; a protein or peptide that binds to a cell-surface receptor such as a receptor for thrombospondin, tumor necrosis factors (TNF), annexin V, interferons, cytokines, transferrin, GM-CSF (granulocyte-macrophage colony- stimulating factor), or growth factors such as vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), (platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), and epi
  • the targeting moiety is a protein scaffold.
  • the protein scaffold may be an antibody-derived protein scaffold.
  • Non-limiting examples include single domain antibody (dAbs), nanobody, single-chain variable fragment (scFv), antigen-binding fragment (Fab), Avibody, minibody, CH2D domain, Fcab, and bispecific T-cell engager (BiTE) molecules.
  • dAbs single domain antibody
  • scFv single-chain variable fragment
  • Fab antigen-binding fragment
  • Avibody minibody
  • CH2D domain CH2D domain
  • Fcab bispecific T-cell engager
  • BiTE bispecific T-cell engager
  • scFv is a stable scFv, wherein the scFv has hyperstable properties.
  • the nanobody may be derived from the single variable domain (VHH) of camelidae antibody.
  • the protein scaffold may be a nonantibody-derived protein scaffold, wherein the protein scaffold is based on nonantibody binding proteins.
  • the protein scaffold may be based on enginnered Kunitz domains of human serine protease inhibitors (e.g., LAC1-D1), DARPins (designed ankyrin repeat domains), avimers created from multimerized low-density lipoprotein receptor class A (LDLR-A), anticalins derived from lipocalins, knottins constructed from cysteine-rich knottin peptides, affibodies that are based on the Z-domain of staphylococcal protein A, adnectins or monobodies and pronectins based on the 10 th or 14 th extracellular domain of human fibronectin III, Fynomers derived from SH3 domains of human Fyn tyrosine kinase, or nanofitins (formerly Affitins) derived from the DNA
  • the protein scaffold may be any protein scaffold disclosed in Mintz and Crea, BioProcess, vol.11(2):40-48 (2013), the contents of which are incorporated herein by reference in their entirety. Any of the protein scaffolds disclosed in Tables 2-4 of Mintz and Crea may be used as a targeting moiety of the conjugate of the invention.
  • the protein scaffold may be based on a fibronectin domain.
  • the protein scaffold may be based on fibronectin type III (FN3) repeat protein.
  • the protein scaffold may be based on a consensus sequence of multiple FN3 domains from human Tenascin-C (hereinafter "Tenascin"). Any protein scaffold based on a fibronectin domain disclosed in US Pat. No. 8569227 to Jacobs et al., the contents of which are incorporated herein by reference in their entirety, may be used as a targeting moiety of the conjugate of the invention.
  • the targeting moiety or targeting ligand may be any molecule that can bind to luteinizing-hormone-releasing hormone receptor (LITRFIR).
  • LITRFIR luteinizing-hormone-releasing hormone receptor
  • Such targeting ligands can be peptides, antibody mimetics, nucleic acids (e.g., aptamers), polypeptides (e.g., antibodies), glycoproteins, small molecules, carbohydrates, or lipids.
  • the targeting moiety is LHRH or a LHRH analog.
  • Luteinizing-hormone-releasing hormone also known as gonadotropin-releasing hormone (GnRH) controls the pituitary release of
  • LHRH gonadotropins
  • FSH gonadotropins
  • LHRH is a 10-amino acid peptide that belongs to the gonadotropin-releasing hormone class. Signaling by LHRH is involved in the first step of the hypothalami c- pituitary-gonadal axis.
  • An approach in the treatment of hormone-sensitive tumors directed to the use of agonists and antagonists of LHRH (A.V. Schally and A.M. Comaru-Schally. Sem. Endocrinol., 5 389-398, 1987) has been reported.
  • Some LHRH agonists, when substituted in position 6, 10, or both are much more active than LHRH and also possess prolonged activity.
  • Some LHRH agonists are approved for clinical use, e.g., Leuprolide, triptorelin, nafarelin and goserelin.
  • Some human tumors are hormone dependent or hormone-responsive and contain hormone receptors. Certain of these tumors are dependent on or responsive to sex hormones or growth factors, or have components that are dependent or responsive to such hormones. Mammary carcinomas contain estrogen, progesterone,
  • glucocorticoid LHRH
  • EGF IGF-I somatostatin receptors.
  • Peptide hormone receptors have been detected in acute leukaemia, prostate-, breast-, pancreatic, ovarian-, endometrial cancer, colon cancer and brain tumors (M.N. Pollak, et al., Cancer Lett. 38 223-230 1987; F. Pekonen, et al., Cancer Res., 48 1343-1347, 1988; M. Fekete, et al., J Clin.Lab. Anal. 3 137-147, 1989; G. Emons, et al., Eur. J. Cancer Oncol., 25215-221 1989). It has been found (M.N. Pollak, et al., Cancer Lett. 38 223-230 1987; F. Pekonen, et al., Cancer Res., 48 1343-1347, 1988; M. Fekete, et al., J Clin.Lab. Anal. 3 137-147,
  • the conjugates of the invention can employ any of the large number of known molecules that recognize the LHRH receptor, such as known LHRH receptor agonists and antagonists.
  • the LHRH analog portion of the conjugate contains between 8 and 18 amino acids.
  • LHRH binding molecules useful in the present invention are described herein. Further non-limiting examples are analogs of pyroGlu-His-Trp-Ser- Tyr-Gly-Leu-Arg-Pro-Gly-NH2, leuprolide, triptorelin, nafarelin, buserelin, goserelin, cetrorelix, ganirelix, azaline-B, degarelix and abarelix.
  • a tumor expressing a LHRH receptor includes a neoplasm of the lung, breast, prostate, colon, brain, gastrointestinal tract, neuroendocrine axis, liver, or kidney (see Schaer et al., Int. J. Cancer, 70:530-537, 1997; Chave et al., Br. J. Cancer 82(1): 124-130, 2000; Evans et al., Br. J. Cancer 75(6):798-803, 1997).
  • the targeting moiety e.g., LHRH analog
  • the targeting moiety used in the invention is hydrophilic, and is therefore water soluble.
  • such targeted constructs are used in treatment paradigms in which this feature is useful, e.g., compared to conjugates comprising hydrophobic analogs.
  • Hydrophilic analogs described herein can be soluble in blood, cerebrospinal fluid, and other bodily fluids, as well as in urine, which may facilitate excretion by the kidneys. This feature can be useful, e.g., in the case of a composition that would otherwise exhibit undesirable liver toxicity.
  • the invention also discloses specific hydrophilic elements (e.g., incorporation of a PEG linker, and other examples in the art) for incorporation into peptide analogs, allowing modulation of the analog's hydrophilicity to adjust for the chemical and structural nature of the various conjugated cytotoxic agents.
  • specific hydrophilic elements e.g., incorporation of a PEG linker, and other examples in the art
  • AnticalinTM an avimers (avidity multimers), a DARPinTM, a FynomerTM, CentyrinTM, a Humabody®, Kunitz domain or an Abdurin peptide.
  • mimetics are artificial peptides or proteins with a molar mass of about 3 to 20 kDa. Nucleic acids and small molecules may be antibody mimetic.
  • the targeting moiety is an Abdurin peptide. It is an engineered antibody domain molecule comprising at least one protein-binding domain derived from a CH2 domain or CH2-like domain of an immunoglobulin (such as IgG, IgA, IgD), or a CH3 domain or CH3-like domain of IgE or IgM, comprising at least one mutation.
  • the mutation may be an N-terminal truncation of at least one amino acid and/or a C-terminal truncation of at least one amino acid.
  • the molecular weight of an Abdurin peptide is usually less than about 15 kD.
  • a targeting moiety can be an aptamer, which is generally an oligonucleotide (e.g., DNA, RNA, or an analog or derivative thereof) that binds to a particular target, such as a polypeptide.
  • the targeting moiety is a polypeptide (e.g., an antibody that can specifically bind a tumor marker).
  • the targeting moiety is an antibody or a fragment thereof.
  • the targeting moiety is an Fc fragment of an antibody.
  • a targeting moiety may be a non-immunoreactive ligand.
  • the non-immunoreactive ligand may be insulin, insulin-like growth factors I and II, lectins, apoprotein from low density lipoprotein, etc. as disclosed in US 20140031535 to Jeffrey, the contents of which are incorporated herein by reference in their entirety.
  • Any protein or peptide comprising a lectin disclosed in WO2013181454 to Radin, the contents of which are incorporated herein by reference in their entirety, may be used as a targeting moiety.
  • the conjugate of the invention may target a hepatocyte intracellularly and a hepatic ligand may be used as a targeting moiety.
  • a hepatic ligand disclosed in US 20030119724 to Ts'o et al., the contents of which are incorporated herein by reference in their entirety, such as the ligands in Fig. 1, may be used.
  • the hepatic ligand specifically binds to a hepatic receptor, thereby directing the conjugate into cells having the hepatic receptor.
  • a targeting moiety may interact with a protein that is overexpressed in tumor cells compared to normal cells.
  • the targeting moiety may bind to a chaperonin protein, such as Hsp90, as disclosed in US 20140079636 to Chimmanamada et al., the contents of which are incorporated herein by reference in their entirety.
  • the targeting moiety may be an Hsp90 inhibitor, such as ganetespib or a ganetespib analog (e.g., 4-(5-hydroxy-4-(l-(2-(piperidin-4-yl)ethyl)-lH-indol-5-yl)- 4H- 1 ,2,4-triazol-3 -yl)-6-isopropylbenzene- 1 ,3 -diol), geldanamycins, macbecins, tripterins, tanespimycins, and radicicols.
  • ganetespib or a ganetespib analog (e.g., 4-(5-hydroxy-4-(l-(2-(piperidin-4-yl)ethyl)-lH-indol-5-yl)- 4H- 1 ,2,4-triazol-3 -yl)-6-isopropylbenzene- 1 ,3 -diol), geldana
  • the targeted construct may have a terminal half-life of longer than about 72 hours and a targeting moiety may be selected from Table 1 or 2 of US 20130165389 to Schellenberger et al., the contents of which are incorporated herein by reference in their entirety.
  • the targeting moiety may be an antibody targeting delta-like protein 3 (DLL3) in disease tissues such as lung cancer, pancreatic cancer, skin cancer, etc., as disclosed in WO2014125273 to Hudson, the contents of which are incorporated herein by reference in their entirety.
  • the targeting moiety may also any targeting moiety in WO2007137170 to Smith, the contents of which are incorporated herein by reference in their entirety.
  • the targeting moiety binds to glypican-3 (GPC-3) and directs the conjugate to cells expressing GPC-3, such as hepatocellular carcinoma cells.
  • a target of the targeting moiety may be a marker that is exclusively or primarily associated with a target cell, or one or more tissue types, with one or more cell types, with one or more diseases, and/or with one or more developmental stages.
  • a target can comprise a protein (e.g., a cell surface receptor, transmembrane protein, glycoprotein, etc.), a carbohydrate (e.g., a glycan moiety, glycocalyx, etc.), a lipid (e.g., steroid, phospholipid, etc.), and/or a nucleic acid (e.g., a DNA, RNA, etc.).
  • targeting moieties may be peptides for regulating cellular activity.
  • the targeting moiety may bind to Toll Like Receptor (TLR).
  • TLR Toll Like Receptor
  • It may be a peptide derived from vaccinia virus A52R protein such as a peptide comprising SEQ ID No. 13 as disclosed in US 7557086, a peptide comprising SEQ ID No. 7 as disclosed in US 8071553 to Hefeneider, et al., or any TLR binding peptide disclosed in WO 2010141845 to McCoy, et al., the contents of each of which are incorporated herein by reference in their entirety.
  • the A52R derived synthetic peptide may significantly inhibit cytokine production in response to both bacterial and viral pathogen associated molecular patterns, and may have application in the treatment of inflammatory conditions that result from ongoing toil -like receptor activation.
  • targeting moieties many be amino acid sequences or single domain antibody fragments for the treatment of cancers and/or tumors.
  • targeting moieties may be an amino acid sequence that binds to Epidermal Growth Factor Receptor 2 (HER2).
  • HER2 Epidermal Growth Factor Receptor 2
  • Targeting moieties may be any HER2 -binding amino acid sequence described in US 20110059090, US8217140, and US 8975382 to Revets, et al., the contents of each of which are incorporated herein by reference in their entirety.
  • the targeting moiety may be a domain antibody, a single domain antibody, a VHH, a humanized VHH or a camelized VH.
  • targeting moieties may be peptidomimetic macrocycles for the treatment of disease.
  • targeting moieties may be peptidomimetic macrocycles that bind to the growth hormone-releasing hormone (GHRH) receptor, such as a peptidomimetic macrocycle comprising an amino acid sequence which is at least about 60% identical to GHRH 1-29 and at least two macrocycle-forming linkers as described in US20130123169 to Kawahata et al, the contents of which are incorporated herein by reference in their entirety.
  • GHRH growth hormone-releasing hormone
  • the peptidomimetic macrocycle targeting moiety may be prepared by introducing a cross-linker between two amino acid residues of a polypeptide as described in US 20120149648 and US 20130072439 to Nash et al., the contents of each of which are incorporated herein by reference in their entirety. Nash et al.
  • the peptidomimetic macrocycle may comprise a peptide sequence that is derived from the BCL-2 family of proteins such as a BH3 domain.
  • peptidomimetic macrocycle may comprise a BID, BAD, BIM, BIK, NOXA, PUMA peptides.
  • targeting moieties may be polypeptide analogues for transport to cells.
  • the polypeptide may be an Angiopep-2
  • polypeptide analog may comprising a polypeptide comprising an amino acid sequence at least 80% identical to SEQ ID No.97 as described in US 20120122798 to Castaigne et al., the contents of which are incorporated herein by reference in their entirety.
  • polypeptides may transport to cells, such as liver, lung, kidney, spleen, and muscle, such as Angiopep-4b, Angiopep-5, Angiopep-6, and Angiopep-7 polypeptide as described in EP 2789628 to Beliveau et al., the contents of each of which are incorporated herein by reference in their entirety.
  • targeting moieties may be homing peptides to target liver cells in vivo.
  • the melittin delivery peptides that are administered with RNAi polynucleotides as described in US 8501930 Rozema, et al., the contents of which are incorporated herein by reference in their entirety may be used as targeting moieties.
  • delivery polymers provide membrane penetration function for movement of the RNAi polynucleotides from the outside the cell to inside the cell as described in US 8313772 to Rozema et al., the contents of each of which are incorporated herein by reference in their entirety. Any delivery peptide disclosed by Rozema et al. may be used as targeting moeities.
  • targeting moieties may be structured polypeptides to target and bind proteins.
  • polypeptides with sarcosine polymer linkers that increase the solubility of structured polypeptides as described in WO
  • polypeptide with variable binding activity produced by the methods described in WO 2014140342 to Stace, et al., the contents of which are incorporated herein by reference in their entirety.
  • the polypeptides may be evaluated for the desired binding activity.
  • modifications of the targeting moieties affect a compound's ability to distribute into tissues.
  • a structure activity relationship analysis was completed on a low orally bioavailable cyclic peptide and the permeability and clearance was determined as described in Rand, AC, et al., Medchemcomm. 2012, 3(10): 1282-1289, the contents of which are incorporated herein by reference in their entirety.
  • Any of the cyclic peptide disclosed by Rand et al., such as N-methylated cyclic hexapeptides, may be used as targeting moieties.
  • targeting moieties may be a polypeptide which is capable of internalization into a cell.
  • targeting moieties may be an Alphabody capable of internalization into a cell and specifically binding to an intracellular target molecule as described in US 20140363434 to Lasters, et al., the contents of which are incorporated herein by reference in their entirety.
  • an 'Alphabody' or an 'Alphabody structure' is a self-folded, single- chain, triple- stranded, predominantly alpha-helical, coiled coil amino acid sequence, polypeptide or protein.
  • the Alphabody may be a parallel Alphabody or an anti- parallel Alphabody.
  • targeting moieties may be any Alphabody in the single-chain Alphabody library used for the screening for and/or selection of one or more Aiphabodies that specifically bind to a target molecule of interest as described in WO 2012092970 to Desmet et al., the contents of which are incorporated herein by reference in their entirety.
  • targeting moieties may consist of an affinity- matured heavy chain-only antibody.
  • targeting moieties may be any VH heavy chain-only antibodies produced in a transgenic non-human mammal as described in US 20090307787 to Grosveld et al., the contents of which are incorporated herein by reference in their entirety.
  • targeting moieties may bind to the hepatocyte growth factor receptor "HGFr" or "cMet".
  • targeting moieties may be a polypeptide moiety that is conjugated to a detectable label for diagnostic detection of cMet as described in US 9000124 to Dransfield et al., the contents of which are incorporated herein by reference in their entirety.
  • targeting moieties may bind to human plasma kallikrein and may comprise BPTI-homologous Kunitz domains, especially LACI homologues, to bind to one or more plasma (and/or tissue) kallikreins as described in WO 1995021601 to Markland et al., the contents of which are incorporated herein by reference in their entirety.
  • targeting moieties are evolved from weak binders and anchor-scaffold conjugates having improved target binding and other desired pharmaceutical properties through control of both synthetic input and selection criteria.
  • targeting moieties may be macrocyclic compounds that bind to inhibitors of apoptosis as described in WO 2014074665 to Borzilleri et al., the contents of which are incorporated herein by reference in their entirety.
  • targeting moieties may comprise pre-peptides that encode a chimeric or mutant lantibiotic.
  • targeting moieties may be pre- tide that encode a chimera that was accurately and efficiently converted to the mature lantibiotic, as demonstrated by a variety of physical and biological activity assays as described in US5861275 to Hansen, the contents of which are incorporated herein by- reference in their entirety. The mixture did contain an active minor component with a biological activity.
  • targeting moieties may comprise a leader peptide of a recombinant manganese superoxide dismutase (rMnSOD-Lp).
  • rMnSOD-Lp which delivers cisplatin directly into tumor cells as described in Borrelli, A., et al., Chem Biol Drug Des. 2012, 80(1):9-16, the contents of which are incorporated herein by reference in their entirety, may be used a targeting moiety.
  • the targeting moiety may be an antibody for the treatment of glioma.
  • an antibody or antigen binding fragment which specifically binds to JAMM-B or JAM-C as described in US8007797 to Dietrich et al., the contents of which are incorporated herein by reference in their entirety, may be used as a targeting moiety
  • JAMs are a family of proteins belonging to a class of adhesion molecules generally localized at sites of cell-cell contacts in tight junctions, the specialized cellular structures that keep ceil polarity and serve as barriers to prevent the diffusion of molecules across intercellular spaces and along the basolateral-apical regions of the plasma membrane.
  • the targeting moiety may be a target interacting modulator.
  • nucleic acid molecules capable of interacting with proteins associated with the Human Hepatitis C virus or corresponding peptides or mimetics capable of interfering with the interaction of the native protein with the HIV accessor ⁇ ' protein as described in WO 2011015379 and US 8685652, the contents of each of which are incorporated herein by reference in their entirety, may be used as a targeting moiety.
  • the targeting moiety may bind with biomolecules.
  • biomolecules for example, any cystine-knot family small molecule polycyclic molecular scaffolds were designed as peptidomsmetics of FSH and used as peptide-vaccine as described in US7863239 to Timmerman, the contents the contents of which are incorporated herein by reference in their entirety, may be used as targeting moieties.
  • the targeting moiety may bind to integrin and thereby block or inhibit integrin binding.
  • any highly selective disulfi.de- rich dimer molecules which inhibit binding of a4B7 to the mucosal addressin cell adhesion molecule (MAdCAM) as described in WO 2014059213 to Bhandari, the contents of which are incorporated herein by reference in their entirety, may be used as a targeting moiety.
  • MAdCAM mucosal addressin cell adhesion molecule
  • Any inhibitor of specific integrins-ligand interactions may be used as a targeting moiety.
  • the conjugates comprising such target moieties may be effective as anti -inflammatory agents for the treatment of various autoimmune diseases.
  • the targeting moiety may comprise novel peptides.
  • any cyclic peptide or mimetic that is a serine protease inhibitor as described in WO 2013172954 to Wang et al., the contents of which are incorporated herein by reference in their entirety, may be used as a targeting moiety.
  • targeting moieties may comprise a targeting peptide that is used in the reduction of cell proliferation and the treatment of cancer.
  • a peptide composition inhibiting the trpv6 calcium channel as described in US 20120316119 to Stewart, the contents of which are incorporated herein by reference in their entirety, may be used as a targeting moiety.
  • the targeting moiety may comprise a cyclic peptide.
  • any cyclic peptides exhibit various types of action in vivo, as described in US20100168380 and WO 2008117833 to Suga et al., and WO
  • Such cyclic peptide targeting moieties have a stabilized secondary structure and may inhibit biological molecule interactions, increase cell membrane permeability and the peptide's half-life in blood serum.
  • the targeting moiety may consist of a therapeutic peptide.
  • peptide targeting moieties may be an AP-1 signaling inhibitor, such as a peptide analog comprising SEQ ID No. 104 of US8946381B2 to Fear that is used for the treatment of wounds, a peptide comprising SEQ ID No. 108 in
  • ARDS acute respiratory distress syndrome
  • the targeting moiety may be any biological modulator isolated from biodiverse gene fragment libraries as described in US7803765 and EP 1754052 to Watt, any inhibitor of c-Jun dimerization as described in EP 1601766 and EP 1793841 to Watt, any peptide inhibitors of CD40L signaling as described in US8802634 and US20130266605 to Watt, or any peptide modulators of cellular phenotype as described in US20110218118 to Watt, the contents of each of which are incorporated herein by reference in their entirety.
  • the targeting moiety may consist of a characterized peptide.
  • targeting moieties may be cell-penetrating peptides.
  • any cell-penetrating peptides linked to a cargo that are capable of passing through the blood brain barrier as described by US20140141452A1 to Watt, et al., the contents of which are incorporated herein by reference, may be used a targeting moiety.
  • the targeting moiety may comprise a LHRH antagonist, agonist, or analog.
  • the targeting moiety may be Cetrorelix, a decapeptide with a terminal acid amide group (AC-D-Nal(2)-D-pCl-Phe-D-Pal(3)- Ser-Tyr-D-Cit-Leu-Arg-Pro-D-Ala-NH2) as described in US 4800191, US 6716817, US 6828415, US 6867191, US 7605121, US 7718599, US 7696149 (Zentaris Ag), or pharmaceutically active decapeptides such as SB-030, SB-075 (cetrorelix) and SB- 088 disclosed in EP 0 299 402 (Asta Pharma), the contents of each of which are incorporated herein by reference in their entirety.
  • the targeting moiety may be LHRH analogues such as D-/L-Mel (4-[bis(2-chloroethyl)amino]-D/L- phenylalanine), cyclopropanealkanoyl, aziridine-2-carbonyl, epoxyalkyl, 1,4- naphthoquinone-5-oxycarbonyl-ethyl, doxorubicinyl (Doxorubicin, DOX), mitomicinyl (Mitomycin C), esperamycinyl or methotrexoyl, as disclosed in US 6214969 to Janaky et al., the contents of which are incorporated herein by reference in their entirety.
  • LHRH analogues such as D-/L-Mel (4-[bis(2-chloroethyl)amino]-D/L- phenylalanine), cyclopropanealkanoyl, aziridine-2-carbonyl, epoxyalky
  • the targeting moiety may be any cell-binding molecule disclosed in US 7741277 or US 7741277 to Guenther et al. (Aeterna Zentaris), the contents of which are incorporated herein by reference in their entirety, such as octamer peptide, nonamer peptide, decamer peptide, luteinizing hormone releasing hormone (LHRH), [D-Lys6]-LHRH, LHRH analogue, LHRH agonist, Triptorelin ([ ⁇ - ⁇ 6]- ⁇ ⁇ ), LHRH antagonist, bombesin, bombesin analogue, bombesin antagonist, somatostatin, somatostatin analogue, serum albumin, human serum albumin (HSA).
  • LHRH luteinizing hormone releasing hormone
  • LHRH luteinizing hormone releasing hormone
  • [D-Lys6]-LHRH LHRH analogue
  • LHRH agonist Triptorelin
  • Triptorelin
  • targeting moieties may bind to growth hormone secretagogue (GHS) receptors, including ghrelin analogue ligands of GHS receptors.
  • GHS growth hormone secretagogue
  • targeting moieties may be any triazole derivatives with improved receptor activity and bioavailability properties as ghrelin analogue ligands of growth hormone secretagogue receptors as describe by US8546435 to Aicher, at al. (Aeterna Zentaris), the contents of which are incorporated herein by reference in their entirety.
  • the targeting moiety X is an aptide or bipodal peptide.
  • X may be any D-Aptamer-Like Peptide (D-Aptide) or retro-inverso Aptide which specifically binds to a target comprising: (a) a structure stabilizing region comprising parallel, antiparallel or parallel and antiparallel D-amino acid strands with interstrand noncovalent bonds; and (b) a target binding region I and a target binding region II comprising randomly selected n and m D-amino acids, respectively, and coupled to both ends of the structure stabilizing region, as disclosed in US Pat.
  • D-Aptide D-Aptide
  • retro-inverso Aptide which specifically binds to a target comprising: (a) a structure stabilizing region comprising parallel, antiparallel or parallel and antiparallel D-amino acid strands with interstrand noncovalent bonds; and (b) a target binding region I and a target binding
  • X may be any bipodal peptide binder (BPB) comprising a structure stabilizing region of parallel or antiparallel amino acid strands or a combination of these strands to induce interstrand non-covalent bonds, and target binding regions I and II, each binding to each of both termini of the structure stabilizing region, as disclosed in US Pat. Application No. 20120321697 to Jon et al., the contents of which are incorporated herein by reference in their entirety.
  • X may be an intracellular targeting bipodal-peptide binder specifically binding to an
  • intracellular target molecule comprising: (a) a structure-stabilizing region comprising a parallel amino acid strand, an antiparallel amino acid strand or parallel and antiparallel amino acid strands to induce interstrand non-covalent bonds; (b) target binding regions I and II each binding to each of both termini of the structure- stabilizing region, wherein the number of amino acid residues of the target binding region I is n and the number of amino acid residues of the target binding region II is m; and (c) a cell-penetrating peptide (CPP) linked to the structure-stabilizing region, the target binding region I or the target binding region II, as disclosed in US Pat. Application No.
  • CPP cell-penetrating peptide
  • X may be any bipodal peptide binder comprising a ⁇ -hairpin motif or a leucine-zipper motif as a structure stabilizing region comprising two parallel amino acid strands or two antiparallel amino acid strands, and a target binding region I linked to one terminus of the first of the strands of the structure stabilizing region, and a target binding region II linked to the terminus of the second of the strands of the structure stabilizing region, as disclosed in US Pat. Application No. 20110152500 to Jon et al., the contents of which are incorporated herein by reference in their entirety.
  • X may be any bipodal peptide binder targeting KPI as disclosed in WO2014017743 to Jon et al, any bipodal peptide binder targeting cytokine as disclosed in WO2011132939 to Jon et al., any bipodal peptide binder targeting transcription factor as disclosed in WO201132941 to Jon et al., any bipodal peptide binder targeting G protein-coupled receptor as disclosed in WO2011132938 to Jon et al., any bipodal peptide binder targeting receptor tyrosine kinase as disclosed in WO2011132940 to Jon et al., the contents of each of which are incorporated herein by reference in their entireties.
  • X may also be bipodal peptide binders targeting cluster differentiation (CD7) or an ion channel.
  • the targeting moiety may be a bicyclic peptide or a modified bicyclic peptide, as disclosed in WO2015063465, EP2464727,
  • the target, target cell or marker is a molecule that is present exclusively or predominantly on the surface of malignant cells, e.g., a tumor antigen.
  • a marker is a prostate cancer marker.
  • the target can be an intra-cellular protein.
  • a marker is a breast cancer marker, a colon cancer marker, a rectal cancer marker, a lung cancer marker, a pancreatic cancer marker, a ovarian cancer marker, a bone cancer marker, a renal cancer marker, a liver cancer marker, a neurological cancer marker, a gastric cancer marker, a testicular cancer marker, a head and neck cancer marker, an esophageal cancer marker, or a cervical cancer marker.
  • the targeting moiety directs the conjugates to specific tissues, cells, or locations in a cell.
  • the target can direct the conjugate in culture or in a whole organism, or both.
  • the targeting moiety binds to a receptor that is present on the surface of or within the targeted cell(s), wherein the targeting moiety binds to the receptor with an effective specificity, affinity and avidity.
  • the targeting moiety targets the conjugate to a specific tissue such as the liver, kidney, lung or pancreas.
  • the targeting moiety can target the conjugate to a target cell such as a cancer cell, such as a receptor expressed on a cell such as a cancer cell, a matrix tissue, or a protein associated with cancer such as tumor antigen.
  • cells comprising the tumor vasculature may be targeted.
  • Targeting moieties can direct the conjugate to specific types of cells such as specific targeting to hepatocytes in the liver as opposed to Kupffer cells.
  • targeting moieties can direct the conjugate to cells of the reticular endothelial or lymphatic system, or to professional phagocytic cells such as macrophages or eosinophils.
  • the target is member of a class of proteins such as receptor tyrosine kinases (RTK) including the following RTK classes: RTK class I (EGF receptor family) (ErbB family), RTK class II (Insulin receptor family), RTK class III (PDGF receptor family), RTK class IV (FGF receptor family), RTK class V (VEGF receptors family), RTK class VI (HGF receptor family), RTK class VII (Trk receptor family), RTK class VIII (Eph receptor family), RTK class IX (AXL receptor family), RTK class X (LTK receptor family), RTK class XI (TIE receptor family), RTK class XII (ROR receptor family), RTK class XIII (DDR receptor family), RTK class XIV (RET receptor family), RTK class XV (KLG receptor family), RTK class XVI (RYK receptor family) and RTK class XVII (MuSK receptor family).
  • RTK class I EGF receptor family
  • ErbB family ErbB family
  • the target is a serine or threonine kinase, G-protein coupled receptor, methyl CpG binding protein, cell surface glycoprotein, cancer stem cell antigen or marker, carbonic anhydrase, cytolytic T lymphocyte antigen, DNA methyltransferase, an ectoenzyme, a glycosylphosphatidylinositol-anchored co- receptor, a gly pi can-related integral membrane proteoglycan, a heat shock protein, a hypoxia induced protein, a multi drug resistant transporter, a Tumor-associated macrophage marker, a tumor associated carbohydrate antigen, a TNF receptor family member, a transmembrane protein, a tumor necrosis factor receptor superfamily member, a tumour differentiation antigen, a zinc dependent metallo-exopeptidase, a zinc transporter, a sodium-dependent transmembrane transport protein, a member of the SIGLEC family of lectins
  • HER-2 HER-2
  • HER-3 EGFR
  • NTSR1 neurotensin receptors
  • the targeting moiety binds a target such as CD 19, CD70, CD56, PSMA, alpha integrin, CD22, CD 138, EphA2, AGS-5, Nectin-4, HER2, GPMNB, CD74 and Le.
  • the target is a protein listed in Table A.
  • the targeting moiety or moieties of the conjugate are present at a predetermined molar weight percentage from about 1% to about 10%, or about 10% to about 20%, or about 20% to about 30%, or about 30% to about 40%, or about 40% to about 50%, or about 50% to about 60%, or about 60% to about 70%, or about 70% to about 80%, or about 80% to about 90%, or about 90% to about 99% such that the sum of the molar weight percentages of the components of the conjugate is 100%.
  • the amount of targeting moieties of the conjugate may also be expressed in terms of proportion to the active agent(s), for example, in a ratio of ligand to active agent of about 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4; 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10.
  • SSTR-binding targeting moieties may also be expressed in terms of proportion to the active agent(s), for example, in a ratio of ligand to active agent of about 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4; 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10.
  • targeting ligands include any molecule that can bind one or more SSTRs, e.g., human SSTR1, SSTR2, SSTR3, SSTR4, or SSTR5.
  • SSTRs e.g., human SSTR1, SSTR2, SSTR3, SSTR4, or SSTR5.
  • Such targeting ligands can be peptides, antibody mimetics, nucleic acids (e.g., aptamers), polypeptides (e.g., antibodies), glycoproteins, small molecules, carbohydrates, or lipids.
  • the targeting moiety is somatostatin or a somatostation analog.
  • the conjugates of the invention can employ any somatostatin analog that binds somatostatin receptor.
  • the somatostatin analog portion of the conjugate contains between 8 and 18 amino acids, and includes the core sequence: cyclo[Cys-Phe-D-Trp-Lys-Thr-Cys] (SEQ ID NO: l) or cyclo[Cys-Tyr-D-Trp-Lys- Thr-Cys] (SEQ ID NO. 2).
  • the C-terminus of the analog is Thr-NH2.
  • the targeting moiety binds preferably to SSTR2.
  • the conjugate comprising the targeting moiety binds preferably to SSTR2.
  • the binding of the conjugate to SSTR2 is stronger than the binding of the conjugate to SSTR1, SSTR3, SSTR4 or SSTR5.
  • the conjugates as described herein have low membrane permeability.
  • Membrane permeability may be low in both the apical to basolateral direction and the basolateral to apical direction. Not wiling to be bound by any theory, low membrane permeability enhances selective uptake by SSTRs by decreasing non-specific permeability. Low permeability leads to decreased uptake in cells that do not express SSTR2, leading to lower toxicity to non-SSTR2 expressing cells.
  • Membrane permeability may be determined by any method known in the art. For example, it may be determined by measuring apparent permeability (Papp) in Caco-2 monolayers.
  • the targeting moiety, X may be selected from somatostatin, octreotide, octreotate, lanreotide, lutathera ( 177 Lu-DOTATATE), 90 Y- DOTATOC, Tyr 3 -octreotate (TATE), vapreotide, cyclo(AA-Tyr-DTrp-Lys-Thr-Phe) where AA is ⁇ - ⁇ -Me lysine or N-Me glutamic acid, pasireotide, lanreotide, seglitide, or any other example of somatostatin receptor binding ligands.
  • the targeting moiety is a somatostatin receptor binding moiety that binds to somatostatin receptors 2 and/or 5.
  • X binds to the linker moiety Y at the C-terminal.
  • X binds to the linker moiety Y at the N- terminal.
  • the targeting moiety X comprises at least one D-Phe residue and the phenyl ring of the D-Phe residue of the targeting moiety X has been replaced by a linker-containing moiety.
  • somatostatin peptides and analogs are well documented and are within the ability of a person of ordinary skill in the art as exemplified in the references listed supra. Further synthetic procedures are provided in the following examples. The following examples also illustrate methods for synthesizing the targeted cytotoxic compounds of the present invention. Specific targeting of therapeutic or cytotoxic agents allows selective destruction of a tumor expressing a receptor specific for a biologically active peptide.
  • a tumor expressing a somatostatin receptor includes a neoplasm of the lung, breast, prostate, colon, brain, gastrointestinal tract, neuroendocrine axis, liver, or kidney (see Schaer et al., Int. J. Cancer, 70:530-537, 1997; Chave et al., Br. J. Cancer 82(1): 124-130, 2000; Evans et al., Br. J. Cancer 75(6):798-803, 1997).
  • the targeting moiety has therapeutic features, e.g., the targeting moiety is cytotoxic or anti -angiogenic.
  • a targeting moiety has some increased affinity for tumor vasculature, or angiogenic blood vessels, e.g., those that over-express somatostatin receptors (see Denzler and Reubi, Cancer 85: 188-198, 1999; Gulec et al., J. Surg. Res. 97(2): 131-137, 2001; Woltering et al., J. Surg. Res. 50:245, 1991).
  • the targeting moiety e.g., somatostatin analog
  • the targeting moiety used in the invention is hydrophilic, and is therefore water soluble.
  • such conjugates and particles containing such conjugates are used in treatment paradigms in which this feature is useful, e.g., compared to conjugates comprising hydrophobic analogs.
  • Hydrophilic analogs described herein can be soluble in blood, cerebrospinal fluid, and other bodily fluids, as well as in urine, which may facilitate excretion by the kidneys. This feature can be useful, e.g., in the case of a composition that would otherwise exhibit undesirable liver toxicity.
  • the invention also discloses specific hydrophilic elements (e.g., incorporation of a PEG linker, and other examples in the art) for incorporation into peptide analogs, allowing modulation of the analog's hydrophilicity to adjust for the chemical and structural nature of the various conjugated cytotoxic agents, e.g., conjugate 6 infra.
  • specific hydrophilic elements e.g., incorporation of a PEG linker, and other examples in the art
  • Neurotensin is a neuropeptide involved in dopamine signaling and thermoregulation.
  • Neurotensin receptor 1 (NTSRl) is normally expressed only in the brain and colon, but some cancers can overexpress NTSRl .
  • NTSRl is expressed in majority of pancreatic cancers, and has high expression in subsets of NSCLC and ductal breast carcinomas. NTSRl is involved in the growth of expressing cancer cells, and NTSRl expression correlates with poor prognosis.
  • Neurotensin is a 13-amino acid peptide with six C-terminal amino acids as the targeting domain for NTSRl .
  • the targeting moiety comprises the targeting domain of neurotensin or derivative thereof, e.g., six or seven C-terminal amino acids of neurotensin.
  • the targeting moiety may futher comprise a linking amino acid which attaches the targeting domain of neurotensin to a variety of releasable linkers.
  • the targeting domain of neurotensin may be modified to increase stability. For example, an isoleucine group on isoleucine residue may be replaced with tert-leucine for greater stability.
  • a targeting moiety comprise seven C-terminal amino acids of neurotensin with tert-leucine modification is shown below:
  • ILl IRa is an important cytokine receptor that is part of a multimeric complex comprising the ubiquitously expressed gpl30R subunit.
  • the complex triggers intracellular signaling and engagement of Stat3, which once activated, promotes cell survival and proliferation as well as immune responses associated with inflammatory diseases and tumor progression.
  • IL-1 IRa links oxidative stress and compensatory proliferation, regulates autoimmune demyelination and the invasion and proliferation of cancer cells.
  • Overexpression of IL-1 IRa indicates a poor long- term prognosis in cancer patients.
  • IL-1 IRa is an established molecular target in primary tumors of bone, such as osteosarcoma, and in secondary bone metastases from solid tumors, such as prostate cancer. It is related to breast cancer development and progression and may play a significant role in the bone metastasis of human breast cancer. It has limited expression in healthy tissues.
  • a mimic motif of ILl 1 (displaying the cyclic nonapeptide CGRRAGGSC) isolated from prostate biopsies binds specifically to IL-1 IRa.
  • the cyclic nonapeptide or its derivatives may be used as a targeting moiety in the conjugates of the present invention. Its structure is shown below.
  • the conjugates contain one or more linkers attaching the active agents and targeting moieties, wherein at least one linker is a penicillamine linker.
  • the penicillamine linker comprises a penicillamine group or derivative thereof.
  • the linker, Y is bound to one or more active agents and one or more targeting ligands to form a conjugate.
  • the linker Y is attached to the targeting moiety X and the active agent Z by functional groups independently selected from an ester bond, disulfide, amide, acylhydrazone, ether, carbamate, carbonate, and urea.
  • the bond between the penicillamine linker Y and the targeting moiety X and/or the active agnet Z may be cleavable.
  • the linker can be attached to either the targeting ligand or the active drug by a non-cleavable group such as provided by the conjugation between a thiol and a maleimide, an azide and an alkyne.
  • the linker comprises a cleavable functionality that is cleavable.
  • the cleavable functionality may be hydrolyzed in vivo or may be designed to be hydrolyzed enzymatically, for example by Cathepsin B.
  • a "cleavable" linker, as used herein, refers to any linker which can be cleaved physically or chemically.
  • Examples for physical cleavage may be cleavage by light, radioactive emission or heat, while examples for chemical cleavage include cleavage by re- dox-reactions, hydrolysis, pH-dependent cleavage or cleavage by enzymes.
  • the active agent is selected from Non-limiting examples of conjugates of the present invention include the following compoumds: a maytansinoid or derivative such as mertansine (DM1) or DM4, cabazitaxel, SN-38, or doxorubicin.
  • the targeting moiety is selected from a SSTR- binding group, a NTSRl-binding group, or an IL1 IRa-binding group.
  • the active agent Z is selected from DM1, DM4, cabazitaxel, SN-38, or doxorubin, and the targeting moiety X is a somatostatin receptor binding agent.
  • X may be selected from somatostatin, cyclo(AA-Tyr-DTrp- Lys-Thr-Phe), octreotide, vapreotide or TATE.
  • the active agent Z is connected to the C-terminus of X with the linker Y. In some embodiments, the active agent Z is connected to the N-terminus of X with the linker Y.
  • the active agent Z is connected to X with the linker Y, wherein the targeting moiety X comprises at least one D-Phe residue and the phenyl ring of the D- Phe residue has been replaced by a group containing linker Y.
  • the active agent Z is selected from DM1, DM4, cabazitaxel, SN-38, or doxorubin
  • the targeting moiety X is a NTSR1 binding agent.
  • X may comprise the targeting domain of neurotensin.
  • the active agent Z is selected from DM1, DM4, cabazitaxel, SN-38, or doxorubin
  • the targeting moiety X is a IL1 IRa binding agent.
  • X may comprise IL11 or derivative thereof.
  • Particles can be polymeric particles, lipid particles, solid lipid particles, inorganic particles, or combinations thereof (e.g., lipid stabilized polymeric particles).
  • the particles are polymeric particles or contain a polymeric matrix.
  • the particles can contain any of the polymers described herein or derivatives or copolymers thereof.
  • the particles generally contain one or more biocompatible polymers.
  • the polymers can be biodegradable polymers.
  • the polymers can be hydrophobic polymers, hydrophilic polymers, or amphiphilic polymers.
  • the particles contain one or more polymers having an additional targeting moiety attached thereto.
  • the size of the particles can be adjusted for the intended application.
  • the particles can be nanoparticles or microparticles.
  • the particle can have a diameter of about 10 nm to about 10 microns, about 10 nm to about 1 micron, about 10 nm to about 500 nm, about 20 nm to about 500 nm, or about 25 nm to about 250 nm.
  • the particle is a nanoparticle having a diameter from about 25 nm to about 250 nm. It is understood by those in the art that a plurality of particles will have a range of sizes and the diameter is understood to be the median diameter of the particle size distribution.
  • a particle may be a nanoparticle, i.e., the particle has a characteristic dimension of less than about 1 micrometer, where the
  • the characteristic dimension of a particle is the diameter of a perfect sphere having the same volume as the particle.
  • the plurality of particles can be characterized by an average diameter (e.g., the average diameter for the plurality of particles).
  • the diameter of the particles may have a Gaussian-type distribution.
  • the plurality of particles have an average diameter of less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, less than about 100 nm, less than about 50 nm, less than about 30 nm, less than about 10 nm, less than about 3 nm, or less than about 1 nm.
  • the particles have an average diameter of at least about 5 nm, at least about 10 nm, at least about 30 nm, at least about 50 nm, at least about 100 nm, at least about 150 nm, or greater. In certain embodiments, the plurality of the particles have an average diameter of about 10 nm, about 25 nm, about 50 nm, about 100 nm, about 150 nm, about 200 nm, about 250 nm, about 300 nm, about 500 nm, or the like.
  • the plurality of particles have an average diameter between about 10 nm and about 500 nm, between about 50 nm and about 400 nm, between about 100 nm and about 300 nm, between about 150 nm and about 250 nm, between about 175 nm and about 225 nm, or the like.
  • the plurality of particles have an average diameter between about 10 nm and about 500 nm, between about 20 nm and about 400 nm, between about 30 nm and about 300 nm, between about 40 nm and about 200 nm, between about 50 nm and about 175 nm, between about 60 nm and about 150 nm, between about 70 nm and about 130 nm, or the like.
  • the average diameter can be between about 70 nm and 130 nm.
  • the plurality of particles have an average diameter between about 20 nm and about 220 nm, between about 30 nm and about 200 nm, between about 40 nm and about 180 nm, between about 50 nm and about 170 nm, between about 60 nm and about 150 nm, or between about 70 nm and about 130 nm.
  • the particles have a size of 40 to 120 nm with a zeta potential close to 0 mV at low to zero ionic strengths (1 to 10 mM), with zeta potential values between + 5 to - 5 mV, and a zero/neutral or a small -ve surface charge.
  • the particles contain one or more conjugates as described above.
  • the conjugates can be present on the interior of the particle, on the exterior of the particle, or both.
  • the particles may comprise hydrophobic ion-pairing complexes or hydrophobic ion-pairs formed by one or more conjugates described above and counterions.
  • Hydrophobic ion-pairing is the interaction between a pair of oppositely charged ions held together by Coulombic attraction.
  • HIP refers to the interaction between the conjugate of the present invention and its counterions, wherein the counterion is not H + or HO " ions.
  • Hydrophobic ion-pairing complex or hydrophobic ion-pair refers to the complex formed by the conjugate of the present invention and its counterions.
  • the counterions are hydrophobic.
  • the counterions are provided by a hydrophobic acid or a salt of a hydrophobic acid.
  • the counterions are provided by bile acids or salts, fatty acids or salts, lipids, or amino acids.
  • the counterions are negatively charged (anionic).
  • Non- limited examples of negative charged counterions include the counterions sodium sulfosuccinate (AOT), sodium oleate, sodium dodecyl sulfate (SDS), human serum albumin (HSA), dextran sulphate, sodium deoxycholate, sodium cholate, anionic lipids, amino acids, or any combination thereof.
  • AOT sodium sulfosuccinate
  • SDS sodium dodecyl sulfate
  • HSA human serum albumin
  • dextran sulphate sodium deoxycholate
  • sodium cholate sodium cholate
  • anionic lipids amino acids, or any combination thereof.
  • HIP may increase the hydrophobicity and/or lipophilicity of the conjugate of the present invention.
  • increasing the hydrophobicity and/or lipophilicity of the conjugate of the present invention may be beneficial for particle formulations and may provide higher solubility of the conjugate of the present invention in organic solvents.
  • particle formulations that include HIP pairs have improved formulation properties, such as drug loading and/or release profile.
  • slow release of the conjugate of the invention from the particles may occur, due to a decrease in the conjugate's solubility in aqueous solution.
  • complexing the conjugate with large hydrophobic counterions may slow diffusion of the conjugate within a polymeric matrix.
  • HIP occurs without covalent conjuatation of the counterion to the conjugate of the present invention.
  • the strength of HIP may impact the drug load and release rate of the particles of the invention.
  • the strength of the HIP may be increased by increasing the magnitude of the difference between the pKa of the conjugate of the present invention and the pKa of the agent providing the counterion.
  • the conditions for ion pair formation may impact the drug load and release rate of the particles of the invention.
  • any suitable hydrophobic acid or a combination thereof may form a HIP pair with the conjugate of the present invention.
  • the hydrophobic acid may be a carboxylic acid (such as but not limited to a monocarboxylic acid, dicarboxylic acid, tricarboxylic acid), a sulfinic acid, a sulfenic acid, or a sulfonic acid.
  • a salt of a suitable hydrophobic acid such as but not limited to a monocarboxylic acid, dicarboxylic acid, tricarboxylic acid
  • a sulfinic acid such as but not limited to a monocarboxylic acid, dicarboxylic acid, tricarboxylic acid
  • a sulfinic acid such as but not limited to a monocarboxylic acid, dicarboxylic acid, tricarboxylic acid
  • a sulfinic acid such as but not limited to a monocarboxylic acid, dicarboxylic acid,
  • hydrophobic acid or a combination thereof may be used to form a HIP pair with the conjugate of the present invention.
  • hydrophobic acids saturated fatty acids, unsaturated fatty acids, aromatic acids, bile acid, polyelectrolyte, their dissociation constant in water (pKa) and logP values were disclosed in
  • WO2014/043,625 the contents of which are incorporated herein by reference in their entirety.
  • the strength of the hydrophobic acid, the difference between the pKa of the hydrophobic acid and the pKa of the conjuagate of the present invention, logP of the hydrophobic acid, the phase transition temperature of the hydrophobic acid, the molar ratio of the hydrophobic acid to the conjugate of the present invention, and the concentration of the hydrophobic acid were also disclosed in WO2014/043,625, the contents of which are incorporated herein by reference in their entirety.
  • particles of the present invention comprising a HIP complex and/or prepared by a process that provides a counterion to form HIP complex with the conjugate may have a highter drug loading than particles without a HIP complex or prepared by a process that does not provide any counterion to form HIP complex with the conjugate.
  • drug loading may increase 50%, 100%), 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, 8 times, 9 times, or 10 times.
  • the particles of the invention may retain the conjugate for at least about 1 minute, at least about 15 minutes, at least about 1 hour, when placed in a phosphate buffer solution at 37°C.
  • the weight percentage of the conjugate in the particles is at least about 0.05%, 0.1%, 0.5%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%), 40%), 45%), or 50%> such that the sum of the weight percentages of the components of the particles is 100%>.
  • the weight percentage of the conjugate in the particles is from about 0.5% to about 10%>, or about 10%> to about 20%, or about 20% to about 30%, or about 30% to about 40%, or about 40% to about 50%, or about 50% to about 60%, or about 60% to about 70%, or about 70% to about 80%, or about 80% to about 90%, or about 90% to about 99% such that the sum of the weight percentages of the components of the particles is 100%>.
  • a conjugate may have a molecular weight of less than about 50,000 Da, less than about 40,000 Da, less than about 30,000 Da, less than about 20,000 Da, less than about 15,000 Da, less than about 10,000 Da, less than about 8,000 Da, less than about 5,000 Da, or less than about 3,000 Da.
  • the conjugate may have a molecular weight of between about 1,000 Da and about 50,000 Da, between about 1,000 Da and about 40,000 Da, in some embodiments between about 1,000 Da and about 30,000 Da, in some embodiments bout 1,000 Da and about 50,000 Da, between about 1,000 Da and about 20,000 Da, in some embodiments between about 1,000 Da and about 15,000 Da, in some embodiments between about 1,000 Da and about 10,000 Da, in some embodiments between about 1,000 Da and about 8,000 Da, in some embodiments between about 1,000 Da and about 5,000 Da, and in some embodiments between about 1,000 Da and about 3,000 Da.
  • the molecular weight of the conjugate may be calculated as the sum of the atomic weight of each atom in the formula of the conjugate multiplied by the number of each atom.
  • the particles may contain one or more polymers.
  • Polymers may contain one more of the following polyesters: homopolymers including glycolic acid units, referred to herein as "PGA”, and lactic acid units, such as poly-L-lactic acid, poly-D- lactic acid, poly-D,L-lactic acid, poly-L-lactide, poly-D-lactide, and poly-D,L-lactide, collectively referred to herein as "PLA”, and caprolactone units, such as poly(s- caprolactone), collectively referred to herein as "PCL”; and copolymers including lactic acid and glycolic acid units, such as various forms of poly(lactic acid-co- gly colic acid) and poly(lactide-co-glycolide) characterized by the ratio of lactic acid:glycolic acid, collectively referred to herein as "PLGA”; and polyacrylates, and derivatives thereof.
  • PGA glycolic acid units
  • PLA poly-L-lactic acid
  • PCL poly
  • Exemplary polymers also include copolymers of polyethylene glycol (PEG) and the aforementioned polyesters, such as various forms of PLGA- PEG or PLA-PEG copolymers, collectively referred to herein as "PEGylated polymers".
  • PEG polyethylene glycol
  • the PEG region can be covalently associated with polymer to yield "PEGylated polymers" by a cleavable linker.
  • the particles may contain one or more hydrophilic polymers.
  • Hydrophilic polymers include cellulosic polymers such as starch and polysaccharides; hydrophilic polypeptides; poly(amino acids) such as poly-L-glutamic acid (PGS), gamma- polyglutamic acid, poly-L-aspartic acid, poly-L-serine, or poly-L-lysine; polyalkylene glycols and polyalkylene oxides such as polyethylene glycol (PEG), polypropylene glycol (PPG), and poly(ethylene oxide) (PEO); poly(oxyethylated polyol);
  • the particles may contain one or more hydrophobic polymers.
  • suitable hydrophobic polymers include polyhydroxyacids such as poly(lactic acid), poly(glycolic acid), and poly(lactic acid-coglycolic acids); polyhydroxyalkanoates such as poly3-hydroxybutyrate or poly4-hydroxybutyrate; polycaprolactones;
  • polyesteramides including synthetic and natural polyamides), polypeptides, and poly(amino acids); polyesteramides; polyesters; poly(dioxanones); poly(alkylene alkylates); hydrophobic polyethers; polyurethanes; polyetheresters; polyacetals; polycyanoacrylates;
  • polyacrylates polymethylmethacrylates; polysiloxanes;
  • poly(oxyethylene)/poly(oxypropylene) copolymers polyketals; polyphosphates; polyhydroxyvalerates; polyalkylene oxalates; polyalkylene succinates; poly(maleic acids), as well as copolymers thereof.
  • the hydrophobic polymer is an aliphatic polyester. In some embodiments, the hydrophobic polymer is poly(lactic acid), poly(glycolic acid), or poly(lactic acid-co-glycolic acid). [00179]
  • the particles can contain one or more biodegradable polymers.
  • Biodegradable polymers can include polymers that are insoluble or sparingly soluble in water that are converted chemically or enzymatically in the body into water-soluble materials.
  • Biodegradable polymers can include soluble polymers crosslinked by hydolyzable cross-linking groups to render the crosslinked polymer insoluble or sparingly soluble in water.
  • Biodegradable polymers in the particle can include polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose such as methyl cellulose and ethyl cellulose, hydroxyalkyl celluloses such as hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, and hydroxybutyl methyl cellulose, cellulose ethers, cellulose esters, nitro celluloses, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose sulphate sodium salt, polymers of acrylic and
  • exemplary biodegradable polymers include polyesters, poly(ortho esters), poly(ethylene imines), poly(caprolactones), poly(hydroxyalkanoates), poly(hydroxyvalerates),
  • polyanhydrides poly(acrylic acids), polyglycolides, poly(urethanes), polycarbonates, polyphosphate esters, polyphosphazenes, derivatives thereof, linear and branched copolymers and block copolymers thereof, and blends thereof.
  • the particle contains biodegradable polyesters or polyanhydrides such as poly(lactic acid), poly(glycolic acid), and poly(lactic-co-glycolic acid).
  • the particles can contain one or more amphiphilic polymers.
  • Amphiphilic polymers can be polymers containing a hydrophobic polymer block and a hydrophilic polymer block.
  • the hydrophobic polymer block can contain one or more of the hydrophobic polymers above or a derivative or copolymer thereof.
  • the hydrophilic polymer block can contain one or more of the hydrophilic polymers above or a derivative or copolymer thereof.
  • the amphiphilic polymer is a di-block polymer containing a hydrophobic end formed from a hydrophobic polymer and a hydrophilic end formed of a hydrophilic polymer.
  • a moiety can be attached to the hydrophobic end, to the hydrophilic end, or both.
  • the particle can contain two or more amphiphilic polymers.
  • the particles may contain one or more lipids or amphiphilic compounds.
  • the particles can be liposomes, lipid micelles, solid lipid particles, or lipid-stabilized polymeric particles.
  • the lipid particle can be made from one or a mixture of different lipids.
  • Lipid particles are formed from one or more lipids, which can be neutral, anionic, or cationic at physiologic pH.
  • the lipid particle in some embodiments, incorporates one or more biocompatible lipids.
  • the lipid particles may be formed using a combination of more than one lipid. For example, a charged lipid may be combined with a lipid that is non-ionic or uncharged at physiological pH.
  • the particle can be a lipid micelle.
  • Lipid micelles for drug delivery are known in the art.
  • Lipid micelles can be formed, for instance, as a water-in-oil emulsion with a lipid surfactant.
  • An emulsion is a blend of two immiscible phases wherein a surfactant is added to stabilize the dispersed droplets.
  • the lipid micelle is a microemulsion.
  • a microemulsion is a thermodynamically stable system composed of at least water, oil and a lipid surfactant producing a transparent and thermodynamically stable system whose droplet size is less than 1 micron, from about 10 nm to about 500 nm, or from about 10 nm to about 250 nm.
  • Lipid micelles are generally useful for encapsulating hydrophobic active agents, including hydrophobic therapeutic agents, hydrophobic prophylactic agents, or hydrophobic diagnostic agents.
  • the particle can be a liposome.
  • Liposomes are small vesicles composed of an aqueous medium surrounded by lipids arranged in spherical bilayers. Liposomes can be classified as small unilamellar vesicles, large unilamellar vesicles, or multilamellar vesicles. Multi-lamellar liposomes contain multiple concentric lipid bilayers. Liposomes can be used to encapsulate agents, by trapping hydrophilic agents in the aqueous interior or between bilayers, or by trapping hydrophobic agents within the bilayer.
  • the lipid micelles and liposomes typically have an aqueous center.
  • the aqueous center can contain water or a mixture of water and alcohol.
  • suitable alcohols include, but are not limited to, methanol, ethanol, propanol, (such as isopropanol), butanol (such as «-butanol, isobutanol, sec-butanol, tert-butanol, pentanol (such as amyl alcohol, isobutyl carbinol), hexanol (such as 1-hexanol, 2-hexanol, 3-hexanol), heptanol (such as 1-heptanol, 2-heptanol, 3-heptanol and 4-heptanol) or octanol (such as 1 -octanol) or a combination thereof.
  • the particle can be a solid lipid particle.
  • Solid lipid particles present an alternative to the colloidal micelles and liposomes.
  • Solid lipid particles are typically submicron in size, i.e. from about 10 nm to about 1 micron, from 10 nm to about 500 nm, or from 10 nm to about 250 nm.
  • Solid lipid particles are formed of lipids that are solids at room temperature. They are derived from oil-in-water emulsions, by replacing the liquid oil by a solid lipid.
  • Suitable neutral and anionic lipids include, but are not limited to, sterols and lipids such as cholesterol, phospholipids, lysolipids, lysophospholipids, sphingolipids or pegylated lipids.
  • Neutral and anionic lipids include, but are not limited to, phosphatidylcholine (PC) (such as egg PC, soy PC), including 1 ,2-diacyl- glycero-3-phosphocholines; phosphatidylserine (PS), phosphatidylglycerol, phosphatidylinositol (PI); glycolipids; sphingophospholipids such as sphingomyelin and sphingoglycolipids (also known as 1-ceramidyl glucosides) such as ceramide galactopyranoside, gangliosides and cerebrosides; fatty acids, sterols, containing a carboxylic acid group for example, cholesterol; 1 ,2-diacyl-sn-glycero-3- phosphoethanolamine, including, but not limited to, 1 ,2-dioleylphosphoethanolamine (DOPE), 1 ,2-dihexadecylphosphoethanolamine (DH
  • the lipids can also include various natural (e.g., tissue derived L-a-phosphatidyl: egg yolk, heart, brain, liver, soybean) and/or synthetic (e.g., saturated and unsaturated l,2-diacyl-s «-glycero-3- phosphocholines, l-acyl-2-acyl-s «-glycero-3-phosphocholines, 1,2-diheptanoyl-SN- glycero-3-phosphocholine) derivatives of the lipids.
  • tissue derived L-a-phosphatidyl egg yolk, heart, brain, liver, soybean
  • synthetic e.g., saturated and unsaturated l,2-diacyl-s «-glycero-3- phosphocholines, l-acyl-2-acyl-s «-glycero-3-phosphocholines, 1,2-diheptanoyl-SN- glycero-3-phosphocholine
  • Suitable cationic lipids include, but are not limited to, N-[l-(2,3- dioleoyloxy)propyl]-N,N,N-trimethyl ammonium salts, also references as TAP lipids, for example methylsulfate salt.
  • Suitable TAP lipids include, but are not limited to, DOTAP (dioleoyl-), DMTAP (dimyristoyl-), DPTAP (dipalmitoyl-), and DSTAP (distearoyl-).
  • Suitable cationic lipids in the liposomes include, but are not limited to, dimethyldioctadecyl ammonium bromide (DDAB), 1 ,2-diacyloxy-3- trimethylammonium propanes, N-[l-(2,3-dioloyloxy)propyl]-N,N-dimethyl amine (DODAP), 1 ,2-diacyloxy-3-dimethylammonium propanes, N-[l-(2,3- dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), 1 ,2-dialkyloxy- 3-dimethylammonium propanes, dioctadecylamidoglycylspermine (DOGS), 3 -[N- (N',N'-dimethylamino-ethane)carbamoyl]cholesterol (DC-Choi); 2,3-dioleoyloxy-N- (2-(spermine
  • the cationic lipids can be l-[2-(acyloxy)ethyl]2-alkyl(alkenyl)-3-(2-hydroxyethyl)- imidazolinium chloride derivatives, for example, l-[2-(9(Z)-octadecenoyloxy)ethyl]- 2-(8(Z)-heptadecenyl-3-(2-hydroxyethyl)imidazolinium chloride (DOTIM), and l-[2- (hexadecanoyloxy)ethyl]-2-pentadecyl-3-(2-hydroxyethyl)imidazolinium chloride (DPTIM).
  • DOTIM DOTIM
  • DPTIM l-[2- (hexadecanoyloxy)ethyl]-2-pentadecyl-3-(2-hydroxyethyl)imidazolinium chloride
  • the cationic lipids can be 2,3-dialkyloxypropyl quaternary ammonium compound derivatives containing a hydroxyalkyl moiety on the quaternary amine, for example, 1 ,2-dioleoyl-3-dimethyl-hydroxy ethyl ammonium bromide (DORI), 1 ,2-dioleyloxypropyl-3-dimethyl-hydroxy ethyl ammonium bromide (DORIE), 1 ,2-dioleyloxypropyl-3-dimetyl-hydroxypropyl ammonium bromide (DORIE-HP), 1 ,2-dioleyl-oxy-propyl-3-dimethyl-hydroxybutyl ammonium bromide (DORIE-HB), 1 ,2-dioleyloxypropyl-3-dimethyl-hydroxypentyl ammonium bromide (DORIE-Hpe), 1 ,2-dimyristyloxypropyl-3-dimethyl-hydroxye
  • Suitable solid lipids include, but are not limited to, higher saturated alcohols, higher fatty acids, sphingolipids, synthetic esters, and mono-, di-, and triglycerides of higher saturated fatty acids.
  • Solid lipids can include aliphatic alcohols having 10-40, for example, 12-30 carbon atoms, such as cetostearyl alcohol.
  • Solid lipids can include higher fatty acids of 10-40, for example, 12-30 carbon atoms, such as stearic acid, palmitic acid, decanoic acid, and behenic acid.
  • Solid lipids can include glycerides, including monoglycerides, diglycerides, and triglycerides, of higher saturated fatty acids having 10-40, for example, 12-30 carbon atoms, such as glyceryl monostearate, glycerol behenate, glycerol palmitostearate, glycerol trilaurate, tricaprin, trilaurin, trimyristin, tripalmitin, tristearin, and hydrogenated castor oil.
  • Suitable solid lipids can include cetyl palmitate, beeswax, or cyclodextrin.
  • Amphiphilic compounds include, but are not limited to, phospholipids, such as 1,2 distearoyl-sn-glycero-3-phosphoethanolamine (DSPE),
  • dipalmitoylphosphatidylcholine DPPC
  • distearoylphosphatidylcholine DSPC
  • diarachidoylphosphatidylcholine DAPC
  • dibehenoylphosphatidylcholine DBPC
  • ditricosanoylphosphatidylcholine DTPC
  • dilignoceroylphatidylcholine DLPC
  • DPPC dipalmitoylphosphatidylcholine
  • DSPC distearoylphosphatidylcholine
  • DAPC diarachidoylphosphatidylcholine
  • DBPC dibehenoylphosphatidylcholine
  • DTPC ditricosanoylphosphatidylcholine
  • DLPC dilignoceroylphatidylcholine
  • Phospholipids that may be used include, but are not limited to, phosphatidic acids, phosphatidyl cholines with both saturated and unsaturated lipids, phosphatidyl ethanolamines, phosphatidylglycerols,
  • phosphatidylserines examples include, but are not limited to, phosphatidylcholines such as dioleoylphosphatidylcholine,
  • dimyristoylphosphatidylcholine dipentadecanoylphosphatidylcholine
  • DPPC dipalmitoylphosphatidylcholine
  • DSPC distearoylphosphatidylcholine
  • DAPC diarachidoylphosphatidylcholine
  • DBPC dibehenoylphosphatidylcho- line
  • DTPC ditricosanoylphosphatidylcholine
  • DLPC dilignoceroylphatidylcholine
  • phosphatidylethanolamines such as dioleoylphosphatidylethanolamine or 1 -hexadecyl-2-palmitoylglycerophos- phoethanolamine.
  • the particles can contain one or more additional active agents in addition to those in the conjugates.
  • the additional active agents can be therapeutic, prophylactic, diagnostic, or nutritional agents as listed above.
  • the additional active agents can be present in any amount, e.g. from about 0.5% to about 90%, from about 0.5% to about 50%), from about 0.5% to about 25%, from about 0.5% to about 20%, from about 0.5% to about 10%), or from about 5% to about 10% (w/w) based upon the weight of the particle.
  • the agents are incorporated in an about 0.5% to about
  • the particles can contain one or more targeting moieties targeting the particle to a specific organ, tissue, cell type, or subcellular compartment in addition to the targeting moieties of the conjugate.
  • the additional targeting moieties can be present on the surface of the particle, on the interior of the particle, or both.
  • the additional targeting moieties can be immobilized on the surface of the particle, e.g., can be covalently attached to polymer or lipid in the particle.
  • the additional targeting moieties are covalently attached to an amphiphilic polymer or a lipid such that the targeting moieties are oriented on the surface of the particle.
  • compositions are administered to humans, human patients or subjects.
  • active ingredient generally refers to the conjugate or particles comprising the conjugates to be delivered as described herein.
  • compositions are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the
  • compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation.
  • Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
  • Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • a pharmaceutical composition in accordance with the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a "unit dose" is discrete amount of the
  • composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • compositions in accordance with the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100%, e.g., between .5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w) active ingredient.
  • the conjugates or particles of the present invention can be formulated using one or more excipients to: (1) increase stability; (2) permit the sustained or delayed release (e.g., from a depot formulation of the monomaleimide); (3) alter the biodistribution (e.g., target the monomaleimide compounds to specific tissues or cell types); (4) alter the release profile of the monomaleimide compounds in vivo.
  • excipients include any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, and preservatives.
  • Excipients of the present invention may also include, without limitation, lipidoids, liposomes, lipid nanoparticles, polymers, lipoplexes, core-shell nanoparticles, peptides, proteins, hyaluronidase, nanoparticle mimics and combinations thereof. Accordingly, the formulations of the invention may include one or more excipients, each in an amount that together increases the stability of the monomaleimide compounds.
  • compositions may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable excipient includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's The Science and Practice of Pharmacy 21st Edition, A. R. Gennaro (Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference in its entirety) discloses various excipients
  • compositions and known techniques for the preparation thereof Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention.
  • a pharmaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure.
  • an excipient is approved for use in humans and for veterinary use.
  • an excipient is approved by United States Food and Drug Administration.
  • an excipient is pharmaceutical grade.
  • an excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
  • compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical compositions.
  • Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
  • Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross- linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (VEEGUM®), sodium lauryl sulfate, quaternary ammonium compounds, etc., and/or combinations thereof.
  • crospovidone cross- linked poly(vinyl-pyrrolidone)
  • Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and VEEGUM® [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g.
  • natural emulsifiers e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin
  • colloidal clays e.g. bentonite [aluminum silicate
  • stearyl alcohol cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
  • polyoxyethylene sorbitan monolaurate [TWEEN®20], polyoxyethylene sorbitan [TWEENn®60], polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], sorbitan monostearate
  • polyoxyethylene esters e.g. polyoxyethylene monostearate [MYRJ®45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and SOLUTOL®
  • sucrose fatty acid esters e.g. CREMOPHOR®
  • polyoxyethylene ethers e.g.
  • polyoxyethylene lauryl ether [BRIJ®30]), poly(vinyl-pyrrolidone), di ethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, PLUORINC®F 68,
  • Exemplary binding agents include, but are not limited to, starch (e.g.
  • cornstarch and starch paste cornstarch and starch paste
  • gelatin e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol,); natural and synthetic gums (e.g.
  • acacia sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum®), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol;
  • inorganic calcium salts silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof.
  • Exemplary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives.
  • Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and/or sodium sulfite.
  • Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate.
  • EDTA ethylenediaminetetraacetic acid
  • citric acid monohydrate disodium edetate
  • dipotassium edetate dipotassium edetate
  • edetic acid fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate.
  • antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and/or thimerosal.
  • Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid.
  • Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol.
  • Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid.
  • preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, GLYDANT PLUS®, PHENONIP®, methylparaben, GERMALL® 115, GERMABEN®II, EOLO ETM, KATHONTM, and/or EUXYL®.
  • Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, is
  • Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.
  • Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl my ri state, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquan
  • cyclomethicone diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and/or combinations thereof.
  • Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator.
  • the conjugates or particles of the present invention may be administered by any route which results in a therapeutically effective outcome. These include, but are not limited to enteral, gastroenteral, epidural, oral, transdermal, epidural (peridural), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself),
  • compositions may be administered in a way which allows them cross the blood-brain barrier, vascular barrier, or other epithelial
  • the formulations described herein contain an effective amount of conjugates or particles in a pharmaceutical carrier appropriate for administration to an individual in need thereof.
  • the formulations may be administered parenterally (e.g., by injection or infusion).
  • the formulations or variations thereof may be administered in any manner including enterally, topically (e.g., to the eye), or via pulmonary administration. In some embodiments the formulations are administered topically.
  • parenteral Formulations e.g., parenteral Formulations
  • the conjugates or particles can be formulated for parenteral delivery, such as injection or infusion, in the form of a solution, suspension or emulsion.
  • the formulation can be administered systemically, regionally or directly to the organ or tissue to be treated.
  • Parenteral formulations can be prepared as aqueous compositions using techniques is known in the art.
  • such compositions can be prepared as injectable formulations, for example, solutions or suspensions; solid forms suitable for using to prepare solutions or suspensions upon the addition of a reconstitution medium prior to injection; emulsions, such as water-in-oil (w/o) emulsions, oil-in- water (o/w) emulsions, and microemulsions thereof, liposomes, or emulsomes.
  • injectable formulations for example, solutions or suspensions
  • solid forms suitable for using to prepare solutions or suspensions upon the addition of a reconstitution medium prior to injection emulsions, such as water-in-oil (w/o) emulsions, oil-in- water (o/w) emulsions, and microemulsions thereof, liposomes, or emulsomes.
  • emulsions such as water-in-oil (w/o) emulsions
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, one or more polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), oils, such as vegetable oils (e.g., peanut oil, corn oil, sesame oil, etc.), and combinations thereof.
  • polyols e.g., glycerol, propylene glycol, and liquid polyethylene glycol
  • oils such as vegetable oils (e.g., peanut oil, corn oil, sesame oil, etc.)
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and/or by the use of surfactants.
  • an isotonic agent is included, for example, one or more sugars, sodium chloride, or other suitable agent known in the art.
  • Solutions and dispersions of the conjugates or particles can be prepared in water or another solvent or dispersing medium suitably mixed with one or more pharmaceutically acceptable excipients including, but not limited to, surfactants, dispersants, emulsifiers, pH modifying agents, and combinations thereof.
  • Suitable surfactants may be anionic, cationic, amphoteric or nonionic surface active agents.
  • Suitable anionic surfactants include, but are not limited to, those containing carboxylate, sulfonate and sulfate ions.
  • anionic surfactants include sodium, potassium, ammonium of long chain alkyl sulfonates and alkyl aryl sulfonates such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium bis-(2-ethylthioxyl)-sulfosuccinate; and alkyl sulfates such as sodium lauryl sulfate.
  • Cationic surfactants include, but are not limited to, quaternary ammonium compounds such as benzalkonium chloride, benzethonium chloride, cetrimonium bromide, stearyl dimethylbenzyl ammonium chloride, polyoxyethylene and coconut amine.
  • nonionic surfactants include ethylene glycol monostearate, propylene glycol myristate, glyceryl monostearate, glyceryl stearate, polyglyceryl-4- oleate, sorbitan acylate, sucrose acylate, PEG- 150 laurate, PEG-400 monolaurate, polyoxyethylene monolaurate, polysorbates, polyoxyethylene octylphenylether, PEG- 1000 cetyl ether, polyoxyethylene tridecyl ether, polypropylene glycol butyl ether, Poloxamer® 401, stearoyl monoisopropanolamide, and polyoxyethylene
  • amphoteric surfactants include sodium N- dodecyl-P-alanine, sodium N-lauryl-P-iminodipropionate, myristoamphoacetate, lauryl betaine and lauryl sulfobetaine.
  • the formulation can contain a preservative to prevent the growth of microorganisms. Suitable preservatives include, but are not limited to, parabens, chlorobutanol, phenol, sorbic acid, and thimerosal.
  • the formulation may also contain an antioxidant to prevent degradation of the active agent(s) or particles.
  • the formulation is typically buffered to a pH of 3-8 for parenteral administration upon reconstitution.
  • Suitable buffers include, but are not limited to, phosphate buffers, acetate buffers, and citrate buffers. If using 10% sucrose or 5% dextrose, a buffer may not be required.
  • Water soluble polymers are often used in formulations for parenteral administration. Suitable water-soluble polymers include, but are not limited to, polyvinylpyrrolidone, dextran, carboxymethylcellulose, and polyethylene glycol.
  • Sterile injectable solutions can be prepared by incorporating the conjugates or particles in the required amount in the appropriate solvent or dispersion medium with one or more of the excipients listed above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized conjugates or particles into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those listed above.
  • examples of methods of preparation include vacuum-drying and freeze-drying techniques that yield a powder of the particle plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the powders can be prepared in such a manner that the particles are porous in nature, which can increase dissolution of the particles. Methods for making porous particles are known in the art.
  • compositions for parenteral administration can be in the form of a sterile aqueous solution or suspension of conjugates or particles formed from one or more polymer-drug conjugates.
  • Acceptable solvents include, for example, water, Ringer's solution, phosphate buffered saline (PBS), and isotonic sodium chloride solution.
  • PBS phosphate buffered saline
  • the formulation may also be a sterile solution, suspension, or emulsion in a nontoxic, parenterally acceptable diluent or solvent such as 1,3- butanediol.
  • the formulation is distributed or packaged in a liquid form.
  • formulations for parenteral administration can be packed as a solid, obtained, for example by lyophilization of a suitable liquid formulation.
  • the solid can be reconstituted with an appropriate carrier or diluent prior to
  • Solutions, suspensions, or emulsions for parenteral administration may be buffered with an effective amount of buffer necessary to maintain a pH suitable for ocular administration.
  • Suitable buffers are well known by those skilled in the art and some examples of useful buffers are acetate, borate, carbonate, citrate, and phosphate buffers.
  • Solutions, suspensions, or emulsions for parenteral administration may also contain one or more tonicity agents to adjust the isotonic range of the formulation.
  • Suitable tonicity agents are well known in the art and some examples include glycerin, sucrose, dextrose, mannitol, sorbitol, sodium chloride, and other electrolytes.
  • Solutions, suspensions, or emulsions for parenteral administration may also contain one or more preservatives to prevent bacterial contamination of the
  • Suitable preservatives are known in the art, and include polyhexamethylenebiguanidine (PHMB), benzalkonium chloride (BAK), stabilized oxychloro complexes (otherwise known as Purite®), phenylmercuric acetate, chlorobutanol, sorbic acid, chlorhexidine, benzyl alcohol, parabens, thimerosal, and mixtures thereof.
  • PHMB polyhexamethylenebiguanidine
  • BAK benzalkonium chloride
  • Purite® stabilized oxychloro complexes
  • phenylmercuric acetate chlorobutanol
  • sorbic acid chlorhexidine
  • chlorhexidine chlorhexidine
  • parabens parabens
  • thimerosal and mixtures thereof.
  • Solutions, suspensions, or emulsions for parenteral administration may also contain one or more excipients known art, such as dispersing agents, wetting agents, and suspending agents.
  • the conjugates or particles can be formulated for topical administration to a mucosal surface Suitable dosage forms for topical administration include creams, ointments, salves, sprays, gels, lotions, emulsions, liquids, and transdermal patches.
  • the formulation may be formulated for transmucosal transepithelial, or
  • compositions contain one or more chemical penetration enhancers, membrane permeability agents, membrane transport agents, emollients, surfactants, stabilizers, and combination thereof.
  • the conjugates or particles can be administered as a liquid formulation, such as a solution or suspension, a semi-solid formulation, such as a lotion or ointment, or a solid formulation.
  • the conjugates or particles are formulated as liquids, including solutions and suspensions, such as eye drops or as a semi-solid formulation, to the mucosa, such as the eye or vaginally or rectally.
  • surfactants are surface-active agents that lower surface tension and thereby increase the emulsifying, foaming, dispersing, spreading and wetting properties of a product.
  • Suitable non-ionic surfactants include emulsifying wax, glyceryl monooleate, polyoxyethylene alkyl ethers, polyoxyethylene castor oil derivatives, polysorbate, sorbitan esters, benzyl alcohol, benzyl benzoate,
  • the non-ionic surfactant is stearyl alcohol.
  • Emmulsifiers are surface active substances which promote the suspension of one liquid in another and promote the formation of a stable mixture, or emulsion, of oil and water. Common emulsifiers are: metallic soaps, certain animal and vegetable oils, and various polar compounds.
  • Suitable emulsifiers include acacia, anionic emulsifying wax, calcium stearate, carbomers, cetostearyl alcohol, cetyl alcohol, cholesterol, diethanolamine, ethylene glycol palmitostearate, glycerin monostearate, glyceryl monooleate, hydroxpropyl cellulose, hypromellose, lanolin, hydrous, lanolin alcohols, lecithin, medium-chain triglycerides, methylcellulose, mineral oil and lanolin alcohols, monobasic sodium phosphate, monoethanolamine, nonionic emulsifying wax, oleic acid, poloxamer, poloxamers, polyoxyethylene alkyl ethers, polyoxyethylene castor oil derivatives, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene stearates, propylene glycol alginate, self-emulsifying glyceryl monostearate, sodium citrate dehydrate, sodium lauryl sulf
  • Suitable classes of penetration enhancers include, but are not limited to, fatty alcohols, fatty acid esters, fatty acids, fatty alcohol ethers, amino acids, phospholipids, lecithins, cholate salts, enzymes, amines and amides, complexing agents (liposomes, cyclodextrins, modified celluloses, and diimides), macrocyclics, such as macrocylic lactones, ketones, and anhydrides and cyclic ureas, surfactants, N-methyl pyrrolidones and derivatives thereof, DMSO and related compounds, ionic compounds, azone and related compounds, and solvents, such as alcohols, ketones, amides, polyols (e.g., glycols). Examples of these classes are known in the art.
  • the present invention provides methods comprising administering conjugates or particles containing the conjugate as described herein to a subject in need thereof.
  • Conjugates or particles containing the conjugates as described herein may be administered to a subject using any amount and any route of administration effective for preventing or treating or imaging a disease, disorder, and/or condition (e.g., a disease, disorder, and/or condition relating to working memory deficits).
  • a disease, disorder, and/or condition e.g., a disease, disorder, and/or condition relating to working memory deficits.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like.
  • compositions in accordance with the invention are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present invention may be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective, prophylactically effective, or appropriate imaging dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of
  • compositions in accordance with the present invention may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.001 mg/kg to about 0.05 mg/kg, from about 0.005 mg/kg to about 0.05 mg/kg, from about 0.001 mg/kg to about 0.005 mg/kg, from about 0.05 mg/kg to about 0.5 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, prophylactic, or imaging effect.
  • the desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks.
  • the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
  • multiple administrations e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations.
  • split dosing regimens such as those described herein may be used.
  • the concentration of the conjugates or particles of the present invention may be between about 0.01 mg/mL to about 50 mg/mL, about 0.1 mg/mL to about 25 mg/mL, about 0.5 mg/mL to about 10 mg/mL, or about 1 mg/mL to about 5 mg/mL in the pharmaceutical composition.
  • a "split dose” is the division of single unit dose or total daily dose into two or more doses, e.g, two or more administrations of the single unit dose.
  • a "single unit dose” is a dose of any therapeutic administed in one dose/at one time/single route/single point of contact, i.e., single administration event.
  • a "total daily dose” is an amount given or prescribed in 24 hr period. It may be administered as a single unit dose.
  • the monomaleimide compounds of the present invention are administed to a subject in split doses.
  • the monomaleimide compounds may be formulated in buffer only or in a formulation described herein.
  • a pharmaceutical composition described herein can be formulated into a dosage form described herein, such as a topical, intranasal, intratracheal, or injectable (e.g., intravenous, intraocular, intravitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous).
  • injectable e.g., intravenous, intraocular, intravitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous.
  • Liquid dosage forms for parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs.
  • liquid dosage forms may comprise inert diluents commonly used in the art including, but not limited to, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art including, but not limited to,
  • compositions may be mixed with solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
  • solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art and may include suitable dispersing agents, wetting agents, and/or suspending agents.
  • Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed include, but are not limited to, water, Ringer's solution, U.S. P., and isotonic sodium chloride solution.
  • Sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • Fatty acids such as oleic acid can be used in the preparation of injectables.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • the absorption of the active ingredient may be desirable to slow the absorption of the active ingredient from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the monomaleimide compounds then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered monomaleimide compound may be accomplished by dissolving or suspending the monomalimide in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the monomaleimide compunds in biodegradable polymers such as polylactide-polyglycolide.
  • the rate of monomaleimide compound release can be controlled.
  • biodegradable polymers include, but are not limited to, poly(orthoesters) and poly(anhydrides). Depot injectable formulations may be prepared by entrapping the monomaleimide compounds in liposomes or microemulsions which are compatible with body tissues.
  • Formulations described herein as being useful for pulmonary delivery may also be used for intranasal delivery of a pharmaceutical composition.
  • Another formulation suitable for intranasal administration may be a coarse powder comprising the active ingredient and having an average particle from about 0.2 ⁇ to 500 ⁇ .
  • Such a formulation may be administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.
  • Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration.
  • Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, contain about 0.1% to 20% (w/w) active ingredient, where the balance may comprise an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein.
  • formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising active ingredient.
  • Such powdered, aerosolized, and/or aerosolized formulations when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.
  • Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active
  • embedding compositions which can be used include polymeric substances and waxes.
  • Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • a method of making the particles includes providing a conjugate; providing a base component such as PLA-PEG or PLGA-PEG for forming a particle; combining the conjugate and the base component in an organic solution to form a first organic phase; and combining the first organic phase with a first aqueous solution to form a second phase; emulsifying the second phase to form an emulsion phase; and recovering particles.
  • the emulsion phase is further homogenized.
  • the first phase includes about 5 to about 50% weight, e.g. about 1 to about 40% solids, or about 5 to about 30% solids, e.g. about 5%), 10%), 15%), and 20%, of the conjugate and the base component. In certain embodiments, the first phase includes about 5% weight of the conjugate and the base component.
  • the organic phase comprises acetonitrile, tetrahydrofuran, ethyl acetate, isopropyl alcohol, isopropyl acetate,
  • the organic phase includes benzyl alcohol, ethyl acetate, or a combination thereof.
  • the aqueous solution includes water, sodium cholate, ethyl acetate, or benzyl alcohol.
  • a surfactant is added into the first phase, the second phase, or both.
  • a surfactant in some instances, can act as an emulsifier or a stabilizer for a composition disclosed herein.
  • a suitable surfactant can be a cationic surfactant, an anionic surfactant, or a nonionic surfactant.
  • a surfactant suitable for making a composition described herein includes sorbitan fatty acid esters, polyoxyethylene sorbitan fatty acid esters and polyoxyethylene stearates.
  • fatty acid ester nonionic surfactants examples include the TWEEN® 80, SPAN® 80, and MYJ® surfactants from ICI.
  • SPAN® surfactants include C12-C18 sorbitan monoesters.
  • TWEEN® surfactants include poly(ethylene oxide) C12-C18 sorbitan monoesters.
  • MYJ® surfactants include poly(ethylene oxide) stearates.
  • the aqueous solution also comprises a surfactant (e.g., an emulsifier), including a polysorbate.
  • the aqueous solution can include polysorbate 80.
  • a suitable surfactant includes a lipid-based surfactant.
  • the composition can include 1 ,2-dihexanoyl-sn-glycero-3 -phosphocholine, 1 ,2-diheptanoyl-sn-glycero-3 - phosphocholine, PEGlyated l,2-distearoyl-sn-glycero-3-phosphoethanolamine (including PEG5000-DSPE), PEGlyated l,2-dioleoyl-sn-glycero-3- phosphoethanolamine (including l,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-5000] (ammonium salt)).
  • Emulsifying the second phase to form an emulsion phase may be performed in one or two emulsification steps.
  • a primary emulsion may be prepared, and then emulsified to form a fine emulsion.
  • the primary emulsion can be formed, for example, using simple mixing, a high pressure homogenizer, probe sonicator, stir bar, or a rotor stator homogenizer.
  • the primary emulsion may be formed into a fine emulsion through the use of e.g. a probe sonicator or a high pressure homogenizer, e.g. by pass(es) through a homogenizer.
  • the pressure used may be about 4000 to about 8000 psi, about 4000 to about 5000 psi, or 4000 or 5000 psi.
  • Either solvent evaporation or dilution may be needed to complete the extraction of the solvent and solidify the particles.
  • a solvent dilution via aqueous quench may be used.
  • the emulsion can be diluted into cold water to a concentration sufficient to dissolve all of the organic solvent to form a quenched phase.
  • Quenching may be performed at least partially at a temperature of about 5°C or less.
  • water used in the quenching may be at a temperature that is less that room temperature (e.g. about 0 to about 10 °C, or about 0 to about 5 °C).
  • the particles are recovered by filtration.
  • ultrafiltration membranes can be used.
  • Exemplary filtration may be performed using a tangential flow filtration system.
  • a membrane with a pore size suitable to retain particles while allowing solutes, micelles, and organic solvent to pass particles can be selectively separated.
  • Exemplary membranes with molecular weight cut-offs of about 300-500 kDa (-5-25 nm) may be used.
  • the particles are freeze-dried or lyophilized, in some instances, to extend their shelf life.
  • the composition also includes a lyoprotectant.
  • a lyoprotectant is selected from a sugar, a polyalcohol, or a derivative thereof.
  • a lyoprotectant is selected from a monosaccharide, a disaccharide, or a mixture thereof.
  • a lyoprotectant can be sucrose, lactulose, trehalose, lactose, glucose, maltose, mannitol, cellobiose, or a mixture thereof.
  • the particles can be polymeric particles, lipid particles, or combinations thereof.
  • the various methods described herein can be adjusted to control the size and composition of the particles, e.g. some methods are best suited for preparing microparticles while others are better suited for preparing particles.
  • the selection of a method for preparing particles having the descried characteristics can be performed by the skilled artisan without undue experimentation.
  • Polymeric particles can be prepared using any suitable method known in the art.
  • Common microencapsulation techniques include, but are not limited to, spray drying, interfacial polymerization, hot melt encapsulation, phase separation encapsulation (spontaneous emulsion microencapsulation, solvent evaporation microencapsulation, and solvent removal microencapsulation), coacervation, low temperature microsphere formation, and phase inversion nanoencapsulation (PEST).
  • Interfacial polymerization can also be used to encapsulate one or more conjugates and/or active agents.
  • a monomer and the conjugates or active agent(s) are dissolved in a solvent.
  • a second monomer is dissolved in a second solvent (typically aqueous) which is immiscible with the first.
  • An emulsion is formed by suspending the first solution through stirring in the second solution. Once the emulsion is stabilized, an initiator is added to the aqueous phase causing interfacial polymerization at the interface of each droplet of emulsion.
  • Microspheres can be formed from polymers such as polyesters and polyanhydrides using hot melt microencapsulation methods as described in
  • polymers with molecular weights between 3,000-75,000 daltons are used.
  • the polymer first is melted and then mixed with the solid particles of one or more active agents to be incorporated that have been sieved to less than 50 microns.
  • the mixture is suspended in a non-miscible solvent (like silicon oil), and, with continuous stirring, heated to 5°C above the melting point of the polymer. Once the emulsion is stabilized, it is cooled until the polymer particles solidify. The resulting microspheres are washed by decanting with petroleum ether to produce a free flowing powder.
  • a non-miscible solvent like silicon oil
  • phase separation microencapsulation techniques a polymer solution is stirred, optionally in the presence of one or more active agents to be encapsulated. While continuing to uniformly suspend the material through stirring, a nonsolvent for the polymer is slowly added to the solution to decrease the polymer's solubility.
  • the polymer either precipitates or phase separates into a polymer rich and a polymer poor phase. Under proper conditions, the polymer in the polymer rich phase will migrate to the interface with the continuous phase, encapsulating the active agent(s) in a droplet with an outer polymer shell.
  • Spontaneous emulsifi cation involves solidifying emulsified liquid polymer droplets formed above by changing temperature, evaporating solvent, or adding chemical cross-linking agents.
  • One or more active agents to be incorporated are optionally added to the solution, and the mixture is suspended in an aqueous solution that contains a surface active agent such as poly(vinyl alcohol).
  • a surface active agent such as poly(vinyl alcohol).
  • the resulting emulsion is stirred until most of the organic solvent evaporated, leaving solid microparticles/nanoparticles. This method is useful for relatively stable polymers like polyesters and polystyrene.
  • the solvent removal microencapsulation technique is primarily designed for polyanhydrides and is described, for example, in WO 93/21906.
  • the substance to be incorporated is dispersed or dissolved in a solution of the selected polymer in a volatile organic solvent, such as methylene chloride. This mixture is suspended by stirring in an organic oil, such as silicon oil, to form an emulsion.
  • Microspheres that range between 1-300 microns can be obtained by this procedure.
  • Substances which can be incorporated in the microspheres include pharmaceuticals, pesticides, nutrients, imaging agents, and metal compounds.
  • Coacervation involves the separation of a macromolecular solution into two immiscible liquid phases.
  • One phase is a dense coacervate phase, which contains a high concentration of the polymer encapsulant (and optionally one or more active agents), while the second phase contains a low concentration of the polymer.
  • the polymer encapsulant forms nanoscale or microscale droplets.
  • Coacervation may be induced by a temperature change, addition of a non-solvent or addition of a micro-salt (simple coacervation), or by the addition of another polymer thereby forming an interpolymer complex (complex coacervation).
  • Particles can also be formed using the phase inversion nanoencapsulation (PIN) method, wherein a polymer is dissolved in a "good” solvent, fine particles of a substance to be incorporated, such as a drug, are mixed or dissolved in the polymer solution, and the mixture is poured into a strong non solvent for the polymer, to spontaneously produce, under favorable conditions, polymeric microspheres, wherein the polymer is either coated with the particles or the particles are dispersed in the polymer.
  • PIN phase inversion nanoencapsulation
  • the method can be used to produce monodisperse populations of nanoparticles and microparticles in a wide range of sizes, including, for example, about 100 nanometers to about 10 microns.
  • an emulsion need not be formed prior to precipitation.
  • the process can be used to form microspheres from thermoplastic polymers.
  • a particle is prepared using an emulsion solvent evaporation method.
  • a polymeric material is dissolved in a water immiscible organic solvent and mixed with a drug solution or a combination of drug solutions.
  • a solution of a therapeutic, prophylactic, or diagnostic agent to be encapsulated is mixed with the polymer solution.
  • the polymer can be, but is not limited to, one or more of the following: PLA, PGA, PCL, their copolymers, polyacrylates, the aforementioned PEGylated polymers.
  • the drug molecules can include one or more conjugates as described above and one or more additional active agents.
  • the water immiscible organic solvent can be, but is not limited to, one or more of the following: chloroform, dichloromethane, and acyl acetate.
  • the drug can be dissolved in, but is not limited to, one or more of the following: acetone, ethanol, methanol, isopropyl alcohol, acetonitrile and Dimethyl sulfoxide (DMSO).
  • DMSO Dimethyl sulfoxide
  • An aqueous solution is added into the resulting polymer solution to yield emulsion solution by emulsification.
  • the emulsification technique can be, but not limited to, probe sonication or homogenization through a homogenizer.
  • a conjugate containing nanoparticle is prepared using nanoprecipitation methods or microfluidic devices.
  • the conjugate containing polymeric material is mixed with a drug or drug combinations in a water miscible organic solvent, optionally containing additional polymers.
  • the additional polymer can be, but is not limited to, one or more of the following: PLA, PGA, PCL, their copolymers, polyacrylates, the aforementioned PEGylated polymers.
  • the water miscible organic solvent can be, but is not limited to, one or more of the following: acetone, ethanol, methanol, isopropyl alcohol, acetonitrile and dimethyl sulfoxide (DMSO).
  • DMSO dimethyl sulfoxide
  • the microfluidic device comprises at least two channels that converge into a mixing apparatus.
  • the channels are typically formed by lithography, etching, embossing, or molding of a polymeric surface.
  • a source of fluid is attached to each channel, and the application of pressure to the source causes the flow of the fluid in the channel.
  • the pressure may be applied by a syringe, a pump, and/or gravity.
  • lipid particles can be lipid micelles, liposomes, or solid lipid particles prepared using any suitable method known in the art.
  • Common techniques for created lipid particles encapsulating an active agent include, but are not limited to high pressure homogenization techniques, supercritical fluid methods, emulsion methods, solvent diffusion methods, and spray drying. A brief summary of these methods is presented below.
  • High pressure homogenization is a reliable and powerful technique, which is used for the production of smaller lipid particles with narrow size distributions, including lipid micelles, liposomes, and solid lipid particles.
  • High pressure homogenizers push a liquid with high pressure (100-2000 bar) through a narrow gap (in the range of a few microns).
  • the fluid can contain lipids that are liquid at room temperature or a melt of lipids that are solid at room temperature.
  • the fluid accelerates on a very short distance to very high velocity (over 1000 Km/h). This creates high shear stress and cavitation forces that disrupt the particles, generally down to the submicron range. Generally 5-10% lipid content is used but up to 40% lipid content has also been investigated.
  • Hot homogenization is carried out at temperatures above the melting point of the lipid and can therefore be regarded as the homogenization of an emulsion.
  • a pre-emulsion of the drug loaded lipid melt and the aqueous emulsifier phase is obtained by a high-shear mixing.
  • HPH of the pre-emulsion is carried out at temperatures above the melting point of the lipid.
  • a number of parameters, including the temperature, pressure, and number of cycles, can be adjusted to produce lipid particles with the desired size. In general, higher temperatures result in lower particle sizes due to the decreased viscosity of the inner phase. However, high temperatures increase the degradation rate of the drug and the carrier. Increasing the
  • homogenization pressure or the number of cycles often results in an increase of the particle size due to high kinetic energy of the particles.
  • Cold homogenization has been developed as an alternative to hot homogenization. Cold homogenization does not suffer from problems such as temperature-induced drug degradation or drug distribution into the aqueous phase during homogenization.
  • the cold homogenization is particularly useful for solid lipid particles, but can be applied with slight modifications to produce liposomes and lipid micelles.
  • the drug containing lipid melt is cooled, the solid lipid ground to lipid microparticles and these lipid microparticles are dispersed in a cold surfactant solution yielding a pre-suspension.
  • the pre-suspension is homogenized at or below room temperature, where the gravitation force is strong enough to break the lipid microparticles directly to solid lipid nanoparticles.
  • Lipid particles including lipid micelles, liposomes, and solid lipid particles, can be prepared by ultrasonication/high speed homogenization. The combination of both ultrasonication and high speed homogenization is particularly useful for the production of smaller lipid particles. Liposomes are formed in the size range from 10 nm to 200 nm, for example, 50 nm to 100 nm, by this process.
  • Lipid particles can be prepared by solvent evaporation approaches.
  • the lipophilic material is dissolved in a water-immiscible organic solvent (e.g.
  • cyclohexane that is emulsified in an aqueous phase.
  • particles dispersion is formed by precipitation of the lipid in the aqueous medium.
  • Parameters such as temperature, pressure, choices of solvents can be used to control particle size and distribution.
  • Solvent evaporation rate can be adjusted through increased/reduced pressure or increased/reduced temperature.
  • Lipid particles can be prepared by solvent emulsification-diffusion methods.
  • the lipid is first dissolved in an organic phase, such as ethanol and acetone.
  • An acidic aqueous phase is used to adjust the zeta potential to induce lipid
  • the continuous flow mode allows the continuous diffusion of water and alcohol, reducing lipid solubility, which causes thermodynamic instability and generates liposomes
  • Lipid particles can be prepared from supercritical fluid methods.
  • Supercritical fluid approaches have the advantage of replacing or reducing the amount of the organic solvents used in other preparation methods.
  • the lipids, active agents to be encapsulated, and excipients can be solvated at high pressure in a supercritical solvent.
  • the supercritical solvent is most commonly CO2, although other supercritical solvents are known in the art.
  • a small amount of co-solvent can be used.
  • Ethanol is a common co-solvent, although other small organic solvents that are generally regarded as safe for formulations can be used.
  • the lipid particles, lipid micelles, liposomes, or solid lipid particles can be obtained by expansion of the supercritical solution or by injection into a non-solvent aqueous phase.
  • the particle formation and size distribution can be controlled by adjusting the supercritical solvent, co-solvent, non- solvent, temperatures, pressures, etc.
  • Microemulsion based methods for making lipid particles are known in the art. These methods are based upon the dilution of a multiphase, usually two-phase, system. Emulsion methods for the production of lipid particles generally involve the formation of a water-in-oil emulsion through the addition of a small amount of aqueous media to a larger volume of immiscible organic solution containing the lipid. The mixture is agitated to disperse the aqueous media as tiny droplets throughout the organic solvent and the lipid aligns itself into a monolayer at the boundary between the organic and aqueous phases. The size of the droplets is controlled by pressure, temperature, the agitation applied and the amount of lipid present.
  • the water-in-oil emulsion can be transformed into a liposomal suspension through the formation of a double emulsion.
  • the organic solution containing the water droplets is added to a large volume of aqueous media and agitated, producing a water-in-oil-in-water emulsion.
  • the size and type of lipid particle formed can be controlled by the choice of and amount of lipid, temperature, pressure, co- surfactants, solvents, etc.
  • Spray drying methods similar to those described above for making polymeric particle can be employed to create solid lipid particles. Typically, this method is used with lipids with a melting point above 70°C.
  • conjugates of the present invention may be encapsulated in polymeric particles using a single oil in water emulsion method.
  • the conjugate and a suitable polymer or block copolymer or a mixture of polymers/block copolymers are dissolved in organic solvents such as, but not limited to, dichloromethane (DCM), ethyl acetate (EtAc) or choloform to form the oil phase.
  • organic solvents such as, but not limited to, dimethyl formamide (DMF), acetonitrile (CAN) or benzyl alcohol (BA) may be used to control the size of the particles and/or to solubilize the conjugate.
  • DCM dichloromethane
  • EtAc ethyl acetate
  • choloform choloform
  • Co-solvents such as, but not limited to, dimethyl formamide (DMF), acetonitrile (CAN) or benzyl alcohol (BA) may be used to control the size of the particles and/
  • particle formulations may be prepared by varying the lipophilicity of conjugates of the present invention.
  • the lipophilicity may be varied by using hydrophobic ion-pairs or hydrophobic ion-paring (HIP) of the conjugates with different counterions.
  • HIP alters the solubility of the conjugates of the present invention.
  • the aqueous solubility may drop and the solubility in organic phases may increase.
  • Any suitable agent may be used to provide counterions to form HIP complex with the conjugate of the present invention.
  • the HIP complex may be formed prior to formulation of the particles.
  • the conjugates or particles as described herein can be administered to treat any hyperproliferative disease, metabolic disease, infectious disease, or cancer, as appropriate.
  • the formulations can be used for immunization.
  • Formulations may be administered by injection, orally, or topically, typically to a mucosal surface (lung, nasal, oral, buccal, sublingual, vaginally, rectally) or to the eye (intraocularly or transocularly).
  • cancer embraces any disease or malady characterized by uncontrolled cell proliferation, e.g., hyperproliferation. Cancers may be characterized by tumors, e.g., solid tumors or any neoplasm.
  • the cancer is a solid tumor.
  • Large drug molecules have limited penetration in solid tumors. The penetration of large drug molecules is slow.
  • small molecules such as conjugates of the present invention may penetrate solid tumors rapidly and more deeply.
  • penetration depth of the drugs larger molecules penetrate less, despite having more durable
  • conjugates of the present invention may penetrate deep and rapidly into the core/center of the solid tumor.
  • conjugates of the present invention reach at least about 25 ⁇ , about 30 ⁇ , about 35 ⁇ , about 40 ⁇ , about 45 ⁇ , about 50 ⁇ , about 75 ⁇ , about 100 ⁇ , about 150 ⁇ , about 200 ⁇ , about 250 ⁇ , about 300 ⁇ , about 400 ⁇ , about 500 ⁇ , about 600 ⁇ , about 700 ⁇ , about 800 ⁇ , about 900 ⁇ , about 1000 ⁇ , about 1100 ⁇ , about 1200 ⁇ , about 1300 ⁇ , about 1400 ⁇ or about 1500 ⁇ into the solid tumor from the vascular surface of the tumor.
  • Zero distance is defined as the vascular surface of the tumor, and every distance greater than zero is defined as the distance measured in three dimensions to the nearest vascular surface.
  • conjugates of the present invention penetrate to the core of the tumor.
  • Core of the tumor refers to the central area of the tumor.
  • the distance from any part of the core area of the tumor to the vascular surface of the tumor is between about 30% to about 50% of the length or width of the tumor.
  • the distance from any part of the core area of the tumor to the center point of the tumor is less than about 20% of the length or width of the tumor.
  • the core area of the tumor is roughly the center 1/3 of the tumor.
  • conjugates of the present invention conjugates of the present invention penetrate to the middle of the solid tumor.
  • "Middle" of the tumor refers to the middle area of the tumor.
  • the distance from any part of the middle area of the tumor to the vascular surface of the tumor is between about 15%) and about 30%> of the length or the width of the tumor.
  • the distance from any part of the middle area of the tumor to the center point of the tumor is between about 20%) to about 35% of the length or width of the tumor.
  • the middle area of the tumor is roughly between the center 1/3 of the tumor and the outer 1/3 of the tumor.
  • the subject may be otherwise free of indications for treatment with the conjugates or particles.
  • methods include use of cancer cells, including but not limited to mammalian cancer cells. In some instances, the mammalian cancer cells are human cancer cells.
  • the conjugates or particles of the present teachings have been found to inhibit cancer and/or tumor growth. They may also reduce, including cell proliferation, invasiveness, and/or metastasis, thereby rendering them useful for the treatment of a cancer.
  • the conjugates or particles of the present teachings may be used to prevent the growth of a tumor or cancer, and/or to prevent the metastasis of a tumor or cancer.
  • compositions of the present teachings may be used to shrink or destroy a cancer.
  • the conjugates or particles provided herein are useful for inhibiting proliferation of a cancer cell.
  • the conjugates or particles provided herein are useful for inhibiting cellular proliferation, e.g., inhibiting the rate of cellular proliferation, preventing cellular proliferation, and/or inducing cell death.
  • the conjugates or particles as described herein can inhibit cellular proliferation of a cancer cell or both inhibiting proliferation and/or inducing cell death of a cancer cell.
  • cell proliferation is reduced by at least about 25%, about 50%, about 75%, or about 90% after treatment with conjguates or particles of the present invention compared with cells with no treatment.
  • cell cycle arrest marker phospho histone H3 (PH3 or PHH3) is increased by at least about 50%, about 75%, about 100%, about 200%, about 400%) or about 600% after treatment with conjguates or particles of the present invention compared with cells with no treatment.
  • cell apoptosis marker cleaved caspase-3 (CC3) is increased by at least 50%, about 75%, about 100%), about 200%, about 400% or about 600% after treatment with conjguates or particles of the present invention compared with cells with no treatment.
  • conjugates or particles of the present invention are effective for inhibiting tumor growth, whether measured as a net value of size (weight, surface area or volume) or as a rate over time, in multiple types of tumors.
  • the size of a tumor is reduced by about 60 % or more after treatment with conjugates or particles of the present invention. In some embodiments, the size of a tumor is reduced by at least about 20%, at least about 30%), at least about 40%, at least about 50%, at least about 60%>, at least about 70%, at least about 80%>, at least about 90%, at least about 95%, at least about 96%, at least about 97%), at least about 98%, at least about 99%, at least about 100%, by a measure of weight, and/or area and/or volume.
  • the cancers treatable by methods of the present teachings generally occur in mammals. Mammals include, for example, humans, non-human primates, dogs, cats, rats, mice, rabbits, ferrets, guinea pigs horses, pigs, sheep, goats, and cattle.
  • the cancer is lung cancer, breast cancer, e.g., mutant BRCA1 and/or mutant BRCA2 breast cancer, non-BRCA-associated breast cancer, colorectal cancer, ovarian cancer, pancreatic cancer, colorectal cancer, bladder cancer, prostate cancer, cervical cancer, renal cancer, leukemia, central nervous system cancers, myeloma, and melanoma.
  • the cancer is a neuroendocrine cancer such as but not limited to small cell lung cancer (SCLC), adrenal medullary tumors (e.g., pheochromocytoma, neuroblastoma, ganglioneuroma, or
  • gastroenteropancreatic neuroendocrine tumors e.g., carcinoids, gastrinoma, glucagonoma, vasoactive intestinal polypeptide-secreting tumor, pancreatic polypeptide-secreting tumor, or nonfunctioning gastroenteropancreatic tumors
  • meduallary thyroid cancer Merkel cell tumor of the skin, pituitary adenoma, and pancreatic cancer.
  • the neuroendocrine cancer is a primary
  • the neuroendocrine cancer is a neuroendocrine metastatsis.
  • Neuroendocrine metastatis may be in liver, lung, bone, or brain of a subject.
  • the cancer is brain cancer, human lung carcinoma, ovarian cancer, pancreatic cancer or colorectal cancer.
  • the conjugates or particles as described herein or formulations containing the conjugates or particles as described herein are used to treat small cell lung cancer.
  • About 12%- 15% of patients having lung cancer have small cell lung cancer. Survival in metastatic small cell lung caner is poor. Survival rate is below 5% five years after diagnosis. US incidence of small cell lung cancer is about 26K-30K. Among these patients, about 40%-80% are SSTR2 positive.
  • the conjugates or particles as described herein or formulations containing the conjugates or particles as described herein are used to treat paitents with tumors that express or over-express somatostatn receptor, NTSR1, or lLHRa.
  • a feature of conjugates or particles of the present invention is relatively low toxicity to an organism while maintaining efficacy at inhibiting, e.g. slowing or stopping tumor growth.
  • toxicity refers to the capacity of a substance or composition to be harmful or poisonous to a cell, tissue organism or cellular environment.
  • Low toxicity refers to a reduced capacity of a substance or composition to be harmful or poisonous to a cell, tissue organism or cellular environment.
  • Such reduced or low toxicity may be relative to a standard measure, relative to a treatment or relative to the absence of a treatment.
  • conjugates or particles of the present invention may have lower toxicity than the active agent moiety Z
  • Toxicity may further be measured relative to a subject's weight loss where weight loss over 15%, over 20% or over 30% of the body weight is indicative of toxicity.
  • Other metrics of toxicity may also be measured such as patient presentation metrics including lethargy and general malaiase.
  • Neutropenia, thrombopenia, white blood cell (WBC) count, complete blood cell (CBC) count may also be metrics of toxicity.
  • Pharmacologic indicators of toxicity include elevated aminotransferases (AST/ ALT) levels, neurotoxicity, kidney damage, GI damage and the like.
  • conjugates or particles of the present invention do not cause a significant change of a subject's body weight.
  • the body weight loss of a subject is less about 30%, about 20%, about 15%, about 10%, or about 5% after treatment with conjguates or particles of the present invention.
  • conjugates or particles of the present invention do not cause a significant increase of a subject's AST/ ALT levels.
  • the AST or ALT level of a subject is increased by less than about 30%), about 20%), about 15%, about 10%, or about 5% after treatment with conjugates or particles of the present invention.
  • conjugates or particles of the present invention do not cause a significant change of a subject's CBC or WBC count after treatment with conjugates or particles of the present invention.
  • conjugates or particles of the present invention are combined with at least one addtional active agent.
  • the active agent may be any suitable drug. It may be selected from any active agent described herein such as a drug for treating cancer. It may also be a cancer symptom relief drug. Non-limiting examples of symptom relief drugs include: octreotide or lanreotide; interferon, cypoheptadine or any other antihistamines.
  • conjugates or particles of the present invention do not have drug-drug interference with the additional active agent.
  • conjugates or particles of the present invention do not inhibit cytochrome P450 (CYP) isozymes.
  • CYP isozymes may include CYP3A4 Midazolam, CYP3A4 Testosterone, CYP2C9, CYP2D6, CYP1A2, CYP2C8, CYP2B6, and CYP2C19.
  • the additional active agent may be administered concomitantly with conjguates or particles of the present invention.
  • the additional active agent may not bind to any somatostatin receptor.
  • the additional active agent is a cancer symptom relief drug.
  • the symptom relief drug may reduce diarrhea or the side effects of chemotherapy or radiation therapy.
  • conjugates or particles of the present invention may be combined with a symptom relief drug for carcinoid symdrome, such as telotristat or telotristat etiprate (LX1032, Lexicon®).
  • Telotristat etiprate is telotristat's crystalline hippurate salt as disclosed in WO2013059146 to Chen et al., the contents of which are incorporated herein by reference in their entirety.
  • Telotristat its salts and crystalline forms can be obtained by methods known in the art (see US 7709493 to Devasagayaraj et al., the contents of which are incorporated herein by reference in their entirety). Any other compound disclosed in US 7709493 may be combined with conjugates or particles of the present invention.
  • conjugates or particles of the present invention may be combined with a moderate dose of chemotherapy agents such as mitomycin C, vinblastine and cisplatin (see Ellis et al., Br J Cancer, vol.71(2): 366-370 (1995), the contents of which are incorporated herein by reference in their entirety).
  • chemotherapy agents such as mitomycin C, vinblastine and cisplatin
  • the conjugates or particles as described herein or formulations containing the conjugates or particles as described herein can be used for the selective tissue delivery of a therapeutic, prophylactic, or diagnostic agent to an individual or patient in need thereof.
  • DM1 conjugates or particles of the present invention are used to deliver DM1 to selective tissues. These tissues may be tumor tissues. Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic.
  • a conjugate contained within a particle is released in a controlled manner.
  • the release can be in vitro or in vivo.
  • particles can be subject to a release test under certain conditions, including those specified in the U.S. Pharmacopeia and variations thereof.
  • less than about 90%, less than about 80%, less than about 70%, less than about 60%, less than about 50%, less than about 40%, less than about 30%, less than about 20% of the conjugate contained within particles is released in the first hour after the particles are exposed to the conditions of a release test. In some embodiments, less that about 90%, less than about 80%, less than about 70%), less than about 60%, or less than about 50% of the conjugate contained within particles is released in the first hour after the particles are exposed to the conditions of a release test. In certain embodiments, less than about 50% of the conjugate contained within particles is released in the first hour after the particles are exposed to the conditions of a release test.
  • the conjugate contained within a particle administered to a subject may be protected from a subject's body, and the body may also be isolated from the conjugate until the conjugate is released from the particle.
  • the conjugate may be substantially contained within the particle until the particle is delivered into the body of a subject. For example, less than about 90%, less than about 80%, less than about 70%, less than about 60%, less than about 50%, less than about 40%, less than about 30%, less than about 20%), less than about 15%, less than about 10%, less than about 5%, or less than about 1%) of the total conjugate is released from the particle prior to the particle being delivered into the body, for example, a treatment site, of a subject.
  • the conjugate may be released over an extended period of time or by bursts (e.g., amounts of the conjugate are released in a short period of time, followed by a periods of time where substantially no conjugate is released).
  • the conjugate can be released over 6 hours, 12 hours, 24 hours, or 48 hours. In certain embodiments, the conjugate is released over one week or one month.
  • kits and devices for conveniently and/or effectively carrying out methods of the present invention.
  • kits will comprise sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.
  • kits for inhibiting tumor cell growth in vitro or in vivo comprising a conjugate and/or particle of the present invention or a combination of conjugates and/or particles of the present invention, optionally in combination with any other active agents.
  • the kit may further comprise packaging and instructions and/or a delivery agent to form a formulation composition.
  • the delivery agent may comprise a saline, a buffered solution, or any delivery agent disclosed herein.
  • the amount of each component may be varied to enable consistent, reproducible higher concentration saline or simple buffer formulations.
  • the components may also be varied in order to increase the stability of the conjugates and/or particles in the buffer solution over a period of time and/or under a variety of conditions.
  • the present invention provides for devices which may incorporate conjugates and/or particles of the present invention. These devices contain in a stable formulation available to be immediately delivered to a subject in need thereof, such as a human patient. In some embodiments, the subject has cancer.
  • Non-limiting examples of the devices include a pump, a catheter, a needle, a transdermal patch, a pressurized olfactory delivery device, iontophoresis devices, multi-layered microfluidic devices.
  • the devices may be employed to deliver conjugates and/or particles of the present invention according to single, multi- or split-dosing regiments.
  • the devices may be employed to deliver conjugates and/or particles of the present invention across biological tissue, intradermal,
  • conjugate as used herein, is also meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • Compounds of the present disclosure also include all of the isotopes of the atoms occurring in the intermediate or final compounds.
  • “Isotopes” refers to atoms having the same atomic number but different mass numbers resulting from a different number of neutrons in the nuclei.
  • isotopes of hydrogen include tritium and deuterium.
  • the compounds and salts of the present disclosure can be prepared in combination with solvent or water molecules to form solvates and hydrates by routine methods.
  • subject or "patient”, as used herein, refer to any organism to which the particles may be administered, e.g., for experimental, therapeutic, diagnostic, and/or prophylactic purposes.
  • Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, guinea pigs, cattle, pigs, sheep, horses, dogs, cats, hamsters, lamas, non-human primates, and humans).
  • treating can include preventing a disease, disorder or condition from occurring in an animal that may be predisposed to the disease, disorder and/or condition but has not yet been diagnosed as having the disease, disorder or condition; inhibiting the disease, disorder or condition, e.g., impeding its progress; and relieving the disease, disorder, or condition, e.g., causing regression of the disease, disorder and/or condition.
  • Treating the disease, disorder, or condition can include ameliorating at least one symptom of the particular disease, disorder, or condition, even if the underlying pathophysiology is not affected, such as treating the pain of a subject by administration of an analgesic agent even though such agent does not treat the cause of the pain.
  • a target shall mean a site to which targeted constructs bind.
  • a target may be either in vivo or in vitro.
  • a target may be cancer cells found in leukemias or tumors (e.g., tumors of the brain, lung (small cell and non-small cell), ovary, prostate, breast and colon as well as other carcinomas and sarcomas).
  • a target may refer to a molecular structure to which a targeting moiety or ligand binds, such as a hapten, epitope, receptor, dsDNA fragment, carbohydrate or enzyme.
  • a target may be a type of tissue, e.g., neuronal tissue, intestinal tissue, pancreatic tissue, liver, kidney, prostate, ovary, lung, bone marrow, or breast tissue.
  • the "target cells” that may serve as the target for the method or conjugates or particles are generally animal cells, e.g., mammalian cells.
  • the present method may be used to modify cellular function of living cells in vitro, i.e., in cell culture, or in vivo, in which the cells form part of or otherwise exist in animal tissue.
  • the target cells may include, for example, the blood, lymph tissue, cells lining the alimentary canal, such as the oral and pharyngeal mucosa, cells forming the villi of the small intestine, cells lining the large intestine, cells lining the respiratory system (nasal passages/lungs) of an animal (which may be contacted by inhalation of the subject invention), dermal/epidermal cells, cells of the vagina and rectum, cells of internal organs including cells of the placenta and the so-called blood/brain barrier, etc.
  • a target cell expresses at least one type of SSTR.
  • a target cell can be a cell that expresses an SSTR and is targeted by a conjugate described herein, and is near a cell that is affected by release of the active agent of the conjugate.
  • a blood vessel expressing an SSTR that is in proximity to a tumor may be the target, while the active agent released at the site will affect the tumor.
  • therapeutic effect is art-recognized and refers to a local or systemic effect in animals, particularly mammals, and more particularly humans caused by a pharmacologically active substance.
  • the term thus means any substance intended for use in the diagnosis, cure, mitigation, treatment or prevention of disease, disorder or condition in the enhancement of desirable physical or mental development and conditions in an animal, e.g., a human.
  • modulation is art-recognized and refers to up regulation (i.e., activation or stimulation), down regulation (i.e., inhibition or suppression) of a response, or the two in combination or apart.
  • the modulation is generally compared to a baseline or reference that can be internal or external to the treated entity.
  • parenteral administration means administration by any method other than through the digestive tract (enteral) or non-invasive topical routes.
  • parenteral administration may include administration to a patient intravenously, intradermally, intraperitoneally, intrapleurally, intratracheally, intraossiously, intracerebrally, intrathecally, intramuscularly, subcutaneously, subjunctivally, by injection, and by infusion.
  • Topical administration means the non-invasive administration to the skin, orifices, or mucosa.
  • Topical administration can be delivered locally, i.e., the therapeutic can provide a local effect in the region of delivery without systemic exposure or with minimal systemic exposure.
  • Some topical formulations can provide a systemic effect, e.g., via adsorption into the blood stream of the individual.
  • Topical administration can include, but is not limited to, cutaneous and transdermal administration, buccal administration, intranasal administration, intravaginal administration, intravesical administration, ophthalmic administration, and rectal administration.
  • Enteral administration means administration via absorption through the gastrointestinal tract. Enteral administration can include oral and sublingual administration, gastric administration, or rectal administration.
  • Pulmonary administration means administration into the lungs by inhalation or endotracheal administration.
  • endotracheal administration means administration into the lungs by inhalation or endotracheal administration.
  • inhalation refers to intake of air to the alveoli.
  • the intake of air can occur through the mouth or nose.
  • a “therapeutically effective amount” is at least the minimum concentration required to effect a measurable improvement or prevention of at least one symptom or a particular condition or disorder, to effect a measurable enhancement of life expectancy, or to generally improve patient quality of life. The therapeutically effective amount is thus dependent upon the specific biologically active molecule and the specific condition or disorder to be treated.
  • therapeutically effective amounts of many active agents, such as antibodies, are known in the art.
  • compositions described herein, e.g., for treating specific disorders may be determined by techniques that are well within the craft of a skilled artisan, such as a physician.
  • bioactive agent and “active agent”, as used interchangeably herein, include, without limitation, physiologically or pharmacologically active substances that act locally or systemically in the body.
  • a bioactive agent is a substance used for the treatment (e.g., therapeutic agent), prevention (e.g.,
  • prophylactic agent diagnosis (e.g., diagnostic agent), cure or mitigation of disease or illness, a substance which affects the structure or function of the body, or pro-drugs, which become biologically active or more active after they have been placed in a predetermined physiological environment.
  • prodrug refers to an agent, including a small organic molecule, peptide, nucleic acid or protein, that is converted into a biologically active form in vitro and/or in vivo.
  • Prodrugs can be useful because, in some situations, they may be easier to administer than the parent compound (the active compound). For example, a prodrug may be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have improved solubility in pharmaceutical compositions compared to the parent drug. A prodrug may also be less toxic than the parent.
  • a prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. Harper, N.J. (1962) Drug Latentiation in Jucker, ed.
  • biocompatible refers to a material that along with any metabolites or degradation products thereof that are generally non-toxic to the recipient and do not cause any significant adverse effects to the recipient.
  • biocompatible materials are materials which do not elicit a significant inflammatory or immune response when administered to a patient.
  • biodegradable generally refers to a material that will degrade or erode under physiologic conditions to smaller units or chemical species that are capable of being metabolized, eliminated, or excreted by the subject.
  • the degradation time is a function of composition and morphology. Degradation times can be from hours to weeks.
  • pharmaceutically acceptable refers to compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio, in accordance with the guidelines of agencies such as the U.S. Food and Drug
  • a "pharmaceutically acceptable carrier”, as used herein, refers to all components of a pharmaceutical formulation that facilitate the delivery of the composition in vivo.
  • Pharmaceutically acceptable carriers include, but are not limited to, diluents, preservatives, binders, lubricants, disintegrators, swelling agents, fillers, stabilizers, and combinations thereof.
  • molecular weight generally refers to the mass or average mass of a material. If a polymer or oligomer, the molecular weight can refer to the relative average chain length or relative chain mass of the bulk polymer. In practice, the molecular weight of polymers and oligomers can be estimated or characterized in various ways including gel permeation chromatography (GPC) or capillary viscometry. GPC molecular weights are reported as the weight-average molecular weight (M w ) as opposed to the number-average molecular weight (M n ).
  • Capillary viscometry provides estimates of molecular weight as the inherent viscosity determined from a dilute polymer solution using a particular set of concentration, temperature, and solvent conditions.
  • small molecule generally refers to an organic molecule that is less than 2000 g/mol in molecular weight, less than 1500 g/mol, less than 1000 g/mol, less than 800 g/mol, or less than 500 g/mol. Small molecules are non-polymeric and/or non-oligomeric.
  • hydrophilic refers to substances that have strongly polar groups that readily interact with water.
  • hydrophobic refers to substances that lack an affinity for water; tending to repel and not absorb water as well as not dissolve in or mix with water.
  • lipophilic refers to compounds having an affinity for lipids.
  • amphiphilic refers to a molecule combining hydrophilic and lipophilic (hydrophobic) properties.
  • Amphiphilic material refers to a material containing a hydrophobic or more hydrophobic oligomer or polymer (e.g., biodegradable oligomer or polymer) and a hydrophilic or more hydrophilic oligomer or polymer.
  • targeting moiety refers to a moiety that binds to or localizes to a specific locale.
  • the moiety may be, for example, a protein, nucleic acid, nucleic acid analog, carbohydrate, or small molecule.
  • the locale may be a tissue, a particular cell type, or a subcellular compartment.
  • a targeting moiety can specifically bind to a selected molecule.
  • reactive coupling group refers to any chemical functional group capable of reacting with a second functional group to form a covalent bond.
  • the selection of reactive coupling groups is within the ability of those in the art.
  • Examples of reactive coupling groups can include primary amines (- H2) and amine-reactive linking groups such as isothiocyanates, isocyanates, acyl azides, NHS esters, sulfonyl chlorides, aldehydes, glyoxals, epoxides, oxiranes, carbonates, aryl halides, imidoesters, carbodiimides, anhydrides, and fluorophenyl esters.
  • reactive coupling groups can include aldehydes (-COH) and aldehyde reactive linking groups such as hydrazides, alkoxyamines, and primary amines.
  • reactive coupling groups can include thiol groups (-SH) and sulfhydryl reactive groups such as maleimides, haloacetyls, and pyridyl disulfides.
  • reactive coupling groups can include photoreactive coupling groups such as aryl azides or diazirines.
  • the coupling reaction may include the use of a catalyst, heat, pH buffers, light, or a combination thereof.
  • protective group refers to a functional group that can be added to and/or substituted for another desired functional group to protect the desired functional group from certain reaction conditions and selectively removed and/or replaced to deprotect or expose the desired functional group.
  • Protective groups are known to the skilled artisan. Suitable protective groups may include those described in Greene and Wuts, Protective Groups in Organic Synthesis, (1991). Acid sensitive protective groups include dimethoxytrityl (DMT), tert- butylcarbamate (tBoc) and trifluoroacetyl (tFA).
  • Base sensitive protective groups include 9- fluorenylmethoxycarbonyl (Fmoc), isobutyrl (iBu), benzoyl (Bz) and phenoxyacetyl (pac).
  • Other protective groups include acetamidomethyl, acetyl, tert- amyloxycarbonyl, benzyl, benzyloxycarbonyl, 2-(4-biph8nylyl)-2-propy!oxycarbonyl, 2- bromobenzyloxycarbonyl, tert-butyh tert-butyloxycarbonyl, 1-carbobenzoxamido- 2,2.2- trifluoroethyl, 2,6-dichlorobenzyl, 2-(3,5-dimethoxyphenyl)-2- propyloxycarbonyl, 2,4- dinitrophenyl, dithiasuccinyl, formyl, 4- methoxybenzenesulfonyl, 4-methoxybenzyl
  • activated ester refers to alkyl esters of carboxylic acids where the alkyl is a good leaving group rendering the carbonyl susceptible to nucleophilic attack by molecules bearing amino groups. Activated esters are therefore susceptible to aminolysis and react with amines to form amides. Activated esters contain a carboxylic acid ester group -CO2R where R is the leaving group.
  • alkyl refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl -substituted cycloalkyl groups, and cycloalkyl-substituted alkyl groups.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C30 for straight chains, C3-C30 for branched chains), 20 or fewer, 12 or fewer, or 7 or fewer.
  • cycloalkyls have from 3-10 carbon atoms in their ring structure, e.g., have 5, 6 or 7 carbons in the ring structure.
  • alkyl (or “lower alkyl) as used throughout the specification, examples, and claims is intended to include both “unsubstituted alkyls” and “substituted alkyls”, the latter of which refers to alkyl moieties having one or more substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents include, but are not limited to, halogen, hydroxyl, carbonyl (such as a carboxyl, alkoxycarbonyl, formyl, or an acyl), thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), alkoxyl, phosphoryl, phosphate, phosphonate, a hosphinate, amino, amido, amidine, imine, cyano, nitro, azido, sulfhydryl, alkylthio, sulfate, sulfonate, sulfamoyl, sulfonamido, sulfonyl, heterocyclyl, aralkyl, or an aromatic or heteroaromatic moiety.
  • carbonyl such as a carboxyl, alkoxycarbonyl, formyl, or an acyl
  • thiocarbonyl such as a thioester, a
  • lower alkyl as used herein means an alkyl group, as defined above, but having from one to ten carbons, or from one to six carbon atoms in its backbone structure. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths. In some embodiments, alkyl groups are lower alkyls. In some embodiments, a substituent designated herein as alkyl is a lower alkyl.
  • the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate.
  • the substituents of a substituted alkyl may include halogen, hydroxy, nitro, thiols, amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), -CF 3 , -CN and the like. Cycloalkyls can be substituted in the same manner.
  • heteroalkyl refers to straight or branched chain, or cyclic carbon-containing radicals, or combinations thereof, containing at least one heteroatom. Suitable heteroatoms include, but are not limited to, O, N, Si, P, Se, B, and S, wherein the phosphorous and sulfur atoms are optionally oxidized, and the nitrogen heteroatom is optionally quaternized. Heteroalkyls can be substituted as defined above for alkyl groups.
  • alkylthio refers to an alkyl group, as defined above, having a sulfur radical attached thereto.
  • the "alkylthio" moiety is represented by one of -S-alkyl, -S-alkenyl, and -S-alkynyl.
  • Representative alkylthio groups include methylthio, and ethylthio.
  • alkylthio also encompasses cycloalkyl groups, alkene and cycloalkene groups, and alkyne groups.
  • Arylthio refers to aryl or heteroaryl groups. Alkylthio groups can be substituted as defined above for alkyl groups.
  • alkenyl and alkynyl refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • alkoxyl or "alkoxy” as used herein refers to an alkyl group, as defined above, having an oxygen radical attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propyloxy, and tert-butoxy.
  • An "ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as can be represented by one of -O-alkyl, -O-alkenyl, and -O-alkynyl.
  • Aroxy can be represented by -O-aiyl or O-heteroaiyl, wherein aryl and heteroaryl are as defined below.
  • the alkoxy and aroxy groups can be substituted as described above for alkyl.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that can be represented by the general formula:
  • R9, Rio, and Rio each independently represent a hydrogen, an alkyl, an alkenyl, -(CH2)m-Rs or R9 and Rio taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure;
  • Rs represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a polycycle;
  • m is zero or an integer in the range of 1 to 8.
  • only one of R9 or Rio can be a carbonyl, e.g., R9, Rio and the nitrogen together do not form an imide.
  • the term "amine” does not encompass amides, e.g., wherein one of R9 and Rio represents a carbonyl.
  • R9 and Rio each independently represent a hydrogen, an alkyl or cycloalkly, an alkenyl or cycloalkenyl, or alkynyl.
  • alkylamine as used herein means an amine group, as defined above, having a substituted (as described above for alkyl) or unsubstituted alkyl attached thereto, i.e., at least one of R9 and Rio is an alkyl group.
  • amido is art-recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula: wherein R9 and Rio are as defined above.
  • Aryl refers to Cs-Cio-membered aromatic, heterocyclic, fused aromatic, fused heterocyclic, biaromatic, or bihetereocyclic ring systems.
  • aryl includes 5-, 6-, 7-, 8-, 9-, and 10-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • aryl heterocycles or "heteroaromatics”.
  • the aromatic ring can be substituted at one or more ring positions with one or more substituents including, but not limited to, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino (or quaternized amino), nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, -CF3, -CN; and combinations thereof.
  • substituents including, but not limited to, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino (or quaternized amino),
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (i.e., "fused rings") wherein at least one of the rings is aromatic, e.g., the other cyclic ring or rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocycles.
  • heterocyclic rings include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-l,5,2-dithiazinyl, dihydrofuro[2,3
  • aralkyl refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).
  • carrier refers to an aromatic or non- aromatic ring in which each atom of the ring is carbon.
  • Heterocycle refers to a cyclic radical attached via a ring carbon or nitrogen of a monocyclic or bicyclic ring containing 3-10 ring atoms, for example, from 5-6 ring atoms, consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(Y) wherein Y is absent or is H, O, (Ci-Cio) alkyl, phenyl or benzyl, and optionally containing 1-3 double bonds and optionally substituted with one or more substituents.
  • heterocyclic rings include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-l,5,2-dithiazinyl, dihydrofuro[2,3-£]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, indolenyl, indolinyl, ind
  • Heterocyclic groups can optionally be substituted with one or more substituents at one or more positions as defined above for alkyl and aryl, for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, and -CN.
  • substituents at one or more positions as defined above for alkyl and aryl, for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imin
  • carbonyl is art-recognized and includes such moieties as can be represented by the general formula:
  • X is a bond or represents an oxygen or a sulfur
  • Ru represents a hydrogen, an alkyl, a cycloalkyl, an alkenyl, an cycloalkenyl, or an alkynyl
  • R'n represents a hydrogen, an alkyl, a cycloalkyl, an alkenyl, an cycloalkenyl, or an alkynyl
  • X is an oxygen and Ru or R' n is not hydrogen
  • the formula represents an "ester”.
  • X is an oxygen and Ru is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when Ru is a hydrogen, the formula represents a "carboxylic acid".
  • monoester refers to an analog of a dicarboxylic acid wherein one of the carboxylic acids is functionalized as an ester and the other carboxylic acid is a free carboxylic acid or salt of a carboxylic acid.
  • monoesters include, but are not limited to, to monoesters of succinic acid, glutaric acid, adipic acid, suberic acid, sebacic acid, azelaic acid, oxalic and maleic acid.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Examples of heteroatoms are boron, nitrogen, oxygen, phosphorus, sulfur and selenium. Other useful heteroatoms include silicon and arsenic.
  • nitro means -NO2; the term “halogen” designates -F, -CI, -Br or -I; the term “sulfhydryl” means -SH; the term “hydroxyl” means -OH; and the term “sulfonyl” means -SO2-.
  • substituted refers to all permissible substituents of the compounds described herein.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, but are not limited to, halogens, hydroxyl groups, or any other organic groupings containing any number of carbon atoms, for example, 1-14 carbon atoms, and optionally include one or more heteroatoms such as oxygen, sulfur, or nitrogen grouping in linear, branched, or cyclic structural formats.
  • substituents include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halo, hydroxyl, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aroxy, substituted aroxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, polyaryl
  • Heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. It is understood that “substitution” or “substituted” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, i.e., a compound that does not spontaneously undergo
  • transformation for example, by rearrangement, cyclization, or elimination.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described herein.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms.
  • the substituent is selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide, and thioketone, each of which optionally is substituted with one or more suitable substituents.
  • the substituent is selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cycloalkyl, ester, ether, formyl, haloalkyl, heteroaryl, heterocyclyl, ketone, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide, and thioketone, wherein each of the alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cycloalkyl, ester, ether, formyl, haloalkyl, heteroaryl, heterocyclyl, ketone, phosphate, sulfide, sulfinyl, sulfony
  • substituents include, but are not limited to, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, thioketone, ester, heterocyclyl, - CN, aryl, aryloxy, perhaloalkoxy, aralkoxy, heteroaryl, heteroaryloxy,
  • heteroarylalkyl heteroaralkoxy, azido, alkylthio, oxo, acylalkyl, carboxy esters, carboxamido, acyloxy, aminoalkyl, alkylaminoaryl, alkylaryl, alkylaminoalkyl, alkoxyaryl, arylamino, aralkylamino, alkylsulfonyl, carboxamidoalkylaryl, carboxamidoaryl, hydroxyalkyl, haloalkyl, alkylaminoalkylcarboxy, aminocarboxamidoalkyl, cyano, alkoxyalkyl, perhaloalkyl, arylalkyloxyalkyl, and the like.
  • the substituent is selected from cyano, halogen, hydroxyl, and nitro.
  • copolymer generally refers to a single polymeric material that is comprised of two or more different monomers.
  • the copolymer can be of any form, for example, random, block, or graft.
  • the copolymers can have any end- group, including capped or acid end groups.
  • mean particle size generally refers to the statistical mean particle size (diameter) of the particles in the composition.
  • the diameter of an essentially spherical particle may be referred to as the physical or hydrodynamic diameter.
  • the diameter of a non-spherical particle may refer to the hydrodynamic diameter.
  • the diameter of a non-spherical particle may refer to the largest linear distance between two points on the surface of the particle.
  • Mean particle size can be measured using methods known in the art such as dynamic light scattering. Two populations can be said to have a "substantially equivalent mean particle size" when the statistical mean particle size of the first population of particles is within 20% of the statistical mean particle size of the second population of particles; for example, within 15%, or within 10%.
  • monodisperse and “homogeneous size distribution”, as used interchangeably herein, describe a population of particles, microparticles, or nanoparticles all having the same or nearly the same size.
  • a monodisperse distribution refers to particle distributions in which 90% of the distribution lies within 5% of the mean particle size.
  • polypeptide generally refer to a polymer of amino acid residues. As used herein, the term also applies to amino acid polymers in which one or more amino acids are chemical analogs or modified derivatives of corresponding naturally-occurring amino acids or are unnatural amino acids.
  • protein refers to a polymer of amino acids linked to each other by peptide bonds to form a polypeptide for which the chain length is sufficient to produce tertiary and/or quaternary structure.
  • protein excludes small peptides by definition, the small peptides lacking the requisite higher- order structure necessary to be considered a protein.
  • nucleic acid refers to a deoxyribonucleotide or ribonucleotide polymer, in linear or circular conformation, and in either single- or double-stranded form. These terms are not to be construed as limiting with respect to the length of a polymer.
  • the terms can encompass known analogs of natural nucleotides, as well as nucleotides that are modified in the base, sugar and/or phosphate moieties (e.g., phosphorothioate backbones).
  • nucleic acid is a term of art that refers to a string of at least two base-sugar-phosphate monomeric units. Nucleotides are the monomeric units of nucleic acid polymers. The term includes deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) in the form of a messenger RNA, antisense, plasmid DNA, parts of a plasmid DNA or genetic material derived from a virus.
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • nucleic acids refers to a string of at least two base-sugar- phosphate combinations. Natural nucleic acids have a phosphate backbone. Artificial nucleic acids may contain other types of backbones, but contain the same bases as natural nucleic acids. The term also includes PNAs (peptide nucleic acids), phosphorothioates, and other variants of the phosphate backbone of native nucleic acids.
  • a "functional fragment" of a protein, polypeptide or nucleic acid is a protein, polypeptide or nucleic acid whose sequence is not identical to the full-length protein, polypeptide or nucleic acid, yet retains at least one function as the full-length protein, polypeptide or nucleic acid.
  • a functional fragment can possess more, fewer, or the same number of residues as the corresponding native molecule, and/or can contain one or more amino acid or nucleotide substitutions.
  • the DNA binding function of a polypeptide can be determined, for example, by filter-binding, electrophoretic mobility shift, or immunoprecipitation assays. DNA cleavage can be assayed by gel electrophoresis.
  • the ability of a protein to interact with another protein can be determined, for example, by co-immunoprecipitation, two-hybrid assays or complementation, e.g., genetic or biochemical. See, for example, Fields et al. (1989) Nature 340:245-246; U.S. Patent No. 5,585,245 and PCT WO 98/44350.
  • linker refers to a carbon chain that can contain heteroatoms (e.g., nitrogen, oxygen, sulfur, etc.) and which may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 atoms long.
  • Linkers may be substituted with various substituents including, but not limited to, hydrogen atoms, alkyl, alkenyl, alkynl, amino, alkylamino, dialkylamino,
  • linkers include, but are not limited to, pH-sensitive linkers, protease cleavable peptide linkers, nuclease sensitive nucleic acid linkers, lipase sensitive lipid linkers, glycosidase sensitive carbohydrate linkers, hypoxia sensitive linkers, photo-cleavable linkers, heat-labile linkers, enzyme cleavable linkers (e.g., esterase cleavable linker), ultrasound-sensitive linkers, and x-ray cleavable linkers.
  • pH-sensitive linkers protease cleavable peptide linkers
  • nuclease sensitive nucleic acid linkers include lipase sensitive lipid linkers, glycosidase sensitive carbohydrate linkers, hypoxia sensitive linkers, photo-cleavable linkers, heat-labile linkers, enzyme cleavable linkers (e.g., esterase cleavable linker), ultrasound-sensitive linkers, and x-ray cleavable linkers.
  • pharmaceutically acceptable counter ion is a pharmaceutically acceptable ion.
  • the pharmaceutically acceptable counter ion is selected from citrate, malate, acetate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate (i.e., 1, 1'- methylene-bis-(2-hydroxy-3-naphthoate)).
  • pharmaceutically acceptable counter ion is selected from chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, citrate, malate, acetate, oxalate, acetate, and lactate.
  • the pharmaceutically acceptable counter ion is selected from chloride, bromide, iodide, nitrate, sulfate, bisulfate, and phosphate.
  • salts refers to salts of acidic or basic groups that may be present in compounds used in the present compositions.
  • Compounds included in the present compositions that are basic in nature are capable of forming a variety of salts with various inorganic and organic acids.
  • the acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including but not limited to sulfate, citrate, malate, acetate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate (i
  • Compounds included in the present compositions that include an amino moiety may form pharmaceutically acceptable salts with various amino acids, in addition to the acids mentioned above.
  • Compounds included in the present compositions, that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations.
  • Examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts.
  • the free base can be obtained by basifying a solution of the acid salt.
  • an addition salt particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.
  • Those skilled in the art will recognize various synthetic methodologies that may be used to prepare non-toxic pharmaceutically acceptable addition salts.
  • a pharmaceutically acceptable salt can be derived from an acid selected from l-hydroxy-2-naphthoic acid, 2,2-dichloroacetic acid, 2-hydroxyethanesulfonic acid, 2-oxoglutaric acid, 4-acetamidobenzoic acid, 4-aminosalicylic acid, acetic acid, adipic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid (decanoic acid), caproic acid (hexanoic acid), caprylic acid (octanoic acid), carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecyl sulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, glu
  • pyroglutamic acid salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tartaric acid, thiocyanic acid, toluenesulfonic acid, trifluoroacetic, and undecylenic acid.
  • bioavailable is art-recognized and refers to a form of the subject invention that allows for it, or a portion of the amount administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered.
  • Fmoc-cystine(Trt) was loaded onto 2-chlorotrityl resin (3.6 g, 1 mmol/g). Iterative deprotection with 4: 1 DMF:piperidine and coupling subsequently with Fmoc-threonine(tBu), Na-Fmoc-Ns-Boc-lysine, Na-Fmoc-N in -Boc-D-tryptophan, Fmoc-tyrosine(tBu), Fmoc-cysteine(Trt), and Fmoc-D-phenylalanine, and a final DMF:piperidine deprotection provided the protected linear peptide on resin.
  • the resin was washed with dichloromethane (3 x 40 mL), then cleaved with tnfluoroacetic acid (40 mL), water (2 mL), and triisopropylsilane (2 mL).
  • the resin was stirred in the deprotection cocktail for 30 min, drained, the resin washed with dichloromethane (40 mL), and the deprotection/washing sequence repeated once more.
  • the collected washings were concentrated in vacuo, and the remaining residue was dissolved in THF (70 mL). 30% hydrogen peroxide (0.65 mL) was added, followed by adding saturated sodium carbonate until reaction pH measured 7.0 (25 mL saturated sodium bicarbonate).
  • Fmoc-cystine(Trt) was loaded onto 2-chlorotrityl resin (875 mg, 1 mmol/g). Iterative deprotection with 4: 1 DMF:piperidine and coupling subsequently with Fmoc-threonine(tBu), Na-Fmoc-Ns-Boc-lysine, Na-Fmoc-N in -Boc-D-tryptophan, Fmoc-tyrosine(tBu), Fmoc-cysteine(Trt), and Fmoc-D-phenylalanine, deprotection, and two treatments each with ethyl isocyanate (0.69 mL, 8.8 mmol) and triethylamine (3.0 mL) in DMF (20 mL) for 2 h gave the protected linear peptide.
  • the resin was washed with dichloromethane (3 x 40 mL), then cleaved with trifluoroacetic acid (20 mL), water (1 mL), and triisopropylsilane (1 mL).
  • the resin was stirred in the deprotection cocktail for 30 min, drained, the resin washed with dichloromethane (20 mL), and the deprotection/washing sequence repeated once more.
  • the collected washings were concentrated in vacuo, and the remaining residue was dissolved in THF (50 mL). 30% hydrogen peroxide (0.32 mL) was added, followed by adding saturated sodium carbonate until reaction pH measured 8.0 (12 mL saturated sodium bicarbonate).
  • Fmoc-cystine(Trt) was loaded onto 2-chlorotrityl resin (30.0 g, 1 mmol/g theoretical loading). Iterative deprotection with 4: 1 DMF:piperidine and coupling subsequently with Fmoc-threonine(tBu), Na-Fmoc-Ns-Boc-lysine, Na-Fmoc-N in - Boc-D-tryptophan, Fmoc-tyrosine(tBu), Fmoc-cysteine(Trt), and Boc-D- phenylalanine to give 90.2 g of the protected linear peptide (60.2 g total peptide loaded, 0.369 mmol/g loading of final protected resin).
  • the resin was drained, washed with DMF (2 x 25 mL) and dichloromethane (4 x 20 mL). The resin was then treated with 4: 1 dichloromethane:hexafluoroisopropanol (60 mL) for 1 h. The dichloromethane: hexafluoroisopropanol solution was collected, the resin washed with dichloromethane (25 mL), and treated again with 4: 1
  • dichloromethane hexafluoroisopropanol (60 mL) for 1 h.
  • Fmoc-cystine(Trt) was loaded onto 2-chlorotrityl resin. Iterative deprotection with 4: 1 DMF:piperidine and coupling subsequently with Fmoc- threonine(tBu), Na-Fmoc-Ns-Boc-lysine, Na-Fmoc-N in -Boc-D-tryptophan, Fmoc- tyrosine(tBu), and Fmoc-cysteine(Trt) provided the Fmoc-capped linear peptide.
  • the resin was washed with dichloromethane (3 x 40 mL), then cleaved with 90:5:5 trifluoroacetic acid, water, and triisopropylsilane.
  • the resin was stirred in the deprotection cocktail for 30 min, drained, the resin washed with dichloromethane, and the deprotection/washing sequence repeated once more.
  • the collected washings were concentrated in vacuo, and the remaining residue was dissolved in THF. 30% hydrogen peroxide was added, followed by adding saturated sodium carbonate until reaction pH measured 8.0. Di-tert-butyl dicarbonate was added, and the reaction stirred at room temperature for 16 h.
  • Fmoc-cystine(Trt) was loaded onto 2-chlorotrityl resin. Iterative deprotection with 4: 1 DMF:piperidine and coupling subsequently with Fmoc- threonine(tBu), Na-Fmoc-Ns-Boc-lysine, Na-Fmoc-N in -Boc-D-tryptophan, Fmoc- tyrosine(tBu), Fmoc-cysteine(Trt), and Fmoc-D-phenylalanine provided the protected linear peptide.
  • Fmoc-leucine was loaded onto 2-chlorotrityl resin (4.0 g, 0.5 mmol/g, 2 mmol loading). Iterative deprotection with 4: 1 DMF :piperi dine and coupling with 3 equiv each HBTU, N-methyl morpholine, and in sequence, Fmoc-tert-leucine, Fmoc- tyrosine(tBu), Fmoc-proline, Fmoc-arginine(Pbf), Fmoc-arginine(Pbf), Fmoc-proline, Fmoc-penicillamine(Trt), and finally acetic anhydride provided the protected linear peptide.
  • the resin was treated with trifluoroacetic acid (95 ml), water (2.5 mL), EDT (2.5 mL), and triisopropylsilane (2.5 mL) for 2 h.
  • the deprotection cocktail was drained into a flask, ether (800 mL) added, and the resulting precipitate was filtered, the solid collected, and purified by reverse phase chromatography to give 23 as the bis-TFA salt (1.18 g, 0.897 mmol, 45% yield).
  • Fmoc-cysteine(Trt) was loaded onto 2-chlorotrityl resin (7.27 g, 0.344 mmol/g loading, 2.5 mmol). Iterative deprotection with 4: 1 DMF:piperidine and coupling with 3 equiv each FIBTU and N-methyl morpholine and, in sequence, Fmoc- serine(tBu), Fmoc-glycine, Fmoc-glycine, Fmoc-alanine, Fmoc-arginine(Pbf), Fmoc- arginine(Pbf), Fmoc-glycine, Fmoc-cysteine(Trt), and finally capping with acetic anhydride provided the linear peptide.
  • a vial was charged with 25 (246 mg, 0.271 mmol), S-trityl cysteine amide (174 mg, 0.406 mmol), diisopropylcarbodiimide (68 mg, 0.54 mmol), HOBt (73 mg, 0.54 mmol), and DMF (5 mL) and diisopropylethylamine (105 mg, 0.813 mmol) were added.
  • the reaction was stirred at room temperature for 40 h, and the resulting reaction mixture purified by preparative HPLC to give 51 (78.1 mg, 0.0625 mmol, 23% yield).
  • LCMS M/Z 1250.3 [M + 1].
  • a vial was charged with 52 (12.8 mg, 10.0 ⁇ ), and this was dissolved in trifluoroethanol (0.1 mL) and 1, 1,3,3-tetramethyldisiloxane (13 ⁇ .).
  • a solution of 12N HCI (13 ⁇ .) in trifluoroethanol (0.1 mL) was added, and the reaction stirred at room temperature for 15 min. All solvent was removed in vacuo, and the remaining residue dissolved in DMF (0.6 mL). This solution was added to 1 (12.7 mg, 15.0 ⁇ ), and 0.2M sodium acetate (0.6 mL) was added. The reaction stirred at room temperature for 1 h, and LCMS shows conversion to the desired product.
  • a vial was charged with 27 (556 mg, 0.366 mmol), and water (0.05 mL), triisopropylsilane (0.05 mL) and trifluoroacetic acid (2.0 mL) were added. The reaction was stirred at room temperature for 10 min, and all solvents were removed in vacuo. To the remaining residue was added a solution of 1 (387 mg, 0.457 mmol) in DMF (4 mL). To this solution was then added pH 7.4 phosphate buffer (4.0 mL) and 0.2M Na 2 HP04 (0.60 mL) dropwise, in that order, over 5 min. The reaction was stirred at room temperature for 1 h, and the reaction judged complete by LCMS.
  • EXAMPLE 2 Ki of conjugates for somatostatin receptor
  • Membranes were incubated with radiolabeled somatostatin (0.03 nM) in the presence of conjugate/compound starting at a dose of either 10 ⁇ (for compounds with a capping group on Lys 5 , 117 and 143) or 10 nM (all other compounds) using 5x serial dilutions to obtain a 6-pt curve. After a four hour incubation, membranes were filtered and washed 3x and counted to determine the remaining [ 125 I] somatostatin bound to the receptor. ICso values were determined by a non-linear, least squares regression analysis using MathlQTM (ID Business Solutions Ltd., UK).
  • the Ki values were calculated using the equation of Cheng and Prusoff (Cheng and Prusoff, Biochem. Pharmacol. 22:3099-3108, 1973) using the observed ICso of the tested conjugate/compound, the concentration of radioligand employed in the assay, and the historical values for the Ki of the ligand obtained at Eurofins.
  • Conjugates were assessed in an in vitro assay evaluating inhibition of cell proliferation.
  • NCI-H524 (ATCC) human lung cancer cells were plated in 96 well, V- bottomed plates (Costar) at a concentration of 5,000 cells/well and 24 hours later were treated with compound for 6 hours and further incubated 66 hours.
  • Compound starting dose was 20 ⁇ and three fold serial dilutions were done for a total of ten points. After 6 hours of treatment, cells were spun down, the drug containing media was removed, and fresh complete medium was added and used to resuspend the cells, which were spun again.
  • the cells were resuspended in complete medium, then transferred into white walled, flat bottomed 96 well plates. Cells were further incubated for an additional 66 hours to measure inhibition of cell proliferation. Octreotide alone had no significant effect on cell proliferation.
  • % inhibition (control- treatment)/ control * 100. Control is defined as vehicle alone.
  • ICso curves were generated using the nonlinear regression analysis (four parameter) with GraphPad Prism 6. ICso values for representative compounds with and without octreotide competition were measured and were shown in Table 4.
  • 1 - Assay shift is the ratio of ICso with octreotide to the ICso without octreotide
  • Conjugates were assessed in an in vitro assay evaluating inhibition of cell proliferation.
  • IMR-32(ATCC), human neuroblastoma, cells were plated in 96 well, flat-bottomed plates (Costar) at a concentration of 4,000 cells/well. Cells were then incubated at 37°C with 5% CO2 for 24hrs. After 24hrs, cells were treated with either 100 ⁇ Octreotide or 0.01% DMSO, and incubated for lhr at 37°C with 5% CO2. Compounds were then added at a starting dose of 20 ⁇ and three fold serial dilutions were done for a total of ten points. Following compound addition cells were incubated at 37°C with 5% CO2 for 6hrs.
  • 1 - Assay shift is the ratio of ICso with octreotide to the ICso without octreotide
  • Claims or descriptions that include “or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context.
  • the invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process.
  • the invention includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process.
  • the term “comprising” is intended to be open and permits but does not require the inclusion of additional elements or steps. When the term “comprising” is used herein, the term “consisting of is thus also encompassed and disclosed.

Landscapes

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

Abstract

L'invention concerne des conjugués d'un agent actif attaché à une fraction de ciblage par l'intermédiaire d'un lieur de pénicillamine, et des particules comprenant de tels conjugués. Ces conjugués et particules peuvent conférer une stabilité accrue et permettre une administration spatiotemporelle améliorée de l'agent actif, une biodistribution et une pénétration améliorées dans la tumeur, et/ou une toxicité réduite. L'invention concerne des procédés de préparation des conjugués, des particules et de leurs formulations. L'invention concerne en outre des procédés d'administration des formulations à un sujet en ayant besoin, par exemple, pour traiter ou prévenir un cancer.
PCT/US2017/035107 2016-05-31 2017-05-31 Conjugués de pénicillamine et particules et formulations associées WO2017210246A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662343445P 2016-05-31 2016-05-31
US62/343,445 2016-05-31

Publications (2)

Publication Number Publication Date
WO2017210246A2 true WO2017210246A2 (fr) 2017-12-07
WO2017210246A3 WO2017210246A3 (fr) 2017-12-28

Family

ID=60479055

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/035107 WO2017210246A2 (fr) 2016-05-31 2017-05-31 Conjugués de pénicillamine et particules et formulations associées

Country Status (1)

Country Link
WO (1) WO2017210246A2 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108440664A (zh) * 2018-03-27 2018-08-24 上海欣科医药有限公司 一种用于癌症检测的生长抑素类似物及其制备方法和应用
WO2019118830A1 (fr) * 2017-12-14 2019-06-20 Tarveda Therapeutics, Inc. Conjugués ciblant hsp90 et formulations de ces derniers
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
WO2021005583A1 (fr) * 2019-07-11 2021-01-14 Sun Pharma Advanced Research Company Ltd. Dérivés de camptothécine ayant une fraction disulfure et une fraction pipérazine
CN113307754A (zh) * 2021-06-01 2021-08-27 上海吉奉生物科技有限公司 一种l-青霉胺盐酸盐的合成方法
WO2022035843A1 (fr) * 2020-08-12 2022-02-17 Nanomedicine Innovation Center, Llc Inhibiteurs de topoisomérase
WO2022099565A1 (fr) * 2020-11-12 2022-05-19 苏州大学 Sonde fluorescente de type à ancrage d'acide nucléique à médiation par la lumière rouge, son procédé de préparation et son utilisation
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014124227A1 (fr) * 2013-02-07 2014-08-14 Immunomedics, Inc. Forme de pro-médicament (p2pdox) de la 2-pyrrolinodoxorubicine fortement puissante conjuguée à des anticorps pour la thérapie ciblée du cancer
BR212016030926U2 (pt) * 2014-06-30 2018-05-29 Tarveda Therapeutics Inc conjugados de alvo e partículas e formulações dos mesmos

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11110178B2 (en) 2016-07-07 2021-09-07 The Board Of Trustees Of The Leland Standford Junior University Antibody adjuvant conjugates
US11547761B1 (en) 2016-07-07 2023-01-10 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
WO2019118830A1 (fr) * 2017-12-14 2019-06-20 Tarveda Therapeutics, Inc. Conjugués ciblant hsp90 et formulations de ces derniers
JP2021506797A (ja) * 2017-12-14 2021-02-22 ターベダ セラピューティクス インコーポレイテッドTarveda Therapeutics,Inc. Hsp90標的化コンジュゲート及びその製剤
CN108440664A (zh) * 2018-03-27 2018-08-24 上海欣科医药有限公司 一种用于癌症检测的生长抑素类似物及其制备方法和应用
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2
WO2021005583A1 (fr) * 2019-07-11 2021-01-14 Sun Pharma Advanced Research Company Ltd. Dérivés de camptothécine ayant une fraction disulfure et une fraction pipérazine
WO2022035843A1 (fr) * 2020-08-12 2022-02-17 Nanomedicine Innovation Center, Llc Inhibiteurs de topoisomérase
WO2022099565A1 (fr) * 2020-11-12 2022-05-19 苏州大学 Sonde fluorescente de type à ancrage d'acide nucléique à médiation par la lumière rouge, son procédé de préparation et son utilisation
CN113307754A (zh) * 2021-06-01 2021-08-27 上海吉奉生物科技有限公司 一种l-青霉胺盐酸盐的合成方法

Also Published As

Publication number Publication date
WO2017210246A3 (fr) 2017-12-28

Similar Documents

Publication Publication Date Title
US11458206B2 (en) Targeted conjugates and particles and formulations thereof
US20220096640A1 (en) Hsp90-targeting conjugates and formulations thereof
US20220096646A1 (en) Sstr-targeted conjugates and particles and formulations thereof
US20200078468A1 (en) Neurotensin receptor binding conjugates and formulations thereof
WO2017210246A2 (fr) Conjugués de pénicillamine et particules et formulations associées
WO2019118830A1 (fr) Conjugués ciblant hsp90 et formulations de ces derniers
US20190125888A1 (en) Targeted conjugates and particles and formulations thereof
US20200237746A1 (en) Hsp90 targeted conjugates and particle formulations thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17807369

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17807369

Country of ref document: EP

Kind code of ref document: A2