WO2021119423A1 - Near infrared fluorescent dyes, formulations and related methods - Google Patents

Near infrared fluorescent dyes, formulations and related methods Download PDF

Info

Publication number
WO2021119423A1
WO2021119423A1 PCT/US2020/064506 US2020064506W WO2021119423A1 WO 2021119423 A1 WO2021119423 A1 WO 2021119423A1 US 2020064506 W US2020064506 W US 2020064506W WO 2021119423 A1 WO2021119423 A1 WO 2021119423A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
childhood
cell
tumor
pharmaceutical composition
Prior art date
Application number
PCT/US2020/064506
Other languages
French (fr)
Inventor
Samuel Achilefu
Rui Tang
Duanwen SHEN
Avik SOM
Baogang XU
Gail SUDLOW
Christopher EGBULEFU
Partha Karmakar
Kexian LIANG
Original Assignee
Washington University
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 Washington University filed Critical Washington University
Priority to EP20897916.1A priority Critical patent/EP4072598A4/en
Publication of WO2021119423A1 publication Critical patent/WO2021119423A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0032Methine dyes, e.g. cyanine dyes
    • A61K49/0034Indocyanine green, i.e. ICG, cardiogreen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0032Methine dyes, e.g. cyanine dyes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0056Peptides, proteins, polyamino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0063Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres
    • A61K49/0069Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form
    • A61K49/0073Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form semi-solid, gel, hydrogel, ointment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0063Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres
    • A61K49/0069Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form
    • A61K49/0076Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form dispersion, suspension, e.g. particles in a liquid, colloid, emulsion
    • A61K49/0084Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form dispersion, suspension, e.g. particles in a liquid, colloid, emulsion liposome, i.e. bilayered vesicular structure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0063Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres
    • A61K49/0069Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form
    • A61K49/0089Particulate, powder, adsorbate, bead, sphere
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09BORGANIC DYES OR CLOSELY-RELATED COMPOUNDS FOR PRODUCING DYES, e.g. PIGMENTS; MORDANTS; LAKES
    • C09B67/00Influencing the physical, e.g. the dyeing or printing properties of dyestuffs without chemical reactions, e.g. by treating with solvents grinding or grinding assistants, coating of pigments or dyes; Process features in the making of dyestuff preparations; Dyestuff preparations of a special physical nature, e.g. tablets, films
    • C09B67/0071Process features in the making of dyestuff preparations; Dehydrating agents; Dispersing agents; Dustfree compositions
    • C09B67/0083Solutions of dyes
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09BORGANIC DYES OR CLOSELY-RELATED COMPOUNDS FOR PRODUCING DYES, e.g. PIGMENTS; MORDANTS; LAKES
    • C09B67/00Influencing the physical, e.g. the dyeing or printing properties of dyestuffs without chemical reactions, e.g. by treating with solvents grinding or grinding assistants, coating of pigments or dyes; Process features in the making of dyestuff preparations; Dyestuff preparations of a special physical nature, e.g. tablets, films
    • C09B67/0071Process features in the making of dyestuff preparations; Dehydrating agents; Dispersing agents; Dustfree compositions
    • C09B67/0084Dispersions of dyes
    • C09B67/0085Non common dispersing agents
    • C09B67/009Non common dispersing agents polymeric dispersing agent
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09BORGANIC DYES OR CLOSELY-RELATED COMPOUNDS FOR PRODUCING DYES, e.g. PIGMENTS; MORDANTS; LAKES
    • C09B69/00Dyes not provided for by a single group of this subclass
    • C09B69/10Polymeric dyes; Reaction products of dyes with monomers or with macromolecular compounds
    • C09B69/101Polymeric dyes; Reaction products of dyes with monomers or with macromolecular compounds containing an anthracene dye
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09BORGANIC DYES OR CLOSELY-RELATED COMPOUNDS FOR PRODUCING DYES, e.g. PIGMENTS; MORDANTS; LAKES
    • C09B69/00Dyes not provided for by a single group of this subclass
    • C09B69/10Polymeric dyes; Reaction products of dyes with monomers or with macromolecular compounds
    • C09B69/105Polymeric dyes; Reaction products of dyes with monomers or with macromolecular compounds containing a methine or polymethine dye
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions

Definitions

  • TECHNICAL FIELD [0003] The present disclosure provides pharmaceutical formulations comprising LS301 or LS838, fluorescent dye-cyclic polypeptide conjugates (dye-conjugates).
  • the formulations herein are suitable for systemic delivery, such as intravenous injection, topical application, and oral gavage.
  • BACKGROUND [0004] Elevated chronic inflammatory milieu, metabolic aberrations, and genetic mutations drive cancer cells’ dynamic adaptation toward their survival, proliferation, and metastasis. Accompanying alterations in cellular processes produce heterogeneous populations of cancer subtypes and distorted stroma characterized by diverse cancer biomarkers. Progress in cancer targeted therapies and imaging relies on effectively and selectively delivering molecules to overexpressed cell surface proteins.
  • ANXA2 annexin A2
  • PTMs cancer- associated posttranslational modifications
  • pANXA2 Phosphorylation of ANXA2 at tyrosine 23 modulates ANXA2 tetramer formation and is a prerequisite for its 1 KC0004-401-PC 019279/WO translocation to the plasma membrane. This PTM occurs in response to growth factor signaling and promotes cancer cell migration and invasion by activating cytoskeletal rearrangements and epithelial-mesenchymal transition.
  • pANXA2 is an inducible hallmark of diverse solid tumor microenvironments in small animal models and primary human cancer tissues, with its expression confined to tumor regions having elevated calcium levels.
  • pANXA2 binds and stabilizes the plasminogen receptor S100A10/p11, which associates with tissue plasminogen activator (tPA) and plasminogen to generate plasmin.
  • tPA tissue plasminogen activator
  • ANXA2 is highly activated (phosphorylated) irrespective of the solid tumor type.
  • pANXA2 is expressed in subtypes of breast cancers, including Er+ and triple-negative cancers, but not in the healthy breast tissue from the same cancer patients.
  • Most ANXA2-based drug delivery strategies rely on the overexpression of ANXA2 in certain tumors.
  • non-tumor tissues also express sufficiently elevated levels to impair selectivity, leading to pre-imaging and tissue biopsy to determine the drugs’ usefulness for treating specific tumors.
  • NIR near-infrared
  • LS301 and LS838 are excellent NIR cancer imaging probes, especially, as disclosed herein, for their specificity toward pANXA2.
  • the clinical formulation for a hydrophobic optical probe should meet several conditions, including adequate and accurate dosing, bioavailability, targeting efficacy, and appropriate absorption and fluorescence characteristics.
  • the long circulation time of LS301 and LS838 delays the tumor uptake and clearance from non-tumor tissue.
  • the formulations described herein accelerate tumor uptake, allowing for quick clinical intervention within an hour post-administration.
  • SUMMARY [0008] The present disclosure provides stable, sustainable formulations of LS301 and LS838 NIR imaging probes for various biological applications, including lyophilized product, IV injection, topical application, and oral gavage delivery, and doxorubicin conjugates of LS301 and LS838 for targeted cancer treatment.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate- Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838), or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration; a divalent metal ion; and a pharmaceutically acceptable carrier.
  • the present disclosure provides a lyophilized product comprising a dye-conjugate chosen from cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate- Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838) or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration, and albumin, wherein the dye-conjugate and the albumin are in a 1:50 (w/w) ratio.
  • a dye-conjugate chosen from cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate- Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838) or
  • the present disclosure also provides aa vial comprising about 102 mg of the lyophilized product disclosed herein.
  • the present disclosure further provides an injectable solution comprising about 102 mg of the lyophilized product disclosed herein and about 10 mL of phosphate-buffered saline.
  • the present disclosure further provides a method for identifying compromised fibroblasts, comprising administering an effective amount of any pharmaceutical composition disclosed herein to a subject in need thereof.
  • the present disclosure also provides a method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with any pharmaceutical composition disclosed herein.
  • pANXA2 phosphorylated annexin A2
  • the present disclosure provides doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser- Pro-Cys)-Lys-OH) and salts thereof, and pharmaceutical compositions thereof.
  • cancer is treated in a subject in need thereof, comprising administering an effective amount of doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and/or salts thereof or a pharmaceutical composition of any one doxorubicin-cypate- cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and/or salts thereof to the subject.
  • the accumulation rate, tissue uptake selectivity, and amount of product retained in target tissue are improved.
  • the disclosure is based on the findings that formulating some drugs and imaging agents in uptake excipients or the presence of ionic solutions, such as an electrolyte, can improve the cellular internalization and biodistribution of these products.
  • a general embodiment of this disclosure relates to the formulation of dyes, dye-like molecules, and cyclic or aromatic drugs with excipients such as albumin, polyethylene glycol (PEG), cyclodextrin, and surfactants.
  • the enabling ionic species include cations and anions.
  • Cations include monovalent and multivalent cations such as Na, K, Ba, Ca, Mg, Al, Cu, Fe, and Zr.
  • Anions can be monovalent or multivalent, including chlorides, carbonates, nitrates, cyanines, and sulfonates.
  • An embodiment includes the formulation of LS301 in calcium or magnesium.
  • Another embodiment is the formulation of 3 KC0004-401-PC 019279/WO LS301 in albumin or PEG or dimethylsulfoxide.
  • LS301 is a cyclic octapeptide labeled with a near-infrared dye which exhibits selective and high-affinity calcium-dependent binding to phosphorylated annexin A2 (pANXA2) protein in diverse solid tumors.
  • the LS301 preferentially binds to the invasive edges of tumors, a process amplified by cancer cell-induced pANXA2 expression in tumor- associated stromal cells. Further, it traffics within macrophages to the necrotic core of tumors. High levels of pANXA2 and calcium are present in most solid cancers’ microenvironment, revealing a pathway to image and selectively deliver drugs to tumors.
  • the divalent metal ion is chosen from Ca 2+ , Mg 2+ , and Mn 2+ , for example, Ca 2+ .
  • the divalent metal ion is present at a concentration between 1 mM and 10 mM, such as 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, or 10 mM. In certain embodiments, the divalent metal ion is present at a concentration of 5 mM.
  • the cypate is .
  • In certain embod residues is D-Cys.
  • the compound is LS301 and comprises the structural formula 4 KC0004-401-PC 019279/WO .
  • the compound is LS301 comprising the structural formula .
  • LS301 selectively binds to pANXA2 over the non-activated ANXA2. Histopathology of tissue samples from mice administered with LS301 in vivo showed that the compound accumulates in pANXA2-positive cancer cells. Cancer cells induce pANXA2 expression in tumor-associated fibroblasts and macrophages to stimulate LS301 accumulation in these cells at the periphery and core. By detecting pANXA2-associated cells in the tumor 5 KC0004-401-PC 019279/WO microenvironment, LS301 targets and delivers drugs to multiple types of solid tumors.
  • LS301 The preferential localization of LS301 at the proliferating edge and the inner core of solid tumors defines tumor margins, improves cancer resection accuracy during surgery, and treats cancer simultaneously from the periphery and interior core of the tumor.
  • Uptake of LS301 into the solid cancer cell lines for lung (A549), breast (MDA-MB- 231 and 4T1), and myeloid leukemia (HL60) reached a maximum concentration at 12 hours, which maintained over 24 hours. Cypate alone exhibited only transient retention in these cell lines.
  • LS301 binds to lipid rafts in the cellular plasma membrane, which endocytosed via a clathrin-dependent pathway after one hour of incubation.
  • the pharmaceutical composition comprises albumin.
  • the cypate dye moiety of LS301 reversibly binds to the hydrophobic pockets of albumin. Without wishing to be bound by theory, this albumin binding is likely not the primary cause of LS301 tumor uptake. Tumors did not retain cypate in vivo with or without formulation with albumin. Rather, the albumin is a nitrogen source for growing tumors.
  • the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin.
  • the albumin is human serum albumin (HSA). In certain embodiments, the albumin is mouse serum albumin (MSA). In certain embodiments, the albumin is bovine serum albumin (BSA).
  • LS301 preferentially targets ANXA2 over ANXA1 and ANXA3. ANXA2 calcium- dependently associates with cholesterol-rich lipid rafts at cell surfaces. LS301 also localizes to these lipids rafts and binds calcium with an apparent dissociation constant (K d ) of 5.67 ⁇ 2.40 nM as determined by microscale thermophoresis (MST).
  • the apparent Kd for LS301 binding to pANXA2, ANXA2, and ANXA3 proteins was 0.075 ⁇ 0.002 nM; 0.389 ⁇ 0.015 nM; and 6.128 ⁇ 0.280 nM, respectively via MST.
  • LS301 preferentially bound to lysates of insulin-stimulated, pANXA2-expressing 4T1 cells with an apparent Kd of 0.017 ⁇ 0.004 nM.
  • NIR fluorescence microscopy showed that LS301 substantially colocalized with pANXA2 in LS301-treated A549 and 4T1 cells after 1-hour incubation.
  • Tissue plasminogen activator (tPA) protein binds to a site in the N-terminal domain of ANXA2 comprising the amino acid 6 KC0004-401-PC 019279/WO sequence LCKLSL.
  • a synthetic LCKLSL peptide inhibited the binding and internalization of LS301 in cells.
  • LS301 Adding LS301 to fibroblast co-cultured with ANXA2-positive GFP-expressing 4T1 cells resulted in LS301 uptake by tumor cells and fibroblasts. By increasing the population of pANXA2-positive cells for tumor tissue, cancer cells amplify LS301 accumulation. Taken together, these results demonstrate that LS301 is selective for pANXA2, regardless of the cell or cancer type. [0029] LS301 selectively accumulated in a tumor from a breast cancer patient-derived xenograft and remained in the lesion for over 96 hours.
  • the pharmaceutical composition comprises the compound LS838.
  • LS838 comprises the structural formula la 7 KC0004-401-PC 019279/WO .
  • PDAC pancreatic intraepithelial neoplasia
  • PanlN pancreatic intraepithelial neoplasia
  • Uptake is mediated by the energy needs of these metabolically active cells.
  • LS838 is then trapped intracellularly under the highly reducing environment of these cells.
  • applying LS838 to distinguish PDAC and to transform PanlN from chronic pancreatitis with high accuracy significantly progresses treatment for these cancers.
  • LS838 enables NIR fluorescence to detect microscopic lesions not visible with prior clinical imaging techniques.
  • LS838 fluoresces more brightly than LS301, allowing smaller amounts of LS838 to achieve the same uptake kinetics as LS301.
  • the placement of the tyrosine in the LS838 molecule is important for retaining LS838 in tumors.
  • LS838 can be radiolabeled at its tyrosine residue, enabling combined intravital fluorescence microscopy and noninvasive imaging.
  • the radionuclide is chosen from, for example, fluorine-18, iodine-123, iodine-124, iodine-125, and iodine-131.
  • chemotherapeutics such as doxorubicin
  • LS838 selectively remains in diverse tumors 8 KC0004-401-PC 019279/WO without significant loss of fluorescence over time.
  • the high specificity of LS838 for cancer cells in the presence of healthy white blood cells allows the same agent to detect circulating tumor cells (CTCs) without additional tagging or expensive antibodies.
  • the pharmaceutically acceptable carrier comprises phosphate- buffered saline.
  • the pharmaceutical composition is administered intravenously.
  • the pharmaceutical composition is administered topically, for example, to the subject’s colon.
  • the pharmaceutical composition is administered orally.
  • the effective amount is up to about 0.6 ⁇ mol/kg including 0.1 ⁇ mol/kg, 0.2 ⁇ mol/kg, 0.3 ⁇ mol/kg, 0.4 ⁇ mol/kg, 0.5 ⁇ mol/kg or 0.6 ⁇ mol/kg.
  • the present disclosure further provides a method for identifying compromised fibroblasts, comprising administering an effective amount of the pharmaceutical composition described herein to a subject in need thereof.
  • the compromised fibroblasts are proximal to dormant cancer cells.
  • the cancer is chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lympho
  • the cancer is acute lymphoblastic leukemia. In some embodiments, the cancer is acute myeloid leukemia. In some embodiments, the cancer is adrenocortical carcinoma. In some embodiments, the cancer is AIDS-related cancer, such as AIDS-related lymphoma. In some embodiments, the cancer is anal cancer. In some embodiments, the cancer is appendix cancer. In some embodiments, the cancer is astrocytomas (childhood cerebellar or cerebral). In some embodiments, the cancer is basal cell carcinoma. In some embodiments, the cancer is bile duct cancer. In some embodiments, the cancer is bladder cancer. In some embodiments, the cancer is bone cancer.
  • the cancer is brainstem glioma. In some embodiments, the cancer is brain tumors, such as cerebellar astrocytoma. In some embodiments, the cancer is cerebral astrocytoma/malignant glioma. In some embodiments, the cancer is ependymoma. In some embodiments, the cancer is medulloblastoma. In some embodiments, the cancer is supratentorial primitive neuroectodermal tumors. In some embodiments, the cancer is a visual pathway and hypothalamic gliomas. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is bronchial adenomas/carcinoids.
  • the cancer is Burkitt lymphoma. In some embodiments, the cancer is carcinoid tumors (childhood, gastrointestinal). In some embodiments, the cancer is carcinoma of an unknown primary. In some embodiments, the cancer is central nervous system lymphoma (primary). In some embodiments, the cancer is cerebellar astrocytoma. In some embodiments, the cancer is cerebral astrocytoma/malignant glioma. In some embodiments, the cancer is cervical cancer. In some embodiments, the cancer is childhood cancer. In some embodiments, the cancer is chronic lymphocytic leukemia. In some embodiments, the cancer is chronic myelogenous leukemia. In some embodiments, the cancer is a chronic myeloproliferative disorder.
  • the cancer is colon cancer. In some embodiments, the cancer is cutaneous T- cell lymphoma. In some embodiments, the cancer is a desmoplastic small round cell tumor. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is ependymoma. In some embodiments, the cancer is esophageal cancer. In some embodiments, the cancer is Ewing’s sarcoma in the Ewing family of tumors. In some embodiments, the cancer is an extracranial germ cell tumor (childhood). In some embodiments, the cancer is an extragonadal germ cell tumor. In some embodiments, the cancer is extrahepatic bile duct cancer.
  • the cancer is eye cancer, such as intraocular melanoma retinoblastoma.
  • the cancer is gallbladder cancer.
  • the cancer is gastric (stomach) cancer.
  • the cancer is a gastrointestinal carcinoid tumor.
  • the cancer is a gastrointestinal stromal tumor.
  • the cancer is germ cell tumors (childhood extracranial, extragonadal, ovarian).
  • the cancer is a gestational trophoblastic tumor.
  • the cancer is gliomas (adult, childhood brain stem.
  • the cancer is gastric carcinoid. In some embodiments, the cancer is hairy cell leukemia. In some embodiments, the cancer is head and neck cancer. In some embodiments, the cancer is hepatocellular (liver) cancer. In some embodiments, the cancer is Hodgkin lymphoma. In some embodiments, the cancer is hypopharyngeal cancer. In some embodiments, the cancer is hypothalamic and visual pathway glioma (childhood). In some embodiments, the cancer is intraocular melanoma. In some embodiments, the cancer is islet cell carcinoma. In some embodiments, the cancer is Kaposi sarcoma.
  • the cancer is kidney cancer (renal cell cancer). In some embodiments, the cancer is laryngeal cancer. In some embodiments, the cancer is leukemia, such as acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, or hairy cell. In some embodiments, the cancer is a lip and oral cavity cancer. In some embodiments, the cancer is liver cancer (primary). In some embodiments, the cancer is lung cancers, such as non-small cell lung cancer and small cell lung cancer. In some embodiments, the cancer is lymphoma, such as AIDS-related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, and primary central nervous system.
  • lymphoma such as AIDS-related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, and primary central nervous system.
  • the cancer is macroglobulinemia (Waldenström). In some embodiments, the cancer is malignant fibrous histiocytoma of bone/osteosarcoma. In some embodiments, the cancer is medulloblastoma (childhood). In some embodiments, the cancer is melanoma. In some embodiments, the cancer is intraocular melanoma. In some embodiments, the cancer is Merkel cell carcinoma. In some embodiments, the cancer is mesotheliomas, such as adult malignant and childhood. In some embodiments, the cancer is metastatic squamous neck cancer with occult primary. In some embodiments, the cancer is mouth cancer.
  • Waldenström macroglobulinemia
  • the cancer is malignant fibrous histiocytoma of bone/osteosarcoma.
  • the cancer is medulloblastoma (childhood).
  • the cancer is melanoma.
  • the cancer is intraocular melanoma.
  • the cancer is Merkel cell carcinoma
  • the cancer is multiple endocrine neoplasia syndrome (childhood). In some embodiments, the cancer is multiple myeloma/plasma cell neoplasm. In some embodiments, the cancer is mycosis fungoides. In some embodiments, the cancer is myelodysplastic syndromes. In some embodiments, the cancer is a myelodysplastic/ myeloproliferative disease. In some embodiments, the cancer is myelogenous leukemia (chronic). In some embodiments, the cancer is myeloid leukemias, such as adult acute and childhood acute. In 12 KC0004-401-PC 019279/WO some embodiments, the cancer is multiple myeloma.
  • the cancer is myeloproliferative disorders (chronic). In some embodiments, the cancer is a nasal cavity and paranasal sinus cancer. In some embodiments, the cancer is nasopharyngeal carcinoma. In some embodiments, the cancer is neuroblastoma. In some embodiments, the cancer is non- Hodgkin lymphoma. In some embodiments, the cancer is non-small cell lung cancer. In some embodiments, the cancer is oral cancer. In some embodiments, the cancer is oropharyngeal cancer. In some embodiments, the cancer is osteosarcoma/malignant fibrous histiocytoma of bone. In some embodiments, the cancer is ovarian cancer.
  • the cancer is ovarian epithelial cancer, such as a surface epithelial-stromal tumor. In some embodiments, the cancer is an ovarian germ cell tumor. In some embodiments, the cancer is an ovarian low malignant potential tumor. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is pancreatic cancer (islet cell). In some embodiments, the cancer is paranasal sinus and nasal cavity cancer. In some embodiments, the cancer is parathyroid cancer. In some embodiments, the cancer is penile cancer. In some embodiments, the cancer is pharyngeal cancer. In some embodiments, the cancer is pheochromocytoma. In some embodiments, the cancer is pineal astrocytoma.
  • the cancer is pineal germinoma. In some embodiments, the cancer is pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood). In some embodiments, the cancer is a pituitary adenoma. In some embodiments, the cancer is plasma cell neoplasia. In some embodiments, the cancer is pleuropulmonary blastoma. In some embodiments, the cancer is a primary central nervous system lymphoma. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is rectal cancer. In some embodiments, the cancer is renal cell carcinoma (kidney cancer). In some embodiments, the cancer is renal pelvis and ureter transitional cell cancer.
  • the cancer is retinoblastoma. In some embodiments, the cancer is rhabdomyosarcoma (childhood). In some embodiments, the cancer is salivary gland cancer. In some embodiments, the cancer is sarcoma, such as the Ewing family of tumors, Kaposi, soft tissue, and uterine. In some embodiments, the cancer is Sézary syndrome. In some embodiments, the cancer is skin cancers, such as nonmelanoma and melanoma. In some embodiments, the cancer is skin carcinoma (Merkel cell). In some embodiments, the cancer is small cell lung cancer. In some embodiments, the cancer is small intestine cancer. In some embodiments, the cancer is soft tissue sarcoma.
  • the cancer is squamous cell carcinoma. In some embodiments, the cancer is squamous neck cancer with occult primary (metastatic). In some embodiments, the cancer is 13 KC0004-401-PC 019279/WO stomach cancer. In some embodiments, the cancer is a supratentorial primitive neuroectodermal tumor (childhood). In some embodiments, the cancer is T-Cell lymphoma (cutaneous). In some embodiments, the cancer is testicular cancer. In some embodiments, the cancer is throat cancer. In some embodiments, the cancer is thymoma (childhood). In some embodiments, the cancer is thymoma and thymic carcinoma. In some embodiments, the cancer is thyroid cancer.
  • the cancer is thyroid cancer (childhood).
  • the cancer is transitional cell cancer of the renal pelvis and ureter.
  • the cancer is a trophoblastic tumor (gestational).
  • the cancer is an unknown primary site, such as adult and childhood.
  • the cancer is ureter and renal pelvis transitional cell cancer.
  • the cancer is urethral cancer.
  • the cancer is uterine cancer (endometrial).
  • the cancer is uterine sarcoma.
  • the cancer is vaginal cancer.
  • the cancer is a visual pathway and hypothalamic glioma (childhood).
  • the cancer is vulvar cancer. In some embodiments, the cancer is Waldenström macroglobulinemia. In some embodiments, the cancer is Wilms tumor (childhood).
  • the pharmaceutical composition is administered intravenously. In certain embodiments, the pharmaceutical composition is administered topically, for example, to the subject’s colon. In certain embodiments, the pharmaceutical composition is administered orally. In certain embodiments, the pharmaceutical composition is administered anally.
  • the present disclosure provides a method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with a pharmaceutical composition disclosed herein.
  • the binding is selective over annexin A1 (ANXA1), non-activated ANXA2, and annexin A3 (ANXA3).
  • tumor margins of cancer are defined.
  • the accuracy of cancer resection is improved during surgery.
  • cancer is treated simultaneously from the periphery and interior core of a tumor.
  • the present disclosure also provides doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp- Ser-Pro-Cys)-Lys-OH) and salts thereof.
  • the cypate is 14 KC0004-401-PC 019279/WO [0043] In is D-Cys.
  • t e compound as t e structura ormu a effective amount of doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition further comprises a divalent metal ion, such as one chosen from Ca 2+ , Mg 2+ , and Mn 2+ .
  • the divalent metal ion is Ca 2+ .
  • the divalent metal ion is present at a concentration between 1 mM and 10 mM, such as 5 mM.
  • the pharmaceutical composition further comprises albumin, such as albumin chosen from human serum albumin, mouse serum albumin, and bovine serum albumin.
  • the pharmaceutically acceptable carrier comprises 15 KC0004-401-PC 019279/WO phosphate-buffered saline.
  • the pharmaceutical composition is administered intravenously. In certain embodiments, the pharmaceutical composition is administered topically, for example, to the subject’s colon. In certain embodiments, the pharmaceutical composition is administered orally.
  • the effective amount up to about 0.6 ⁇ mol/kg including 0.1 ⁇ mol/kg, 0.2 ⁇ mol/kg, 0.3 ⁇ mol/kg, 0.4 ⁇ mol/kg, 0.5 ⁇ mol/kg or 0.6 ⁇ mol/kg.
  • the present disclosure provides a method for treating cancer in a subject in need thereof, comprising administering an effective amount of doxorubicin-cypate-cyclic(Cys- Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and salts thereof, or a pharmaceutical composition of doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and/or salts thereof to the subject.
  • the conjugate with LS301 does not accumulate in the heart, thus avoiding dose-limiting toxicity.
  • the present disclosure provides a lyophilized product comprising a dye-conjugate chosen from cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate- Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838), or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration, and albumin.
  • the dye-conjugate and the albumin are in a 1:50 (w/w) ratio.
  • the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin.
  • the lyophilized product may be formed by a method comprising suspending the dye-conjugate in 1% albumin solution at a ratio of 1 gram of dye- conjugate per 10 liters of the albumin solution, mixing the suspension, filtering the mixed suspension, and lyophilizing the filtered suspension to form the lyophilized product.
  • the suspension is mixed on a shaker of at least about 30 minutes.
  • the concentration of the dye-conjugate and albumin in the suspension is measured via absorbance.
  • the present disclosure further provides a vial comprising about 102 mg of the lyophilized product described herein.
  • an injectable solution comprises about 102 mg of the lyophilized product of any one of claims 20 to 24 and about 10 mL of phosphate-buffered saline.
  • the injectable solution comprises about 0.2 mg/mL dye-conjugate. 16 KC0004-401-PC 019279/WO [0053]
  • the present disclosure also provides a method of preparing an injectable solution.
  • the method comprises mixing about 10 mL of phosphate-buffered saline into a vial comprising about 102 mg of the lyophilized product described herein to form an injectable solution comprising about 0.2 mg/mL dye-conjugate.
  • EMBODIMENTS [0054] Provided herein are the following specific embodiments: [0055] Embodiment 1.
  • a pharmaceutical composition comprising an effective amount of cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate-Cyclo(Cys-Gly-Arg- Asp-Ser-Pro-Cys)-Tyr-OH (LS838) or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration; a divalent metal ion; and a pharmaceutically acceptable carrier.
  • Embodiment 2 The pharmaceutical composition of Embodiment 1, wherein the divalent metal ion is chosen from Ca 2+ , Mg 2+ , and Mn 2+ .
  • Embodiment 2 wherein the divalent metal ion is Ca 2+ .
  • Embodiment 4 The pharmaceutical composition of any preceding Embodiment, wherein the divalent metal ion is present at a concentration between 1 mM and 10 mM.
  • Embodiment 5. The pharmaceutical composition of Embodiment 4, wherein the divalent metal ion is present at a concentration of 5 mM.
  • Embodiment 6. The pharmaceutical composition of any preceding Embodiment, wherein the cypate is .
  • Embodiment ding Embodiment, wherein at least one of the Cys amino acid residues is D-Cys.
  • Embodiment 13 The pharmaceutical composition of Embodiment 12, wherein the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin.
  • Embodiment 14 The pharmaceutical composition of any preceding Embodiment, wherein the pharmaceutically acceptable carrier comprises phosphate-buffered saline.
  • Embodiment 15 The pharmaceutical composition of any preceding Embodiment formulated for intravenous administration.
  • Embodiment 16 The pharmaceutical composition of any one of Embodiments 1–14 formulated for topical administration.
  • Embodiment 17. The pharmaceutical composition of any preceding Embodiment, wherein the effective amount is 0.2 ⁇ mol/kg.
  • Embodiment 18. The pharmaceutical composition of any one of Embodiments 1–16, wherein the effective amount is 0.4 ⁇ mol/kg.
  • Embodiment 19 The pharmaceutical composition of any one of Embodiments 1–16, wherein the effective amount is 0.6 ⁇ mol/kg.
  • Embodiment 20 The pharmaceutical composition of any preceding Embodiment formulated for intravenous administration.
  • Embodiment 16 The pharmaceutical composition of any one of Embodiments 1–14 formulated for topical administration.
  • Embodiment 17. The pharmaceutical composition of any preceding Embodiment, wherein the effective amount is 0.2 ⁇ mol/kg.
  • Embodiment 18 The pharmaceutical composition
  • a method for identifying compromised fibroblasts comprising administering an effective amount of the pharmaceutical composition of any preceding Embodiment to a subject in need thereof.
  • Embodiment 21 The method of Embodiment 20, wherein the compromised fibroblasts are proximal to dormant cancer cells.
  • Embodiment 22 The method of Embodiment 22, wherein the compromised fibroblasts are proximal to dormant cancer cells.
  • the dormant cancer cells are chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (primary), cerebellar astrocytoma, cerebral
  • Embodiment 23 The method of Embodiment 22, wherein the dormant cancer cells are pancreatic cancer.
  • Embodiment 24 The method of Embodiment 21, wherein the pharmaceutical composition is administered intravenously.
  • Embodiment 25 The method of Embodiment 21, wherein the pharmaceutical composition is administered topically.
  • Embodiment 26 The method of Embodiment 21, wherein the topical administration is to the subject’s colon.
  • Embodiment 27 A method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with a pharmaceutical composition of any one of Embodiments 1–19.
  • Embodiment 28 A method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with a pharmaceutical composition of any one of Embodiments 1–19.
  • Embodiment 27 wherein the binding is selective over annexin A1 (ANXA1), non-activated ANXA2, and annexin A3 (ANXA3).
  • Embodiment 29 The method of Embodiment 27, wherein tumor margins of cancer are defined.
  • Embodiment 30 The method of Embodiment 27, wherein the accuracy of cancer resection is improved during surgery.
  • Embodiment 31 The method of Embodiment 27, wherein cancer is treated simultaneously from the periphery and interior core of a tumor.
  • Embodiment 32 Embodiment 32.
  • the cancer is chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial 22 KC0004-401-PC 019279/WO adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (
  • annexin A2 Recombinant annexin A2, annexin A2 with glutathione-S-transferase (GST) tag, annexin A1, and annexin A3 proteins were purchased from MyBioSource, Inc. (San Diego, CA, USA). Purified ANXA2 protein and purified pANXA2 protein were gifts from Dr. Gabriel Birrane (Beth Israel Deaconess Medical Center, 24 KC0004-401-PC 019279/WO Boston, MA).
  • Rabbit monoclonal anti-annexin A2 antibody (D11G2, #8235), Rabbit monoclonal anti-Rab5 (C8B1, #3547), and rabbit monoclonal anti-Rab7 (D95F2, #9367) primary antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA).
  • Mouse monoclonal anti- ⁇ -actin 2A3 (sc-517582), mouse monoclonal anti-pANXA2 antibody (11.Tyr 24) (sc-135752), rabbit polyclonal anti-NOS2 (C-19, sc-649), rabbit polyclonal anti-ARG1 (H-52, sc-20150) antibodies, and protein A/G agarose were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA).
  • Rabbit polyclonal anti-pANXA2 (phosphor-Tyr24) antibody was purchased from Signalway Antibody (College Park, MD, USA).
  • Rabbit polyclonal anti-LS301 antibody was generated by Antibody Research Corporation (St.
  • Human Anxa2 cDNA subcloned into the pCMV6-AC-GFP vector was purchased from OriGene Technologies (Rockville, MD, USA).
  • cDNA for the mutant pEZ- M98-GFP- Anxa2-Y23A (tyrosine replaced by alanine) was obtained from GeneCopoeia (Rockville, MD, USA).
  • the recombinant plasmids were characterized by restriction digest, and the quality of the expressed recombinant protein was assessed by confocal microscopy for fluorescence integrity and by western blot. [0090] Cell lines and culture.
  • the cancer cell lines 4T1, Lewis Lung Carcinoma (LLC), A431, A549, MDA-MB- 231, BxPC-3, and HL-60 were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA).
  • the 4T1-luciferase (4T1-luc) cell line was a gift from Dr. Katherine Weilbaecher (Washington University School of Medicine, St Louis, MO, USA).
  • Human dermal fibroblasts were obtained from Coriell Cell Technologies (Camden, NJ, USA). Unless otherwise indicated, all cell lines were cultured at 37 °C in a 5% CO2 incubator in appropriate media supplemented with 10% FBS.
  • mice Five to seven-week-old nude Balb/c or C57BL/6 mice were purchased from Charles River Laboratories (Wilmington, MA) and housed in designated animal facilities. Mice were fed ad libitum and inspected regularly. All animal studies were approved by the Washington University School of Medicine Animal Studies Committee (protocol numbers 25 KC0004-401-PC 019279/WO 20130207 and 20160207) and performed per humane care and use of research animals.
  • LS301 (cypate-cyclic (DCys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) was synthesized from the linear GRD peptide, H- D Cys(Acm)-Gly-Arg(Pbf)- Asp(tBu)-Ser(tBu)-Pro- Cys(Acm)-Lys(Boc)-OH, was prepared via a CEM Liberty Blue microwave peptide synthesizer (Matthews, NC, USA) on the Fmoc-Lys(Boc)-Wang resin.
  • the resin (0.1 mmol) was swelled in dichloromethane for 1 hour before use. Fmoc-amino acids (0.5 mmol, 5 eq), coupling reagent (HBTU, 0.5 mmol, 5 eq), and diisopropylethylamine (DIEA, 1 mmol, 10 eq) were added to the resin, and the mixture was reacted for 15 min under microwave irradiation (100W, 90 °C). The resin was washed three times with DMF. The Fmoc group’s deprotection was carried out by treatment of 20% piperidine/DMF for 5 min under microwave irradiation (100 W, 90°C).
  • peptidyl resin was washed, and the peptide cyclized through the disulfide bridge with iodine (1.2 eq) in DMF for 90 min.
  • cypate (3 eq) was conjugated to the cyclic peptide on a solid support in the presence of N,N’-diisopropylcarbodiimide (DIC, 5 eq) in DMF to afford the LS301 peptidyl resin.
  • DIC, 5 eq N,N’-diisopropylcarbodiimide
  • the resin was then treated with a trifluoroacetic acid cleavage cocktail: thioanisol: phenol: water (85:5:5:5, v/v/v/v) for 90 min at room temperature.
  • the cleaved peptide product was concentrated in vacuo before purifying via reverse-phase HPLC (Gilson, Middleton, WI, USA).
  • the molecular weight of the final product (1469 Da) was confirmed by electrospray ionization mass spectrometry with peaks observed at 1470 (M+1) and 735 (M+2/2).
  • LS637 (Ac-Cyclo( D Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH), a non-fluorescent analog of LS301, was synthesized by a similar method as LS301, except acetic anhydride was used instead of cypate.
  • the molecular weight of the final product (904 Da) was confirmed by electrospray ionization mass spectrometry with peaks observed at 905 (M+1) and 453 (M+2/2).
  • the resulting product was purified by preparative HPLC using a Grace VydacTM C-18 column (250 ⁇ 21.2 mm) with a UV detector at 254 nm.
  • the desired compound was obtained by linear gradient elution using solvents A (0.1% TFA in water) and B (0.1% TFA in acetonitrile) from 90% to 10% over 30 minutes at 10 mL/min.
  • Analytical HPLC characterized the purity of the peptide. The identity was confirmed by electrospray mass spectrometry (ES+MS): LCKLSL, calculated MW 676 g/mol, observed m/z 677 (M+1); LGKLSL calculated 629.80 g/mol, observed m/z 575 (M+1).
  • Example 5 Synthesis of LS301-doxorubicin conjugate (LS766) 27 KC0004-401-PC 019279/WO [0098]
  • Doxorubicin (3 eq, LC Laboratories, Woburn, MA) was added to a mixture of the LS301 peptidyl resin, (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (HATU, 6 eq), and DIEA (6 eq) in DMF.
  • the resin was treated with a cleavage cocktail consisting of TFA: thioanisol: phenol: water (85:5:5:5, v/v/v/v) for 90 minutes at room temperature.
  • the cleaved peptide product was concentrated in vacuo, then purified on reverse-phase HPLC (Gilson, Middleton, WI, USA) to obtain LS766 (doxorubicin-cypate-cyclic(DCys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys- OH), M/W 1995 g/mol, ESI-MS observed 998 (M+2/2) and 666 (M+3/3).
  • the LS838 peptidyl resin can be reacted with doxorubicin to yield doxorubicin-cypate-cyclic(DCys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH, comprising the structural formula 28 KC0004-401-PC 019279/WO .
  • immunoprecipitation experiments were performed by incubation of LS301 with either 4T1 cell lysates or live 4T1 cells, followed by pulldown of LS301-associated proteins with anti-LS301 antibody and protein A/G agarose beads.
  • the protein concentration was determined by BAC protein assay (Bio-Rad) with bovine serum albumin (BSA) as a standard. The supernatants were stored at -80 °C. [0101]
  • BSA bovine serum albumin
  • 400 ⁇ g of cellular protein extract was incubated with LS301 (40 ⁇ M) in 0.5 mL 1X phosphate-buffered saline (PBS) for 2 hours at room temperature.
  • Annexin A2-GST, annexin A1, or annexin A3 (1.0 ⁇ g protein per sample) was pre-incubated with or without 2.5 mM of LS637 (non-fluorescent LS301 analog; blocking peptide) in binding buffer (25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM MgCl2, 1 mM CaCl2) for 1 hour at 37 °C.
  • binding buffer 25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM MgCl2, 1 mM CaCl2
  • Ten ⁇ M LS301 was then added to each sample. After incubation for 2 hours at 37 °C, proteins were subjected to SDS-PAGE and imaged fluorescently.
  • LS301 was incubated with 4T1 tumor cells before SDS-PAGE.
  • MST Microscale thermophoresis studies biomolecular interactions in an aqueous environment without immobilization. MST exploits the directed movement of molecules along a microscopic temperature gradient. The movement rate and direction of movement depend on the species’ size, charge, and solvation shell under study. Changes in this environment can change the thermophoretic movement of the species. MST can distinguish very small changes that occur, for example, when a small fluorescent molecule binds to the surface of a protein. [0104] MST was performed using the Monolith NT.115 (Nanotemper Technologies, Kunststoff, Germany).
  • MST-1 buffer 50 mM Tris-HCl, pH 7.4; 150 mM NaCl; 10 mM MgCl2; 2% DMSO; 0.1% Tween- 20; 4% SDS, 4 mM dithiothreitol (DTT)
  • MST-2 buffer 50 mM Tris-HCl, pH 7.4; 150 mM NaCl; 10 mM MgCl2.
  • the protein was prepared in MST-2 buffer, and 20 ⁇ L serial dilutions were made with protein concentration ranging from 3.05 pM to 50 nM (pANXA2) or 3.81 pM to 62.5 nM (ANXA2). All samples were loaded into NT.115 standard capillaries. The analysis was performed at 37 °C, 30% LED power, 60% MST power, and a constant LS301 concentration of 20 nM. The apparent 30 KC0004-401-PC 019279/WO K d values were calculated from fragment concentration-dependent changes in normalized fluorescence (Fraction Bound) of LS301 after 5 seconds of thermophoresis based on mass action law using the MO Affinity Analysis v2.3 software.
  • Total cellular protein or pure proteins were denatured in SDS gel-loading buffer (100 mM Tris-HCl, 200 mM DTT, 4% SDS, 0.2% bromophenol blue, and 20% glycerol) for 7 minutes at 100 °C and then separated on 12% TGX gel (Bio-Rad, Hercules, CA, USA). After electrophoresis, proteins were transferred to PVDF-FL membrane using an EC140 Mini Blot Module (Thermo EC, Holbrook, NY, USA) apparatus.
  • SDS gel-loading buffer 100 mM Tris-HCl, 200 mM DTT, 4% SDS, 0.2% bromophenol blue, and 20% glycerol
  • the membrane was treated with Odyssey blocking buffer for 1 hour at room temperature, followed by incubation with rabbit monoclonal anti-annexin A2 (D11G2, Cell Signaling Technology, Inc.) and mouse monoclonal pannexin II (11.Tyr24; Santa Cruz Biotechnology) or rabbit ANXA2 (Phospho- Tyr24; MyBioSource, Inc.) primary antibodies in Odyssey blocking buffer at 4 °C overnight. After washing three times for 10 minutes each in PBS with 0.1% Tween-20 (PBS-T), the membrane was incubated for 1 hour with diluted IRDye 680RD goat anti-rabbit IgG or IRDye 800CW goat anti-mouse IgG in Odyssey blocking buffer.
  • rabbit monoclonal anti-annexin A2 D11G2, Cell Signaling Technology, Inc.
  • mouse monoclonal pannexin II 11.Tyr24; Santa Cruz Biotechnology
  • rabbit ANXA2 Phospho- Tyr24; MyBioSource, Inc.
  • Example 7 Proteomics [0106] For mass spectrometric protein identification, the appropriate bands were excised from protein gels and submitted for LC-MS/MS protein identification at the Donald Danforth Plant Science Center, Proteomics & Mass Spectrometry Facility (St. Louis, MO, USA).
  • Proteins that contained similar peptides and could not be differentiated based on MS/MS analysis alone were grouped to satisfy parsimony. Proteins sharing significant peptide evidence were grouped into clusters. [0110] In general, cultured cancer cells were grown and incubated with the indicated amount of probe in Lab-Tek 8-chambered slides for the indicated time durations. Cells were washed with PBS at selected time points and underwent conventional slide preparation, ICC if indicated, and viewing under epifluorescence or confocal.
  • LS301 was added to 4T1 cells at 5 ⁇ M for 1 hour, followed by staining with AlexaFluor 488-conjugated cholera toxin B for 10 minutes at 4 °C, washing with PBS, and followed by adding anti-cholera toxin B antibody for 15 min at 4 °C to crosslink labeled lipid 32
  • LS301 and AlexaFluor 488 fluorescence were imaged using the cypate filter set (Ex/Em 750–800 nm/818–873 nm) and fluorescein isothiocyanate (FITC) filter set (Ex/Em 460–500/510–560nm), respectively.
  • FITC fluorescein isothiocyanate
  • HEK 293T cells (or in some cases 4T1 cells) were maintained in DMEM medium supplemented with 10% fetal bovine serum and 100 units/mL penicillin.
  • DMEM Dulbecco’s Modified Eagle’s medium
  • FBS fetal bovine serum
  • 4T1 murine breast cancer cells were seeded in an 8-well plate at a density of 5000 cells/well with DMEM media supplemented with 10% FBS and penicillin/streptomycin and 33 KC0004-401-PC 019279/WO incubated overnight at 37 °C. The next day, culture media was removed, followed by pre- treatment of cells with chlorpromazine hydrochloride at 13 ⁇ g/mL for 30 min, filipin complex at 3.2 ⁇ g/mL for 1 hour, or amiloride hydrochloride at 300 ⁇ M for 1 hour in a 300- ⁇ L volume of cell culture media.
  • the drug-containing media was removed after incubation and replaced with fresh culture media containing 6 ⁇ M or 0.9 ⁇ g/mL of LS301 in complete DMEM for 1 hour at 37°C.
  • 4T1 cells were incubated with Pitstop 2 reagent at 25 ⁇ M for 20 minutes, followed by incubation with LS301 at 2 ⁇ M for 1 or 3 hours.
  • the culture media was removed, followed by adding culture media containing SYTO 17 nuclear dye for 40 minutes at 37 °C. Finally, nuclear stain media was removed. Cells were washed 2 times with PBS.
  • LS301-containing media was removed, followed by three PBS washes. Fluorescence organelle staining and imaging were performed per manufacturers’ protocols using VectaCell Rhodamine 123 for mitochondria, Lysotracker for lysosomes, BODIPY TR Ceramide for Golgi apparatus and CellLight Peroxisome-GFP, BacMam 2.0 for peroxisomes.
  • Immunocytochemistry was performed with 4% paraformaldehyde fixation, permeabilization with 0.1% Triton X- 100 solution, and blocking with 5% normal goat serum and 1% BSA in 1X PBS blocking solution.
  • LS301 (1 ⁇ M) was incubated with co-cultures of FB and ANXA2-YFP transfected HL60 cells, co-cultures of FB and wild-type HL60 cells, or co- cultures of FB and 4T1-GFP cells at 37 °C for 3 hours and then washed twice with 0.01 M PBS (pH 7.4). Then, the cells were fixed with 4% paraformaldehyde for 10 minutes. [0119] For immunocytochemistry, slides were blocked with appropriate serum for 35 minutes or 5% non-fat milk PBS (pH 7.4) overnight at 4 °C.
  • the slides were incubated with 1:500 monoclonal rabbit anti-ANXA2 antibody (Cell signaling Technology. Inc. Danvers, MA, USA), 1:500 monoclonal mouse anti-pANXA2 antibody (Santa Cruz Biotechnology, Inc. Santa Cruz, CA, USA), and/or 1:250 monoclonal mouse anti-vimentin antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA) for 1 hour at 37 °C.
  • 1:500 monoclonal rabbit anti-ANXA2 antibody Cell signaling Technology. Inc. Danvers, MA, USA
  • 1:500 monoclonal mouse anti-pANXA2 antibody Santa Cruz Biotechnology, Inc. Santa Cruz, CA, USA
  • 1:250 monoclonal mouse anti-vimentin antibody Santa Cruz Biotechnology, Santa Cruz, CA, USA
  • tissue sections were incubated with 1:1000 Alexa 488 anti-rabbit antibody (Thermo Fisher Scientific, Waltham, MA) and/or 1: 1000 TRITC labeled donkey anti-mouse IgG in MPBS (Jackson ImmunoResearch Lab, West Grove, PA, USA), for 1 hour at 25oC. Finally, slides were imaged under a confocal microscope as described in the Fluorescence Microscopy methods section.
  • ANXA2-YFP expressing HL60 cells were generated by lentiviral transduction as follows. Human full-length ANXA2 cDNA in the pCMV6-AC-GFP vector was purchased from OriGene Technologies (Rockville, MD, USA). BamHI, XhoI, and AgeI restriction enzymes and Quick Ligation kit were purchased from New England Biolabs (Ipswich, MA, USA).
  • FCIV lentiviral vector containing IRES-Venus YFP under the ubiquitin promoter was obtained as a generous gift from Dr. Mingjie Lee at the Hope Center, Washington University School of Medicine (St Louis, MO, USA).
  • the ANXA2 cDNA was subcloned into the FCIV lentiviral vector by restriction digest of the ANXA2 cDNA vector with BamHI and XhoI enzymes, restriction digest of the FCIV vector with BamHI and AgeI enzymes, and ligation using Quick Ligation per manufacturer’s instructions. Klenow fragment and dNTP nucleotide mix (New England Biolabs) was used to fill sticky ends.
  • HEK 293T cells were cultured overnight in DMEM with 10% fetal bovine serum, 100 units/mL penicillin 100 ⁇ g/mL streptomycin before transfection. DNA constructs were transfected into HEK 293T cells using Genejuice (EMD Millipore, Burlington, MA) per manufacturer’s instructions. Culture supernatants were collected 40 hours after transfection, mixed 1:1 with Iscove’s Modified Dulbecco’s medium (IMDM) (Thermo Fisher Scientific) supplemented with 10% fetal bovine serum (FBS) and 1% Streptomycin, and added to HL60 cells in 6-well plates.
  • IMDM Modified Dulbecco’s medium
  • the HL60 cells were checked under a confocal microscope for fluorescence. Populations of HL60 cells strongly expressing ANXA2-YFP were analyzed and collected by flow cytometry using a BD FACS AriaII Cell Sorter at a core laboratory at Washington University in St. Louis School of Medicine (St. Louis, MO, USA). Cell viability was checked by luciferase MTS assay as described in the MTS Assay section of methods.
  • transwell culture and extracellular vesicle transfer studies For transwell studies, human dermal fibroblasts (300,000 cells/well) were cultured alone in 6- well plates, or transwell with 4T1 cells (100,000 cells/insert) suspended within transwell inserts (3 ⁇ m pore polycarbonate membrane, MilliporeSigma, St. Louis, MO, USA). Cells were plated and grown in extracellular vesicle-depleted media for 72 hours and subsequently stained for pANXA2 using rabbit anti-pANXA2 primary antibody (15 min. at 4 °C) and 36 KC0004-401-PC 019279/WO donkey anti-rabbit AlexaFluor 594 conjugated secondary antibody (15 min.
  • Extracellular vesicle-depleted media were generated by collecting flow-through after centrifugal filtration of complete DMEM media using Amicon Ultra-100K (100 kDa molecular weight cutoff membrane) devices (MilliporeSigma, St. Louis, MO). For extracellular vesicle transfer studies, supernatants from 4T1 cells cultured in extracellular vesicle-depleted media for 16 hours were harvested.
  • the 4T1-derived extracellular vesicle fraction was concentrated by serial centrifugal filtration on Amicon Ultra-100K devices.
  • the 4T1-derived extracellular vesicle fraction was then added to fibroblasts (60 ⁇ g/well) for 24 h, with untreated fibroblasts as control.
  • LS301 was then added at 5 ⁇ M for 1 hour, and LS301 uptake was visualized by fluorescent microscopy using the cypate filter set (Ex/Em 750–800 nm/818– 873 nm).
  • fibroblasts with or without added 4T1-derived extracellular vesicles was stained for pANXA2 using rabbit anti-pANXA2 primary antibody and donkey anti-rabbit AlexaFluor 594 conjugated secondary antibody at 1:500 dilutions and visualized by fluorescence microscopy as above.
  • Cell viability was evaluated using CellTiterGlo Luminescent cell viability assay (Promega, USA) per manufacturer’s instructions by standard plate reader for luminescence. The luminescence intensity from cells was normalized to reagent alone. [0124] Overnight cultured 4T1 tumor cells in 96-well plates were treated with 0.5 ⁇ g/ml of doxorubicin (DOX) for 3 hours as a positive control. After treatment with different concentrations of LS301 for 48 hours or 72 hours, the cells were trypsinized, centrifuged, and washed twice with PBS (pH 7.4) before cell cytometry.
  • DOX doxorubicin
  • LS301 fluorescence signal was then assessed by fluorescence microscopy using Cy7 filters or confocal microscope. Sections were subsequently fixed with 4% paraformaldehyde for 10 minutes. [0126] For immunohistochemistry, slides were blocked with appropriate serum for 35 minutes or with 5% non-fat milk PBS (pH 7.4) overnight at 4 °C and incubated with primary antibody overnight at 4 °C or 1 hour at 37 °C. For ANXA2/pANXA2 studies, tissue sections were incubated with 1:250 monoclonal rabbit anti-ANXA2 antibody (Cell signaling Technology. Inc. Danvers, MA) or 1:250 monoclonal mouse anti-pANXA2 antibody (Santa Cruz Biotechnology, Inc. Santa Cruz, CA).
  • tissue sections were incubated with 1:1000 AlexaFluor 488 anti-rabbit antibody (Thermo Fisher Scientific, Waltham, MA) and 1:800 TRITC labeled donkey anti-mouse IgG in MPBS (Jackson ImmunoResearch Lab, West Grove, PA, USA) for 1 hour at 25 oC respectively.
  • Slides were washed again and stained with Hoechst or DAPI nuclear stains for 5 min or stained with 1:4000 nucleus dye ToPro3 (Thermo Fisher Scientific, Waltham, MA) for 45 minutes at 37 °C. After final washes, a coverslip with aqueous fluorescence-saving mounting media was applied before imaging.
  • spontaneous tumor models were used. Animals injected subcutaneously with saline served as sham tumor controls. For near-infrared imaging, 100 ⁇ L volumes of 60 ⁇ M LS301 or cypate in PBS were injected intravenously by lateral tail vein, and animals were imaged using a Pearl Small Animal Imager (LI-COR Biotechnology, Lincoln, NE, USA) at the indicated time points. In some experiments, probes were formulated in 1% albumin in PBS. Animals were shaved using commercially available hair removal equipment and products before imaging. For experiments involving further tissue analysis, animals were anesthetized with isoflurane and euthanized by cervical dislocation under anesthesia. Organs were harvested by surgical dissection.
  • the tissue was dissociated and filtered with a 40- ⁇ m cell strainer tube (Falcon 352235) to collect the sample and washed with PBS (pH 7.4) three times and further centrifuged for collection.
  • the cells were sorted using a BD FACS AriaII Cell Sorter at a core laboratory at Washington 39 KC0004-401-PC 019279/WO University in St. Louis School of Medicine (St. Louis, MO, USA). Untreated cells, cells treated with LS301 (4 ⁇ M for 24h), wild-type 4T1 cells (without GFP), and 4T1 luc/GFP cells were used for compensation and gating.
  • the sorted cells were classified and collected in fresh IMDM supplemented with 10% FBS and 1% streptomycin.
  • Example 9 Fluorescence imaging-guided surgical margin assessment
  • Three 6-week old Balb/c mice were implanted with 100,0004T1-Luc/GFP murine breast cancer cells orthotopically in the mammary fat pad below the right front leg. Seven to ten days post-implantation, all three mice were injected with 100 ⁇ L of calcium-formulated LS301 (5 mM Ca/60 ⁇ M LS301) via tail vein injection. Twenty-four hours post-injection, these mice were subjected to image-guided surgery using the CancerVisionTM Goggle (CVG) system. [0134] Mice were maintained under anesthesia using a continuous 5% isoflurane gas inhalation.
  • TBR Tumor-to-background ratio
  • Example 10 In vivo LS301-doxorubicin drug conjugate therapy
  • LS301-doxorubicin treatment Example 11 – DMSO-Based Formulation
  • 200 ⁇ L of DMSO was added to 0.5 mg LS301 and vortexed for about 1 minute to homogenize. The solution was centrifuged to remove any non- dissolved components. The concentration was measured via absorbance.
  • solution concentrations ranged between 290 and 330 ⁇ M. This concentrated solution was diluted dropwise into 800 ⁇ L phosphate-buffered saline (PBS) calculated such that the final concentration was 60 ⁇ M LS301 and that the final concentration was 20% DMSO/ 80% PBS by volume (“20% DMSO”).
  • PBS phosphate-buffered saline
  • Example 12 – Ethanol-Based Formulation A 1:1 PEG400/Ethanol solution (v/v) was prepared by mixing 4 mL of PEG400 and 4 mL of ethanol and vortexing at high speed for 1 minute.200 ⁇ L of this 1:1 solution was added to about 0.5 mg of LS301 to produce a transparent solution (>300 ⁇ M). No precipitate was observed after centrifugation. The solution was then added dropwise to a PBS/Ethanol solution, such that the final solution had a concentration of 60 ⁇ M, 10% PEG400, 10% ethanol solution, 80% PBS (“1:1 PEG400/Ethanol”).
  • LS301 exhibited variable contrast when placed in different media, with demonstrable significantly increased contrast in 20% PEG400 (contrast in vivo over time) and 3e (contrast ex vivo). LS301 cleared via renal and hepatic mechanisms. [0142] LS301 placed in 20% PEG400 solutions demonstrated increased tumor uptake compared to renal or hepatic clearance. Compared to DMSO based solvents, all the ethanol- 41 KC0004-401-PC 019279/WO based solvents demonstrated a more rapid increase in bladder fluorescence in vivo, suggesting increased renal clearance by ethanol-based solvents.
  • Example 14 Performance Test of PEG/Ethanol Formulation in Different Tumor Models [0143] LS301’s solubility in ethanol ranged near the cutoff for producing a 20% ethanol/PBS solution. Besides, although 20% PEG400 + 20% ethanol had a higher contrast than the other solvent basis, efforts were made to reduce the non-aqueous component and increase the ease in producing the stock solution. The 1:1 PEG400/ethanol solution could maintain stability up to about 1-to-16 dilution. In vivo, a screening test was done by IV injection of 60 ⁇ M solution into a Balb/c mouse with two 4T1luc tumors dorsally injected. The data demonstrated increased contrast compared to 20% DMSO.
  • LS301 multitumor affinity in the new solvent
  • solutions were tested against HT1080, A431, and DBT tumor-bearing mice.
  • LS301 in 20% 1:1 PEG400 demonstrates a tumor-to-muscle ratio of greater than 10-fold in all three models and visible over time and ex vivo studies.
  • Example 15 - HSA/MSA Formulation [0144] One percent human serum albumin (HSA) or mouse serum albumin (MSA) was prepared by diluting 100 mL 25% HSA to 2.5 L with sterile water or dissolving 1 g MSA in sterile water.
  • One gram LS301 was suspended in 10 L of 1% HSA or 1% MSA solution. The solution vortexed on a shaker for 30 minutes.
  • the resulting LS301-HSA or LS301-MSA formulation was sterile filtered through SteriflipTM filter. Aliquots of the filtered sample were taken. The concentration of LS301 and HSA or MSA was confirmed via absorbance.
  • the sample vials containing filtered LS301-HSA or LS301-MSA formulation were placed on dry ice to quick-freeze the sample. The frozen sample vials were placed onto the lyophilizer’s tray dryer, then loaded into the lyophilizer to dry overnight (at least 12 hours). After lyophilization, the sample vials were removed from the lyophilizer’s tray dryer, then crimp sealed. The sample vials were labeled and stored at -20 °C.
  • LS301-HSA or LS301-MSA lyophilized product was reconstituted by injecting 10 mL of saline into vials to form a 0.2 mg/mL injection solution.
  • LS301-HSA formulation was evaluated in multiple cancer models with an injection dose of 0.35 mg/kg. NIR fluorescent images were monitored over time to confirm tumor accumulation.
  • LS301 improved uptake 42 KC0004-401-PC 019279/WO into cancer cells, which use albumin for energy and express cell surface receptors, such as secreted protein acidic and rich in cysteine (SPARC), to internalize the LS301-albumin complex.
  • Example 16 - HSA/MSA Formulation With Metal Ions Five mM of metal chloride (Na, K, Ca, Mg, Mn, Al) solutions were prepared. To the LS301-HSA or LS301-MSA lyophilized product, 10 mL of 5 mM metal chloride solution were added to reconstitute the solution, which was then incubated at room temperature for at least 10 minutes before administration.
  • metal chloride Na, K, Ca, Mg, Mn, Al
  • a solution of LS301 (0.5 mL; 60 ⁇ M) was dissolved in 10% w/v aqueous Pluronic solution to form the sol-gel system, which was stored on ice before administration to maintain its liquid state.
  • Pluronic gel-LS301 formulation a topical approach of the formulation in-vivo study was conducted using a colon cancer HT29 mice model.10 ⁇ L of the Pluronic gel-LS301 formulation was applied to the skin where the tumor was located. In-vivo NIR fluorescent images were taken at different time points post-application.
  • Example 18 Thermal Sensitive Hydrogel Formulation For Topical Application [0152] PLGA-b-PEG-b-PLGA triblock copolymers (1500:1000:1500, transition temperature ⁇ 30 °C, 250 mg) dissolved in 1 mL of cold water (5 °C) were mixed with 44 ⁇ g of LS301 in 1 mL tert-butanol at 60 °C and lyophilized for 24 hours. The lyophilized cake was then 43 KC0004-401-PC 019279/WO rehydrated with 1 mL of cold DI water at 4 °C and gently stirred for at least 6 hours in the cold room at 300 rpm.
  • Example 19 Liposomal Formulation of LS301 [0153]
  • DPPC 1,2-dipalmitoyl-sn-glycero-3-phosphocholine
  • DSPE-PEG 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-5000 amine)
  • cholesterol phosphocholine 67.5%, PEGylated phosphocholine 2.5%, cholesterol 30%
  • Lipid stock solutions were prepared by dissolving each lipid in chloroform in different glass vials.
  • the phosphocholine class lipids that were used comprised: 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC); 1,2-didodecanoyl-sn-glycero-3-phosphocholine (DLPC); 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC); 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC); 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC); 1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE); 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE); 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE); 1,2-dimyristoyl-sn-glycero-3-phosphate (DMPA); 44 KC0004-401-PC 019279
  • the PEGylated phosphocholine class lipids used comprised: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)- 5000 [DSPE- PEG (5000) Amine]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)- 2000 [DSPE- PEG (2000) Amine]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)- 5000 [DSPE- PEG (5000) carboxylic acid]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)- 2000 [DSPE- PEG (2000) carboxylic acid]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene
  • compositions were described as including components or materials, it is contemplated that the compositions can also consist essentially of, or consist of, any combination of the recited components or materials, unless described otherwise.

Abstract

Provided herein is a pharmaceutical composition comprising an effective amount of cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate-Cyclo(Cys-Gly-Arg-Asp- Ser-Pro-Cys)-Tyr-OH (LS838) or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration; a divalent metal ion; and a pharmaceutically acceptable carrier. Further provided are lyophilized products comprising a dye-conjugate and m methods for identifying compromised and for binding phosphorylated annexin A2 (pANXA2) protein in a biological sample using a composition described herein.

Description

KC0004-401-PC 019279/WO NEAR INFRARED FLUORESCENT DYES, FORMULATIONS AND RELATED METHODS CROSS-REFERENCE [0001] This application claims the benefit of priority of United States Provisional Patent Application Serial No.62/947,974 filed December 13, 2019, and also claims the benefit of priority of United States Provisional Patent Application Serial No.62/976,702 filed February 14, 2020, the disclosures of which are each incorporated by reference in their entireties for all purposes. GOVERNMENT LICENSE RIGHTS [0002] This invention was made with government support under Award No. R01 CA171651 awarded by the National Institutes of Health. The government has certain rights in the invention. TECHNICAL FIELD [0003] The present disclosure provides pharmaceutical formulations comprising LS301 or LS838, fluorescent dye-cyclic polypeptide conjugates (dye-conjugates). The formulations herein are suitable for systemic delivery, such as intravenous injection, topical application, and oral gavage. BACKGROUND [0004] Elevated chronic inflammatory milieu, metabolic aberrations, and genetic mutations drive cancer cells’ dynamic adaptation toward their survival, proliferation, and metastasis. Accompanying alterations in cellular processes produce heterogeneous populations of cancer subtypes and distorted stroma characterized by diverse cancer biomarkers. Progress in cancer targeted therapies and imaging relies on effectively and selectively delivering molecules to overexpressed cell surface proteins. Yet, the evolving landscape of tumor survival mechanisms frustrates developing molecularly targeted drugs and imaging agents for each cancer type. [0005] One such cell surface protein is annexin A2 (ANXA2), a member of the annexin family of calcium-dependent phospholipid-binding proteins. ANXA2 exhibits cancer- associated posttranslational modifications (PTMs). It is upregulated in many cancers, including breast, colon, liver, pancreatic, and brain tumors, suggesting a role in tumor proliferation, angiogenesis, invasion, and metastasis. Phosphorylation of ANXA2 at tyrosine 23 (pANXA2) modulates ANXA2 tetramer formation and is a prerequisite for its 1 KC0004-401-PC 019279/WO translocation to the plasma membrane. This PTM occurs in response to growth factor signaling and promotes cancer cell migration and invasion by activating cytoskeletal rearrangements and epithelial-mesenchymal transition. [0006] pANXA2 is an inducible hallmark of diverse solid tumor microenvironments in small animal models and primary human cancer tissues, with its expression confined to tumor regions having elevated calcium levels. Cell surface-associated pANXA2 binds and stabilizes the plasminogen receptor S100A10/p11, which associates with tissue plasminogen activator (tPA) and plasminogen to generate plasmin. The enhanced matrix invasion of tumor cells and migration of tumor-promoting macrophages into tumors. ANXA2 is highly activated (phosphorylated) irrespective of the solid tumor type. In human cancers, pANXA2 is expressed in subtypes of breast cancers, including Er+ and triple-negative cancers, but not in the healthy breast tissue from the same cancer patients. Most ANXA2-based drug delivery strategies rely on the overexpression of ANXA2 in certain tumors. Still, non-tumor tissues also express sufficiently elevated levels to impair selectivity, leading to pre-imaging and tissue biopsy to determine the drugs’ usefulness for treating specific tumors. [0007] A variety of cancers are imaged by administering targeted near-infrared (NIR) fluorescence imaging probes. LS301 and LS838 are excellent NIR cancer imaging probes, especially, as disclosed herein, for their specificity toward pANXA2. The clinical formulation for a hydrophobic optical probe should meet several conditions, including adequate and accurate dosing, bioavailability, targeting efficacy, and appropriate absorption and fluorescence characteristics. The long circulation time of LS301 and LS838 delays the tumor uptake and clearance from non-tumor tissue. The formulations described herein accelerate tumor uptake, allowing for quick clinical intervention within an hour post-administration. SUMMARY [0008] The present disclosure provides stable, sustainable formulations of LS301 and LS838 NIR imaging probes for various biological applications, including lyophilized product, IV injection, topical application, and oral gavage delivery, and doxorubicin conjugates of LS301 and LS838 for targeted cancer treatment. [0009] The present disclosure provides a pharmaceutical composition comprising an effective amount of cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate- Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838), or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration; a divalent metal ion; and a pharmaceutically acceptable carrier. 2 KC0004-401-PC 019279/WO [0010] The present disclosure provides a lyophilized product comprising a dye-conjugate chosen from cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate- Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838) or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration, and albumin, wherein the dye-conjugate and the albumin are in a 1:50 (w/w) ratio. [0011] The present disclosure also provides aa vial comprising about 102 mg of the lyophilized product disclosed herein. [0012] The present disclosure further provides an injectable solution comprising about 102 mg of the lyophilized product disclosed herein and about 10 mL of phosphate-buffered saline. [0013] The present disclosure further provides a method for identifying compromised fibroblasts, comprising administering an effective amount of any pharmaceutical composition disclosed herein to a subject in need thereof. [0014] The present disclosure also provides a method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with any pharmaceutical composition disclosed herein. [0015] The present disclosure provides doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser- Pro-Cys)-Lys-OH) and salts thereof, and pharmaceutical compositions thereof. In certain embodiments, cancer is treated in a subject in need thereof, comprising administering an effective amount of doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and/or salts thereof or a pharmaceutical composition of any one doxorubicin-cypate- cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and/or salts thereof to the subject. DETAILED DESCRIPTION [0016] In one aspect of this disclosure, the accumulation rate, tissue uptake selectivity, and amount of product retained in target tissue are improved. The disclosure is based on the findings that formulating some drugs and imaging agents in uptake excipients or the presence of ionic solutions, such as an electrolyte, can improve the cellular internalization and biodistribution of these products. A general embodiment of this disclosure relates to the formulation of dyes, dye-like molecules, and cyclic or aromatic drugs with excipients such as albumin, polyethylene glycol (PEG), cyclodextrin, and surfactants. The enabling ionic species include cations and anions. Cations include monovalent and multivalent cations such as Na, K, Ba, Ca, Mg, Al, Cu, Fe, and Zr. Anions can be monovalent or multivalent, including chlorides, carbonates, nitrates, cyanines, and sulfonates. An embodiment includes the formulation of LS301 in calcium or magnesium. Another embodiment is the formulation of 3 KC0004-401-PC 019279/WO LS301 in albumin or PEG or dimethylsulfoxide. [0017] LS301 is a cyclic octapeptide labeled with a near-infrared dye which exhibits selective and high-affinity calcium-dependent binding to phosphorylated annexin A2 (pANXA2) protein in diverse solid tumors. LS301 preferentially binds to the invasive edges of tumors, a process amplified by cancer cell-induced pANXA2 expression in tumor- associated stromal cells. Further, it traffics within macrophages to the necrotic core of tumors. High levels of pANXA2 and calcium are present in most solid cancers’ microenvironment, revealing a pathway to image and selectively deliver drugs to tumors. [0018] In certain embodiments, the divalent metal ion is chosen from Ca2+, Mg2+, and Mn2+, for example, Ca2+. In certain embodiments, the divalent metal ion is present at a concentration between 1 mM and 10 mM, such as 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, or 10 mM. In certain embodiments, the divalent metal ion is present at a concentration of 5 mM. [0019] In certain embodiments, the cypate is . [0020] In certain embod
Figure imgf000005_0001
residues is D-Cys. [0021] In certain embodiments, the compound is LS301 and comprises the structural formula 4 KC0004-401-PC 019279/WO . [0022] In certain embodiments, the compound is LS301 comprising the structural formula . [0023] LS301 selectively binds to pANXA2 over the non-activated ANXA2. Histopathology of tissue samples from mice administered with LS301 in vivo showed that the compound accumulates in pANXA2-positive cancer cells. Cancer cells induce pANXA2 expression in tumor-associated fibroblasts and macrophages to stimulate LS301 accumulation in these cells at the periphery and core. By detecting pANXA2-associated cells in the tumor 5 KC0004-401-PC 019279/WO microenvironment, LS301 targets and delivers drugs to multiple types of solid tumors. The preferential localization of LS301 at the proliferating edge and the inner core of solid tumors defines tumor margins, improves cancer resection accuracy during surgery, and treats cancer simultaneously from the periphery and interior core of the tumor. [0024] Uptake of LS301 into the solid cancer cell lines for lung (A549), breast (MDA-MB- 231 and 4T1), and myeloid leukemia (HL60) reached a maximum concentration at 12 hours, which maintained over 24 hours. Cypate alone exhibited only transient retention in these cell lines. Without wishing to be bound by theory, initially, LS301 binds to lipid rafts in the cellular plasma membrane, which endocytosed via a clathrin-dependent pathway after one hour of incubation. LS301 was then trafficked into endosomes. After 5 hours, LS301 was primarily found in lysosomes, mitochondria, and peroxisomes. [0025] In certain embodiments, the pharmaceutical composition comprises albumin. The cypate dye moiety of LS301 reversibly binds to the hydrophobic pockets of albumin. Without wishing to be bound by theory, this albumin binding is likely not the primary cause of LS301 tumor uptake. Tumors did not retain cypate in vivo with or without formulation with albumin. Rather, the albumin is a nitrogen source for growing tumors. In certain embodiments, the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin. In certain embodiments, the albumin is human serum albumin (HSA). In certain embodiments, the albumin is mouse serum albumin (MSA). In certain embodiments, the albumin is bovine serum albumin (BSA). [0026] LS301 preferentially targets ANXA2 over ANXA1 and ANXA3. ANXA2 calcium- dependently associates with cholesterol-rich lipid rafts at cell surfaces. LS301 also localizes to these lipids rafts and binds calcium with an apparent dissociation constant (Kd) of 5.67 ± 2.40 nM as determined by microscale thermophoresis (MST). The apparent Kd for LS301 binding to pANXA2, ANXA2, and ANXA3 proteins was 0.075 ± 0.002 nM; 0.389 ± 0.015 nM; and 6.128 ± 0.280 nM, respectively via MST. LS301 preferentially bound to lysates of insulin-stimulated, pANXA2-expressing 4T1 cells with an apparent Kd of 0.017 ± 0.004 nM. [0027] NIR fluorescence microscopy showed that LS301 substantially colocalized with pANXA2 in LS301-treated A549 and 4T1 cells after 1-hour incubation. Cells expressing the wild-type ANXA2 had increased pANXA2 expression and cellular uptake of LS301 upon stimulation with insulin, while cells expressing mutant ANXA2 (Y23A) showed minimal LS301 uptake or pANXA2 expression with insulin stimulation. Tissue plasminogen activator (tPA) protein binds to a site in the N-terminal domain of ANXA2 comprising the amino acid 6 KC0004-401-PC 019279/WO sequence LCKLSL. A synthetic LCKLSL peptide inhibited the binding and internalization of LS301 in cells. [0028] Adding LS301 to fibroblast co-cultured with ANXA2-positive GFP-expressing 4T1 cells resulted in LS301 uptake by tumor cells and fibroblasts. By increasing the population of pANXA2-positive cells for tumor tissue, cancer cells amplify LS301 accumulation. Taken together, these results demonstrate that LS301 is selective for pANXA2, regardless of the cell or cancer type. [0029] LS301 selectively accumulated in a tumor from a breast cancer patient-derived xenograft and remained in the lesion for over 96 hours. Control experiments with cypate alone or with a scrambled LS301 peptide analog (Cypate-cyclo(Cys-Arg-Gly-Asp-Ser-Pro- Cys)-Lys-OH) were not retained. In small tumors (<5 mm), LS301 coverage is about 100%. In advanced tumors, LS301 accumulated at the periphery as correlated to pANXA2 and calcium expression. In capsular tumors such as breast cancer, LS301 intensely fluoresced in tumor cells and cancer-associated fibroblasts. Identifying these compromised fibroblasts enables one to detect dormant cancer cells early and to help prevent subsequent relapse. [0030] In certain embodiments, the pharmaceutical composition comprises the compound LS838. In certain embodiments, LS838 comprises the structural formula la
Figure imgf000008_0001
7 KC0004-401-PC 019279/WO . PDAC)
Figure imgf000009_0001
and pancreatic intraepithelial neoplasia (PanlN) 2/3 lesions, with high accuracy. Uptake is mediated by the energy needs of these metabolically active cells. LS838 is then trapped intracellularly under the highly reducing environment of these cells. In certain embodiments, applying LS838 to distinguish PDAC and to transform PanlN from chronic pancreatitis with high accuracy significantly progresses treatment for these cancers. [0033] LS838 enables NIR fluorescence to detect microscopic lesions not visible with prior clinical imaging techniques. LS838 fluoresces more brightly than LS301, allowing smaller amounts of LS838 to achieve the same uptake kinetics as LS301. [0034] Without wishing to be bound by theory, the placement of the tyrosine in the LS838 molecule is important for retaining LS838 in tumors. Unlike LS301, LS838 can be radiolabeled at its tyrosine residue, enabling combined intravital fluorescence microscopy and noninvasive imaging. In certain embodiments, the radionuclide is chosen from, for example, fluorine-18, iodine-123, iodine-124, iodine-125, and iodine-131. This radiolabeling allows the imaging of cancer in the human body noninvasively using nuclear imaging methods. The fluorescence allows optical methods to guide tissue biopsy, surgery, and assessment of surgical margins. [0035] Conjugation of chemotherapeutics, such as doxorubicin, to the free carboxylic acid group of LS838, allows for the highly selective treatment of cancer, including PDAC, with minimal off-target effect on the healthy cells. LS838 selectively remains in diverse tumors 8 KC0004-401-PC 019279/WO without significant loss of fluorescence over time. The high specificity of LS838 for cancer cells in the presence of healthy white blood cells allows the same agent to detect circulating tumor cells (CTCs) without additional tagging or expensive antibodies. [0036] In certain embodiments, the pharmaceutically acceptable carrier comprises phosphate- buffered saline. In certain embodiments, the pharmaceutical composition is administered intravenously. In certain embodiments, the pharmaceutical composition is administered topically, for example, to the subject’s colon. In certain embodiments, the pharmaceutical composition is administered orally. In certain embodiments, the effective amount is up to about 0.6 µmol/kg including 0.1 µmol/kg, 0.2 µmol/kg, 0.3 µmol/kg, 0.4 µmol/kg, 0.5 µmol/kg or 0.6 µmol/kg. [0037] The present disclosure further provides a method for identifying compromised fibroblasts, comprising administering an effective amount of the pharmaceutical composition described herein to a subject in need thereof. In certain embodiments, the compromised fibroblasts are proximal to dormant cancer cells. [0038] In certain embodiments, the cancer is chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (primary), cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, cervical cancer, childhood cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, desmoplastic small round cell tumor, endometrial cancer, ependymoma, esophageal cancer, Ewing’s sarcoma in the Ewing family of tumors, extracranial germ cell tumor (childhood), extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancers (intraocular melanoma, retinoblastoma), gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumors (childhood extracranial, extragonadal, ovarian), gestational trophoblastic tumor, gliomas (adult, childhood brain stem, childhood cerebral astrocytoma, childhood visual pathway and hypothalamic), gastric carcinoid, hairy cell leukemia, head and neck cancer, hepatocellular 9 KC0004-401-PC 019279/WO (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, hypothalamic and visual pathway glioma (childhood), intraocular melanoma, islet cell carcinoma, Kaposi sarcoma, kidney cancer (renal cell cancer), laryngeal cancer, leukemias (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, hairy cell), lip and oral cavity cancer, liver cancer (primary), lung cancers (non-small cell, small cell), lymphomas (AIDS-related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, primary central nervous system), macroglobulinemia (Waldenström), malignant fibrous histiocytoma of bone/osteosarcoma, medulloblastoma (childhood), melanoma, intraocular melanoma, Merkel cell carcinoma, mesotheliomas (adult malignant, childhood), metastatic squamous neck cancer with occult primary, mouth cancer, multiple endocrine neoplasia syndrome (childhood), multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, myelogenous leukemia (chronic), myeloid leukemias (adult acute, childhood acute), multiple myeloma, myeloproliferative disorders (chronic), nasal cavity and paranasal sinus cancer, nasopharyngeal carcinoma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma/malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer (surface epithelial-stromal tumor), ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, pancreatic cancer (islet cell), paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pineal astrocytoma, pineal germinoma, pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood), pituitary adenoma, plasma cell neoplasia, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell carcinoma (kidney cancer), renal pelvis and ureter transitional cell cancer, retinoblastoma, rhabdomyosarcoma (childhood), salivary gland cancer, sarcoma (Ewing family of tumors, Kaposi, soft tissue, uterine), Sézary syndrome, skin cancers (nonmelanoma, melanoma), skin carcinoma (Merkel cell), small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer with occult primary (metastatic), stomach cancer, supratentorial primitive neuroectodermal tumor (childhood), T- Cell lymphoma (cutaneous), testicular cancer, throat cancer, thymoma (childhood), thymoma and thymic carcinoma, thyroid cancer, thyroid cancer (childhood), transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor (gestational), unknown primary site (adult, childhood), ureter and renal pelvis transitional cell cancer, urethral cancer, uterine cancer (endometrial), uterine sarcoma, vaginal cancer, visual pathway and hypothalamic glioma 10 KC0004-401-PC 019279/WO (childhood), vulvar cancer, Waldenström macroglobulinemia, and Wilms tumor (childhood). [0039] In some embodiments, the cancer is acute lymphoblastic leukemia. In some embodiments, the cancer is acute myeloid leukemia. In some embodiments, the cancer is adrenocortical carcinoma. In some embodiments, the cancer is AIDS-related cancer, such as AIDS-related lymphoma. In some embodiments, the cancer is anal cancer. In some embodiments, the cancer is appendix cancer. In some embodiments, the cancer is astrocytomas (childhood cerebellar or cerebral). In some embodiments, the cancer is basal cell carcinoma. In some embodiments, the cancer is bile duct cancer. In some embodiments, the cancer is bladder cancer. In some embodiments, the cancer is bone cancer. In some embodiments, the cancer is brainstem glioma. In some embodiments, the cancer is brain tumors, such as cerebellar astrocytoma. In some embodiments, the cancer is cerebral astrocytoma/malignant glioma. In some embodiments, the cancer is ependymoma. In some embodiments, the cancer is medulloblastoma. In some embodiments, the cancer is supratentorial primitive neuroectodermal tumors. In some embodiments, the cancer is a visual pathway and hypothalamic gliomas. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is bronchial adenomas/carcinoids. In some embodiments, the cancer is Burkitt lymphoma. In some embodiments, the cancer is carcinoid tumors (childhood, gastrointestinal). In some embodiments, the cancer is carcinoma of an unknown primary. In some embodiments, the cancer is central nervous system lymphoma (primary). In some embodiments, the cancer is cerebellar astrocytoma. In some embodiments, the cancer is cerebral astrocytoma/malignant glioma. In some embodiments, the cancer is cervical cancer. In some embodiments, the cancer is childhood cancer. In some embodiments, the cancer is chronic lymphocytic leukemia. In some embodiments, the cancer is chronic myelogenous leukemia. In some embodiments, the cancer is a chronic myeloproliferative disorder. In some embodiments, the cancer is colon cancer. In some embodiments, the cancer is cutaneous T- cell lymphoma. In some embodiments, the cancer is a desmoplastic small round cell tumor. In some embodiments, the cancer is endometrial cancer. In some embodiments, the cancer is ependymoma. In some embodiments, the cancer is esophageal cancer. In some embodiments, the cancer is Ewing’s sarcoma in the Ewing family of tumors. In some embodiments, the cancer is an extracranial germ cell tumor (childhood). In some embodiments, the cancer is an extragonadal germ cell tumor. In some embodiments, the cancer is extrahepatic bile duct cancer. In some embodiments, the cancer is eye cancer, such as intraocular melanoma retinoblastoma. In some embodiments, the cancer is gallbladder cancer. In some 11 KC0004-401-PC 019279/WO embodiments, the cancer is gastric (stomach) cancer. In some embodiments, the cancer is a gastrointestinal carcinoid tumor. In some embodiments, the cancer is a gastrointestinal stromal tumor. In some embodiments, the cancer is germ cell tumors (childhood extracranial, extragonadal, ovarian). In some embodiments, the cancer is a gestational trophoblastic tumor. In some embodiments, the cancer is gliomas (adult, childhood brain stem. childhood cerebral astrocytoma, childhood visual pathway, and hypothalamic). In some embodiments, the cancer is gastric carcinoid. In some embodiments, the cancer is hairy cell leukemia. In some embodiments, the cancer is head and neck cancer. In some embodiments, the cancer is hepatocellular (liver) cancer. In some embodiments, the cancer is Hodgkin lymphoma. In some embodiments, the cancer is hypopharyngeal cancer. In some embodiments, the cancer is hypothalamic and visual pathway glioma (childhood). In some embodiments, the cancer is intraocular melanoma. In some embodiments, the cancer is islet cell carcinoma. In some embodiments, the cancer is Kaposi sarcoma. In some embodiments, the cancer is kidney cancer (renal cell cancer). In some embodiments, the cancer is laryngeal cancer. In some embodiments, the cancer is leukemia, such as acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, or hairy cell. In some embodiments, the cancer is a lip and oral cavity cancer. In some embodiments, the cancer is liver cancer (primary). In some embodiments, the cancer is lung cancers, such as non-small cell lung cancer and small cell lung cancer. In some embodiments, the cancer is lymphoma, such as AIDS-related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, and primary central nervous system. In some embodiments, the cancer is macroglobulinemia (Waldenström). In some embodiments, the cancer is malignant fibrous histiocytoma of bone/osteosarcoma. In some embodiments, the cancer is medulloblastoma (childhood). In some embodiments, the cancer is melanoma. In some embodiments, the cancer is intraocular melanoma. In some embodiments, the cancer is Merkel cell carcinoma. In some embodiments, the cancer is mesotheliomas, such as adult malignant and childhood. In some embodiments, the cancer is metastatic squamous neck cancer with occult primary. In some embodiments, the cancer is mouth cancer. In some embodiments, the cancer is multiple endocrine neoplasia syndrome (childhood). In some embodiments, the cancer is multiple myeloma/plasma cell neoplasm. In some embodiments, the cancer is mycosis fungoides. In some embodiments, the cancer is myelodysplastic syndromes. In some embodiments, the cancer is a myelodysplastic/ myeloproliferative disease. In some embodiments, the cancer is myelogenous leukemia (chronic). In some embodiments, the cancer is myeloid leukemias, such as adult acute and childhood acute. In 12 KC0004-401-PC 019279/WO some embodiments, the cancer is multiple myeloma. In some embodiments, the cancer is myeloproliferative disorders (chronic). In some embodiments, the cancer is a nasal cavity and paranasal sinus cancer. In some embodiments, the cancer is nasopharyngeal carcinoma. In some embodiments, the cancer is neuroblastoma. In some embodiments, the cancer is non- Hodgkin lymphoma. In some embodiments, the cancer is non-small cell lung cancer. In some embodiments, the cancer is oral cancer. In some embodiments, the cancer is oropharyngeal cancer. In some embodiments, the cancer is osteosarcoma/malignant fibrous histiocytoma of bone. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is ovarian epithelial cancer, such as a surface epithelial-stromal tumor. In some embodiments, the cancer is an ovarian germ cell tumor. In some embodiments, the cancer is an ovarian low malignant potential tumor. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is pancreatic cancer (islet cell). In some embodiments, the cancer is paranasal sinus and nasal cavity cancer. In some embodiments, the cancer is parathyroid cancer. In some embodiments, the cancer is penile cancer. In some embodiments, the cancer is pharyngeal cancer. In some embodiments, the cancer is pheochromocytoma. In some embodiments, the cancer is pineal astrocytoma. In some embodiments, the cancer is pineal germinoma. In some embodiments, the cancer is pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood). In some embodiments, the cancer is a pituitary adenoma. In some embodiments, the cancer is plasma cell neoplasia. In some embodiments, the cancer is pleuropulmonary blastoma. In some embodiments, the cancer is a primary central nervous system lymphoma. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is rectal cancer. In some embodiments, the cancer is renal cell carcinoma (kidney cancer). In some embodiments, the cancer is renal pelvis and ureter transitional cell cancer. In some embodiments, the cancer is retinoblastoma. In some embodiments, the cancer is rhabdomyosarcoma (childhood). In some embodiments, the cancer is salivary gland cancer. In some embodiments, the cancer is sarcoma, such as the Ewing family of tumors, Kaposi, soft tissue, and uterine. In some embodiments, the cancer is Sézary syndrome. In some embodiments, the cancer is skin cancers, such as nonmelanoma and melanoma. In some embodiments, the cancer is skin carcinoma (Merkel cell). In some embodiments, the cancer is small cell lung cancer. In some embodiments, the cancer is small intestine cancer. In some embodiments, the cancer is soft tissue sarcoma. In some embodiments, the cancer is squamous cell carcinoma. In some embodiments, the cancer is squamous neck cancer with occult primary (metastatic). In some embodiments, the cancer is 13 KC0004-401-PC 019279/WO stomach cancer. In some embodiments, the cancer is a supratentorial primitive neuroectodermal tumor (childhood). In some embodiments, the cancer is T-Cell lymphoma (cutaneous). In some embodiments, the cancer is testicular cancer. In some embodiments, the cancer is throat cancer. In some embodiments, the cancer is thymoma (childhood). In some embodiments, the cancer is thymoma and thymic carcinoma. In some embodiments, the cancer is thyroid cancer. In some embodiments, the cancer is thyroid cancer (childhood). In some embodiments, the cancer is transitional cell cancer of the renal pelvis and ureter. In some embodiments, the cancer is a trophoblastic tumor (gestational). In some embodiments, the cancer is an unknown primary site, such as adult and childhood. In some embodiments, the cancer is ureter and renal pelvis transitional cell cancer. In some embodiments, the cancer is urethral cancer. In some embodiments, the cancer is uterine cancer (endometrial). In some embodiments, the cancer is uterine sarcoma. In some embodiments, the cancer is vaginal cancer. In some embodiments, the cancer is a visual pathway and hypothalamic glioma (childhood). In some embodiments, the cancer is vulvar cancer. In some embodiments, the cancer is Waldenström macroglobulinemia. In some embodiments, the cancer is Wilms tumor (childhood). [0040] In certain embodiments, the pharmaceutical composition is administered intravenously. In certain embodiments, the pharmaceutical composition is administered topically, for example, to the subject’s colon. In certain embodiments, the pharmaceutical composition is administered orally. In certain embodiments, the pharmaceutical composition is administered anally. [0041] The present disclosure provides a method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with a pharmaceutical composition disclosed herein. In certain embodiments, the binding is selective over annexin A1 (ANXA1), non-activated ANXA2, and annexin A3 (ANXA3). In certain embodiments, tumor margins of cancer are defined. In certain embodiments, the accuracy of cancer resection is improved during surgery. In certain embodiments, cancer is treated simultaneously from the periphery and interior core of a tumor. [0042] The present disclosure also provides doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp- Ser-Pro-Cys)-Lys-OH) and salts thereof. In certain embodiments, the cypate is 14 KC0004-401-PC 019279/WO [0043] In is D-Cys. [0044] In
Figure imgf000016_0001
certan embod ments, t e compound as t e structura ormu a
Figure imgf000016_0002
effective amount of doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. [0046] In certain embodiments, the pharmaceutical composition further comprises a divalent metal ion, such as one chosen from Ca2+, Mg2+, and Mn2+. In certain embodiments, the divalent metal ion is Ca2+. In certain embodiments, the divalent metal ion is present at a concentration between 1 mM and 10 mM, such as 5 mM. [0047] In certain embodiments, the pharmaceutical composition further comprises albumin, such as albumin chosen from human serum albumin, mouse serum albumin, and bovine serum albumin. In certain embodiments, the pharmaceutically acceptable carrier comprises 15 KC0004-401-PC 019279/WO phosphate-buffered saline. [0048] In certain embodiments, the pharmaceutical composition is administered intravenously. In certain embodiments, the pharmaceutical composition is administered topically, for example, to the subject’s colon. In certain embodiments, the pharmaceutical composition is administered orally. In certain embodiments, the effective amount up to about 0.6 µmol/kg including 0.1 µmol/kg, 0.2 µmol/kg, 0.3 µmol/kg, 0.4 µmol/kg, 0.5 µmol/kg or 0.6 µmol/kg. [0049] The present disclosure provides a method for treating cancer in a subject in need thereof, comprising administering an effective amount of doxorubicin-cypate-cyclic(Cys- Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and salts thereof, or a pharmaceutical composition of doxorubicin-cypate-cyclic(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) and/or salts thereof to the subject. Unlike free doxorubicin, the conjugate with LS301 does not accumulate in the heart, thus avoiding dose-limiting toxicity. This same effect is expected for the doxorubicin conjugate with LS838. [0050] The present disclosure provides a lyophilized product comprising a dye-conjugate chosen from cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate- Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838), or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration, and albumin. In certain embodiments, the dye-conjugate and the albumin are in a 1:50 (w/w) ratio. In certain embodiments, the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin. [0051] In certain embodiments, the lyophilized product may be formed by a method comprising suspending the dye-conjugate in 1% albumin solution at a ratio of 1 gram of dye- conjugate per 10 liters of the albumin solution, mixing the suspension, filtering the mixed suspension, and lyophilizing the filtered suspension to form the lyophilized product. In certain embodiments, the suspension is mixed on a shaker of at least about 30 minutes. In certain embodiments, the concentration of the dye-conjugate and albumin in the suspension is measured via absorbance. [0052] The present disclosure further provides a vial comprising about 102 mg of the lyophilized product described herein. In certain embodiments, an injectable solution comprises about 102 mg of the lyophilized product of any one of claims 20 to 24 and about 10 mL of phosphate-buffered saline. In certain embodiments, the injectable solution comprises about 0.2 mg/mL dye-conjugate. 16 KC0004-401-PC 019279/WO [0053] The present disclosure also provides a method of preparing an injectable solution. The method comprises mixing about 10 mL of phosphate-buffered saline into a vial comprising about 102 mg of the lyophilized product described herein to form an injectable solution comprising about 0.2 mg/mL dye-conjugate. EMBODIMENTS [0054] Provided herein are the following specific embodiments: [0055] Embodiment 1. A pharmaceutical composition comprising an effective amount of cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate-Cyclo(Cys-Gly-Arg- Asp-Ser-Pro-Cys)-Tyr-OH (LS838) or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration; a divalent metal ion; and a pharmaceutically acceptable carrier. [0056] Embodiment 2. The pharmaceutical composition of Embodiment 1, wherein the divalent metal ion is chosen from Ca2+, Mg2+, and Mn2+. [0057] Embodiment 3. The pharmaceutical composition of Embodiment 2, wherein the divalent metal ion is Ca2+. [0058] Embodiment 4. The pharmaceutical composition of any preceding Embodiment, wherein the divalent metal ion is present at a concentration between 1 mM and 10 mM. [0059] Embodiment 5. The pharmaceutical composition of Embodiment 4, wherein the divalent metal ion is present at a concentration of 5 mM. [0060] Embodiment 6. The pharmaceutical composition of any preceding Embodiment, wherein the cypate is . [0061] Embodiment ding Embodiment,
Figure imgf000018_0001
wherein at least one of the Cys amino acid residues is D-Cys. [0062] Embodiment 8. The pharmaceutical composition to Embodiment 1, wherein the compound is LS301 comprising the structural formula 17 KC0004-401-PC 019279/WO
Figure imgf000019_0001
compound is LS301 comprising the structural formula
Figure imgf000019_0002
compound is LS838 comprising the structural formula 18 KC0004-401-PC 019279/WO
Figure imgf000020_0001
compound is LS838 comprising the structural formula
Figure imgf000020_0002
further comprising albumin. 19 KC0004-401-PC 019279/WO [0067] Embodiment 13. The pharmaceutical composition of Embodiment 12, wherein the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin. [0068] Embodiment 14. The pharmaceutical composition of any preceding Embodiment, wherein the pharmaceutically acceptable carrier comprises phosphate-buffered saline. [0069] Embodiment 15. The pharmaceutical composition of any preceding Embodiment formulated for intravenous administration. [0070] Embodiment 16. The pharmaceutical composition of any one of Embodiments 1–14 formulated for topical administration. [0071] Embodiment 17. The pharmaceutical composition of any preceding Embodiment, wherein the effective amount is 0.2 µmol/kg. [0072] Embodiment 18. The pharmaceutical composition of any one of Embodiments 1–16, wherein the effective amount is 0.4 µmol/kg. [0073] Embodiment 19. The pharmaceutical composition of any one of Embodiments 1–16, wherein the effective amount is 0.6 µmol/kg. [0074] Embodiment 20. A method for identifying compromised fibroblasts, comprising administering an effective amount of the pharmaceutical composition of any preceding Embodiment to a subject in need thereof. [0075] Embodiment 21. The method of Embodiment 20, wherein the compromised fibroblasts are proximal to dormant cancer cells. [0076] Embodiment 22. The method of Embodiment 21, wherein the dormant cancer cells are chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (primary), cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, cervical cancer, childhood cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, desmoplastic small round cell tumor, endometrial cancer, ependymoma, esophageal cancer, Ewing’s sarcoma in the Ewing family 20 KC0004-401-PC 019279/WO of tumors, extracranial germ cell tumor (childhood), extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancers (intraocular melanoma, retinoblastoma), gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumors (childhood extracranial, extragonadal, ovarian), gestational trophoblastic tumor, gliomas (adult, childhood brain stem, childhood cerebral astrocytoma, childhood visual pathway and hypothalamic), gastric carcinoid, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, hypothalamic and visual pathway glioma (childhood), intraocular melanoma, islet cell carcinoma, Kaposi sarcoma, kidney cancer (renal cell cancer), laryngeal cancer, leukemias (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, hairy cell), lip and oral cavity cancer, liver cancer (primary), lung cancers (non-small cell, small cell), lymphomas (AIDS-related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, primary central nervous system), macroglobulinemia (Waldenström), malignant fibrous histiocytoma of bone/osteosarcoma, medulloblastoma (childhood), melanoma, intraocular melanoma, Merkel cell carcinoma, mesotheliomas (adult malignant, childhood), metastatic squamous neck cancer with occult primary, mouth cancer, multiple endocrine neoplasia syndrome (childhood), multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, myelogenous leukemia (chronic), myeloid leukemias (adult acute, childhood acute), multiple myeloma, myeloproliferative disorders (chronic), nasal cavity and paranasal sinus cancer, nasopharyngeal carcinoma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma/malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer (surface epithelial-stromal tumor), ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, pancreatic cancer (islet cell), paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pineal astrocytoma, pineal germinoma, pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood), pituitary adenoma, plasma cell neoplasia, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell carcinoma (kidney cancer), renal pelvis and ureter transitional cell cancer, retinoblastoma, rhabdomyosarcoma (childhood), salivary gland cancer, sarcoma (Ewing family of tumors, Kaposi, soft tissue, uterine), Sézary syndrome, skin cancers (nonmelanoma, melanoma), skin carcinoma (Merkel cell), small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer with occult primary 21 KC0004-401-PC 019279/WO (metastatic), stomach cancer, supratentorial primitive neuroectodermal tumor (childhood), T- Cell lymphoma (cutaneous), testicular cancer, throat cancer, thymoma (childhood), thymoma and thymic carcinoma, thyroid cancer, thyroid cancer (childhood), transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor (gestational), unknown primary site (adult, childhood), ureter and renal pelvis transitional cell cancer, urethral cancer, uterine cancer (endometrial), uterine sarcoma, vaginal cancer, visual pathway and hypothalamic glioma (childhood), vulvar cancer, Waldenström macroglobulinemia, and Wilms tumor (childhood). [0077] Embodiment 23. The method of Embodiment 22, wherein the dormant cancer cells are pancreatic cancer. [0078] Embodiment 24. The method of Embodiment 21, wherein the pharmaceutical composition is administered intravenously. [0079] Embodiment 25. The method of Embodiment 21, wherein the pharmaceutical composition is administered topically. [0080] Embodiment 26. The method of Embodiment 21, wherein the topical administration is to the subject’s colon. [0081] Embodiment 27. A method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with a pharmaceutical composition of any one of Embodiments 1–19. [0082] Embodiment 28. The method of Embodiment 27, wherein the binding is selective over annexin A1 (ANXA1), non-activated ANXA2, and annexin A3 (ANXA3). [0083] Embodiment 29. The method of Embodiment 27, wherein tumor margins of cancer are defined. [0084] Embodiment 30. The method of Embodiment 27, wherein the accuracy of cancer resection is improved during surgery. [0085] Embodiment 31. The method of Embodiment 27, wherein cancer is treated simultaneously from the periphery and interior core of a tumor. [0086] Embodiment 32. The method of any one of Embodiments 29–31, wherein the cancer is chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial 22 KC0004-401-PC 019279/WO adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (primary), cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, cervical cancer, childhood cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, desmoplastic small round cell tumor, endometrial cancer, ependymoma, esophageal cancer, Ewing’s sarcoma in the Ewing family of tumors, extracranial germ cell tumor (childhood), extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancers (intraocular melanoma, retinoblastoma), gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumors (childhood extracranial, extragonadal, ovarian), gestational trophoblastic tumor, gliomas (adult, childhood brain stem, childhood cerebral astrocytoma, childhood visual pathway and hypothalamic), gastric carcinoid, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, hypothalamic and visual pathway glioma (childhood), intraocular melanoma, islet cell carcinoma, Kaposi sarcoma, kidney cancer (renal cell cancer), laryngeal cancer, leukemias (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, hairy cell), lip and oral cavity cancer, liver cancer (primary), lung cancers (non-small cell, small cell), lymphomas (AIDS-related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, primary central nervous system), macroglobulinemia (Waldenström), malignant fibrous histiocytoma of bone/osteosarcoma, medulloblastoma (childhood), melanoma, intraocular melanoma, Merkel cell carcinoma, mesotheliomas (adult malignant, childhood), metastatic squamous neck cancer with occult primary, mouth cancer, multiple endocrine neoplasia syndrome (childhood), multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, myelogenous leukemia (chronic), myeloid leukemias (adult acute, childhood acute), multiple myeloma, myeloproliferative disorders (chronic), nasal cavity and paranasal sinus cancer, nasopharyngeal carcinoma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma/malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer (surface epithelial-stromal tumor), ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, pancreatic cancer (islet cell), paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pineal astrocytoma, pineal germinoma, pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood), pituitary adenoma, plasma cell neoplasia, 23 KC0004-401-PC 019279/WO pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell carcinoma (kidney cancer), renal pelvis and ureter transitional cell cancer, retinoblastoma, rhabdomyosarcoma (childhood), salivary gland cancer, sarcoma (Ewing family of tumors, Kaposi, soft tissue, uterine), Sézary syndrome, skin cancers (nonmelanoma, melanoma), skin carcinoma (Merkel cell), small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer with occult primary (metastatic), stomach cancer, supratentorial primitive neuroectodermal tumor (childhood), T- Cell lymphoma (cutaneous), testicular cancer, throat cancer, thymoma (childhood), thymoma and thymic carcinoma, thyroid cancer, thyroid cancer (childhood), transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor (gestational), unknown primary site (adult, childhood), ureter and renal pelvis transitional cell cancer, urethral cancer, uterine cancer (endometrial), uterine sarcoma, vaginal cancer, visual pathway and hypothalamic glioma (childhood), vulvar cancer, Waldenström macroglobulinemia, and Wilms tumor (childhood). EXAMPLES General methods [0087] Chemicals. All the fluorenylmethyloxycarbonyl (Fmoc) amino acids, Wang resin Fmoc- Tyr(tBu)-Wang resin, and Fmoc-Lys(Boc)-Wang Resin were purchased from AAPPTec (Louisville, KY, USA). Dichloromethane (DCM), acetic acid, acetic anhydride, thioanisole, phenol, hydroxybenzotriazole (HOBt), N,N-diisopropylethylamine (DIEA), N- trityl-1,2-ethanediamine, phenol, thioanisol, dimethylformamide (DMF), N,N’- diisopropylcarbodiimide (DIC), trifluoroacetic acid (TFA), iodine, methyl tert-butyl ether (MTBE) and O-(7-azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (HATU) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Water was obtained from a Millipore Q3 system. Lysotracker, BODIPY TR Ceramide, CellLight Peroxisome- GFP, BacMam 2.0, and Fluo-4 calcium probe were purchased from Thermo Fisher Scientific (Waltham, MA). Chlorpromazine hydrochloride, filipin complex, and amiloride hydrochloride were purchased from Sigma-Aldrich (St Louis, MO, USA). Reagents for organelle staining included VectaCell Rhodamine 123 (Vector Laboratories, Burlingame, CA, USA). [0088] Recombinant proteins and antibodies. Recombinant annexin A2, annexin A2 with glutathione-S-transferase (GST) tag, annexin A1, and annexin A3 proteins were purchased from MyBioSource, Inc. (San Diego, CA, USA). Purified ANXA2 protein and purified pANXA2 protein were gifts from Dr. Gabriel Birrane (Beth Israel Deaconess Medical Center, 24 KC0004-401-PC 019279/WO Boston, MA). Rabbit monoclonal anti-annexin A2 antibody (D11G2, #8235), Rabbit monoclonal anti-Rab5 (C8B1, #3547), and rabbit monoclonal anti-Rab7 (D95F2, #9367) primary antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Mouse monoclonal anti-β-actin 2A3 (sc-517582), mouse monoclonal anti-pANXA2 antibody (11.Tyr 24) (sc-135752), rabbit polyclonal anti-NOS2 (C-19, sc-649), rabbit polyclonal anti-ARG1 (H-52, sc-20150) antibodies, and protein A/G agarose were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Rabbit polyclonal anti-pANXA2 (phosphor-Tyr24) antibody was purchased from Signalway Antibody (College Park, MD, USA). Rabbit polyclonal anti-LS301 antibody was generated by Antibody Research Corporation (St. Charles, MO, USA) via immunization of New Zealand rabbits with keyhole limpet hemocyanin (KLH)-conjugated LS301 followed by antigen affinity purification of rabbit immunoglobulins from blood; antibody titer was tested by ELISA. Tyr24- phosphorylated ANXA2 protein (pANXA2) was generated via in vitro phosphorylation by overnight incubation of recombinant ANXA2 protein with recombinant ephrin B1 (kinase) and ATP in a buffer. The Vybrant Alexa Fluor 488 lipid raft labeling kit was purchased from Thermo Fisher Scientific (Waltham, MA, USA). [0089] Plasmids. Human Anxa2 cDNA subcloned into the pCMV6-AC-GFP vector was purchased from OriGene Technologies (Rockville, MD, USA). cDNA for the mutant pEZ- M98-GFP- Anxa2-Y23A (tyrosine replaced by alanine) was obtained from GeneCopoeia (Rockville, MD, USA). The recombinant plasmids were characterized by restriction digest, and the quality of the expressed recombinant protein was assessed by confocal microscopy for fluorescence integrity and by western blot. [0090] Cell lines and culture. The cancer cell lines 4T1, Lewis Lung Carcinoma (LLC), A431, A549, MDA-MB- 231, BxPC-3, and HL-60, were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). The 4T1-luciferase (4T1-luc) cell line was a gift from Dr. Katherine Weilbaecher (Washington University School of Medicine, St Louis, MO, USA). Human dermal fibroblasts were obtained from Coriell Cell Technologies (Camden, NJ, USA). Unless otherwise indicated, all cell lines were cultured at 37 °C in a 5% CO2 incubator in appropriate media supplemented with 10% FBS. [0091] Animals. Five to seven-week-old nude Balb/c or C57BL/6 mice were purchased from Charles River Laboratories (Wilmington, MA) and housed in designated animal facilities. Mice were fed ad libitum and inspected regularly. All animal studies were approved by the Washington University School of Medicine Animal Studies Committee (protocol numbers 25 KC0004-401-PC 019279/WO 20130207 and 20160207) and performed per humane care and use of research animals. Human tissues De-identified human breast tissue sections including malignant triple-negative (n = 4) and ER-positive (n = 4) and control normal tissues (n = 3) from the breast cancer patients were obtained from the Tissue Procurement Core (TPC)–Siteman Cancer Center and Pathology and Immunology at Washington University School of Medicine (St. Louis, MO, USA) per Washington University’s Institutional Review Board. [0092] Statistics. Unless otherwise noted, differences between sample means were analyzed by a two-tailed unpaired t-test with p<0.05 denoting statistical significance. Error bars denote standard error (standard deviation / square root of n); for replicate experiments, the sample size was at least (n) = 3 for each experimental group, and data were taken from different samples. Pearson’s correlation coefficient, image quantification, and analysis were calculated using the ImageJ software plugin Colocalization Finder. Statistical analyses of the images were performed using Origin Pro 8.0 software (OriginLabs). Example 1 - Synthesis of LS301 [0093] LS301 (cypate-cyclic (DCys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH) was synthesized from the linear GRD peptide, H-DCys(Acm)-Gly-Arg(Pbf)- Asp(tBu)-Ser(tBu)-Pro- Cys(Acm)-Lys(Boc)-OH, was prepared via a CEM Liberty Blue microwave peptide synthesizer (Matthews, NC, USA) on the Fmoc-Lys(Boc)-Wang resin. The resin (0.1 mmol) was swelled in dichloromethane for 1 hour before use. Fmoc-amino acids (0.5 mmol, 5 eq), coupling reagent (HBTU, 0.5 mmol, 5 eq), and diisopropylethylamine (DIEA, 1 mmol, 10 eq) were added to the resin, and the mixture was reacted for 15 min under microwave irradiation (100W, 90 °C). The resin was washed three times with DMF. The Fmoc group’s deprotection was carried out by treatment of 20% piperidine/DMF for 5 min under microwave irradiation (100 W, 90°C). The peptidyl resin was washed, and the peptide cyclized through the disulfide bridge with iodine (1.2 eq) in DMF for 90 min. [0094] Subsequently, cypate (3 eq) was conjugated to the cyclic peptide on a solid support in the presence of N,N’-diisopropylcarbodiimide (DIC, 5 eq) in DMF to afford the LS301 peptidyl resin. The resin was then treated with a trifluoroacetic acid cleavage cocktail: thioanisol: phenol: water (85:5:5:5, v/v/v/v) for 90 min at room temperature. The cleaved peptide product was concentrated in vacuo before purifying via reverse-phase HPLC (Gilson, Middleton, WI, USA). The molecular weight of the final product (1469 Da) was confirmed by electrospray ionization mass spectrometry with peaks observed at 1470 (M+1) and 735 (M+2/2). 26
Figure imgf000028_0001
Example 2 - Synthesis of LS301 peptide (LS637) [0095] LS637 (Ac-Cyclo(DCys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH), a non-fluorescent analog of LS301, was synthesized by a similar method as LS301, except acetic anhydride was used instead of cypate. The molecular weight of the final product (904 Da) was confirmed by electrospray ionization mass spectrometry with peaks observed at 905 (M+1) and 453 (M+2/2). Example 3 - Synthesis of ANXA2 blocking (LCKLSL) and scrambled (LGKLSL) peptides [0096] AnXA2 binding and scrambled peptides, LCKLSL and LGKLSL, were synthesized on solid support by standard automated Fmoc chemistry at room temperature. Starting with the Rink amide resin (30 μmol), amino acids were subsequently coupled to the resin using the appropriate Fmoc-protected amino acids (90 μmol) and coupling reagent 2-(1H-benzotriazol- 1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU, 90 μmol), hydroxybenzotriazole (HOBT, 90 μmol), and N,N-diisopropylethylamine (DIEA, 180 μmol). Cleavage of the peptide from the resin and concomitant removal of all protecting groups was achieved with 95% trifluoroacetic acid (TFA) and 5% water. The resulting product was purified by preparative HPLC using a Grace Vydac™ C-18 column (250 × 21.2 mm) with a UV detector at 254 nm. The desired compound was obtained by linear gradient elution using solvents A (0.1% TFA in water) and B (0.1% TFA in acetonitrile) from 90% to 10% over 30 minutes at 10 mL/min. Analytical HPLC characterized the purity of the peptide. The identity was confirmed by electrospray mass spectrometry (ES+MS): LCKLSL, calculated MW 676 g/mol, observed m/z 677 (M+1); LGKLSL calculated 629.80 g/mol, observed m/z 575 (M+1). Example 4 - Synthesis of KLH-conjugated LS301 for antibody production [0097] Synthesis of KLH-conjugated LS301 was accomplished via conjugation of the linker to LS301 followed by the conjugation of LS301-linker to KLH. For linker conjugation, 180 μmol (34.5 mg) 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC•HCl, 90 μmol, 41 mg), S-trityl-L-cystine tert-butyl ester hydrochloride (Chem-Impex, Wood Dale, IL, USA), and DIEA (180 μmol, 31.2 μL) in 2 mL DMF was sonicated until clear. The solution was added to 30 μmol LS301 and mixed overnight before the resin was filtered and washed with DMF and DCM. Finally, the product was cleaved from the resin and purified by reverse-phase HPLC. About 3 mg of the product was obtained (MW 1572 g/mol). Example 5 - Synthesis of LS301-doxorubicin conjugate (LS766) 27
Figure imgf000028_0002
KC0004-401-PC 019279/WO [0098] Doxorubicin (3 eq, LC Laboratories, Woburn, MA) was added to a mixture of the LS301 peptidyl resin, (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (HATU, 6 eq), and DIEA (6 eq) in DMF. After 12 hours of reaction, the resin was treated with a cleavage cocktail consisting of TFA: thioanisol: phenol: water (85:5:5:5, v/v/v/v) for 90 minutes at room temperature. The cleaved peptide product was concentrated in vacuo, then purified on reverse-phase HPLC (Gilson, Middleton, WI, USA) to obtain LS766 (doxorubicin-cypate-cyclic(DCys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys- OH), M/W 1995 g/mol, ESI-MS observed 998 (M+2/2) and 666 (M+3/3). [0099] Following the procedure above, the LS838 peptidyl resin can be reacted with doxorubicin to yield doxorubicin-cypate-cyclic(DCys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH, comprising the structural formula 28
Figure imgf000029_0001
KC0004-401-PC 019279/WO .
Figure imgf000030_0001
[0100] In general, immunoprecipitation experiments were performed by incubation of LS301 with either 4T1 cell lysates or live 4T1 cells, followed by pulldown of LS301-associated proteins with anti-LS301 antibody and protein A/G agarose beads. For cell lysis, whole 4T1 cells were homogenized in a 15-mL tube on homogenizer using 3 mL of homogenizing RIPA buffer (10 mM Tris-HCl pH 8.0, 140 mM NaCl, 1mM EDTA, 0.5mM EGTA, 1% Triton X- 100, 0.1% sodium deoxycholate, 0.1% SDS, 1 mM PMSF, protease inhibitor cocktail III (10 μL/mL Calbiochem, 539134), and phosphatase inhibitor cocktail 1 (10 μL/mL, Sigma, P2850)). Lysates were clarified by centrifugation at 15,000 x g. The protein concentration was determined by BAC protein assay (Bio-Rad) with bovine serum albumin (BSA) as a standard. The supernatants were stored at -80 °C. [0101] For immunoprecipitation studies with cell lysates, 400 µg of cellular protein extract was incubated with LS301 (40 µM) in 0.5 mL 1X phosphate-buffered saline (PBS) for 2 hours at room temperature. For immunoprecipitation studies with live cells, 4T1 cells in a T- 75 flask at about 80% confluence were incubated with LS301 (10 µM) for 24 h at 37 °C; the cells were rinsed with PBS, harvested using trypsin-EDTA, and lysed as above. Next, 10 µg of anti-LS301 polyclonal Ab (Antibody Research Corp., St. Charles, MO) was added to the sample and incubated for 18 hours at 4 °C. Forty µL of protein A/G PLUS-agarose beads (Santa Cruz Biotechnology) was then added and incubated overnight at 4 °C under rotation. 29 KC0004-401-PC 019279/WO The pellets were collected by centrifugation (1200 x g) and washed three times with 1X PBS for 5 minutes. The pellets were resuspended in 60 µL of 1 x electrophoresis loading buffer containing sodium dodecyl sulfate (SDS). Samples were boiled for 7 min. Then 30 µL of the samples were resolved on TGX gels (Any Size gradient TGX gels, Bio-Rad) and visualized by near-infrared fluorescence imaging using the Pearl Imaging System, Coomassie staining, or Western blot analysis. [0102] For fluorescent gel binding experiments, 1 μg recombinant annexin A2 protein was incubated with LS301 or cypate (10 μM) at 37 °C for the periods indicated. Samples were subjected to SDS-PAGE on precast gels (Bio-Rad, Hercules, CA), and gels were imaged for near-infrared fluorescence (800 nm) using a Pearl Small Animal Imager (LI-COR Biosciences, Lincoln, NE, USA). For competitive binding experiments, Annexin A2-GST, annexin A1, or annexin A3 (1.0 μg protein per sample) was pre-incubated with or without 2.5 mM of LS637 (non-fluorescent LS301 analog; blocking peptide) in binding buffer (25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM MgCl2, 1 mM CaCl2) for 1 hour at 37 °C. Ten μM LS301 was then added to each sample. After incubation for 2 hours at 37 °C, proteins were subjected to SDS-PAGE and imaged fluorescently. For some experiments, LS301 was incubated with 4T1 tumor cells before SDS-PAGE. [0103] Microscale thermophoresis (MST) studies biomolecular interactions in an aqueous environment without immobilization. MST exploits the directed movement of molecules along a microscopic temperature gradient. The movement rate and direction of movement depend on the species’ size, charge, and solvation shell under study. Changes in this environment can change the thermophoretic movement of the species. MST can distinguish very small changes that occur, for example, when a small fluorescent molecule binds to the surface of a protein. [0104] MST was performed using the Monolith NT.115 (Nanotemper Technologies, Munich, Germany). The appropriate agent(s) and LS301 stock samples were denatured in MST-1 buffer (50 mM Tris-HCl, pH 7.4; 150 mM NaCl; 10 mM MgCl2; 2% DMSO; 0.1% Tween- 20; 4% SDS, 4 mM dithiothreitol (DTT)) at 97 °C for 10 min. Fluorescence was assessed in MST-2 buffer (50 mM Tris-HCl, pH 7.4; 150 mM NaCl; 10 mM MgCl2). The protein was prepared in MST-2 buffer, and 20 μL serial dilutions were made with protein concentration ranging from 3.05 pM to 50 nM (pANXA2) or 3.81 pM to 62.5 nM (ANXA2). All samples were loaded into NT.115 standard capillaries. The analysis was performed at 37 °C, 30% LED power, 60% MST power, and a constant LS301 concentration of 20 nM. The apparent 30 KC0004-401-PC 019279/WO Kd values were calculated from fragment concentration-dependent changes in normalized fluorescence (Fraction Bound) of LS301 after 5 seconds of thermophoresis based on mass action law using the MO Affinity Analysis v2.3 software. [0105] For Western blot analysis, cells were homogenized using an ultrasonic processor in radioimmunoprecipitation assay (RIPA) buffer (10 mM Tris-HCl, pH 8.0; 140 mM NaCl; 1 mM ethylenediaminetetraacetic acid (EDTA); 0.5mM egtazic acid (EGTA); 1% Triton X- 100; 0.1% sodium deoxycholate; 0.1% SDS; 1 mM PMSF). Total cell lysates were clarified by centrifugation. Total cellular protein or pure proteins were denatured in SDS gel-loading buffer (100 mM Tris-HCl, 200 mM DTT, 4% SDS, 0.2% bromophenol blue, and 20% glycerol) for 7 minutes at 100 °C and then separated on 12% TGX gel (Bio-Rad, Hercules, CA, USA). After electrophoresis, proteins were transferred to PVDF-FL membrane using an EC140 Mini Blot Module (Thermo EC, Holbrook, NY, USA) apparatus. The membrane was treated with Odyssey blocking buffer for 1 hour at room temperature, followed by incubation with rabbit monoclonal anti-annexin A2 (D11G2, Cell Signaling Technology, Inc.) and mouse monoclonal pannexin II (11.Tyr24; Santa Cruz Biotechnology) or rabbit ANXA2 (Phospho- Tyr24; MyBioSource, Inc.) primary antibodies in Odyssey blocking buffer at 4 °C overnight. After washing three times for 10 minutes each in PBS with 0.1% Tween-20 (PBS-T), the membrane was incubated for 1 hour with diluted IRDye 680RD goat anti-rabbit IgG or IRDye 800CW goat anti-mouse IgG in Odyssey blocking buffer. The membrane was then washed three times for 10 minutes each in PBS-T and imaging using the Odyssey CLx imaging system. The membrane was probed again with mouse monoclonal anti-actin (Santa Cruz Biotech) or rabbit polyclonal anti-Actin (Cell Signaling Technology) for immunoblotting and imaging. In some experiments, the Coomassie blue stain was used (Bio- Rad, Hercules, CA, USA). Example 7 - Proteomics [0106] For mass spectrometric protein identification, the appropriate bands were excised from protein gels and submitted for LC-MS/MS protein identification at the Donald Danforth Plant Science Center, Proteomics & Mass Spectrometry Facility (St. Louis, MO, USA). Samples were digested with trypsin and run on the LTQ-Orbitrap Velos using the 1-hour LC- MS/MS method. [0107] All MS/MS samples were analyzed using Mascot (Matrix Science, London, UK; version 2.4.1. Mascot was set to search the cRAP_20110301 database and NCBInr database (selected for Mus musculus, 174101 entries), assuming the digestion enzyme to be trypsin. 31 KC0004-401-PC 019279/WO Mascot was searched with a fragment ion mass tolerance of 0.80 Da and a parent ion tolerance of 15 ppm. Deamidated asparagine and glutamine, oxidation of methionine, and carbamidomethyl of cysteine were specified in Mascot as variable modifications. [0108] Scaffold (version Scaffold_4.3.4, Proteome Software Inc., Portland, OR, USA) validated MS/MS-based peptide and protein identifications. Peptide identifications were accepted if they could be established at greater than 80.0% probability by the Scaffold Local FDR algorithm. Protein identifications were accepted if they could be established at greater than 99.0% probability and contained at least two identified peptides. [0109] Protein probabilities were assigned by the Protein Prophet algorithm (Nesvizhskii et al., Anal. Chem.2003;75(17):4646-58). Proteins that contained similar peptides and could not be differentiated based on MS/MS analysis alone were grouped to satisfy parsimony. Proteins sharing significant peptide evidence were grouped into clusters. [0110] In general, cultured cancer cells were grown and incubated with the indicated amount of probe in Lab-Tek 8-chambered slides for the indicated time durations. Cells were washed with PBS at selected time points and underwent conventional slide preparation, ICC if indicated, and viewing under epifluorescence or confocal. For live-cell imaging of LS301, internalization at 37 ºC in human dermal fibroblasts (Coriell Cell Technologies) and tumor cells including 4T1luc, MDA-MB-231, 5TGM, A549, and HL60 (ATCC) cells were cultured in appropriate media supplemented with 10% FBS and penicillin/streptomycin and incubated at 37 °C in 5% CO2 incubator. The cells were grown on Lab-Tek 8-chamber slides (Nunc Inc. Rochester, NY) in the culture medium overnight before the experiment. The compounds were dissolved in 20% DMSO in 10 μM PBS (pH 7.4) (Sigma, St. Louis, MO) for stocking solution and mixed with culture medium to reach the final concentration. Cells were incubated with 1 μM compounds in chamber slides at different time points at 37 ºC. After LS301 treatment, the culture media was removed and followed by adding culture media containing SYTO 13 nuclear dye from (Thermo Fisher Scientific, Waltham, MA) for 45 minutes at 37 °C. For assessing cellular colocalization of LS301 with lipid rafts, the Vybrant AlexaFluor 488 Lipid Raft Labeling Kit (Thermo Fisher Scientific, Waltham, MA, USA) was used per the manufacturer’s instructions. Briefly, 4T1 cells were grown overnight in eight- chambered slides (Thermo-Fisher Scientific, Waltham, MA, USA) in phenol-free DMEM + 10% FBS. LS301 was added to 4T1 cells at 5 μM for 1 hour, followed by staining with AlexaFluor 488-conjugated cholera toxin B for 10 minutes at 4 °C, washing with PBS, and followed by adding anti-cholera toxin B antibody for 15 min at 4 °C to crosslink labeled lipid 32
Figure imgf000034_0001
rafts and washing with PBS. Slides were mounted with Fluoromount-G mounting medium (Thermo-Fisher Scientific) before viewing. LS301 and AlexaFluor 488 fluorescence were imaged using the cypate filter set (Ex/Em 750–800 nm/818–873 nm) and fluorescein isothiocyanate (FITC) filter set (Ex/Em 460–500/510–560nm), respectively. [0111] For live-cell imaging of LS301 cell surface binding at 4 ºC, at first, the tumor cells and fibroblast cells were incubated with 1 μM of LS301 in culture medium for 1 hour at 4 ºC in 35 mm glass-bottom culture dishes (MatTek Co., Ashland, MA, USA). In the 1.5 mL tubes, 1:250 monoclonal rabbit anti-ANXA2 antibody (Cell signaling Technology. Inc. Danvers, MA, USA) was incubated with 1:500 Alexa 488 anti-rabbit antibody (Thermo Fisher Scientific), and 1:250 mouse anti-pANXA2 antibody (Santa Cruz Biotechnology, Inc. Santa Cruz, CA) was incubated with 1:500 Alexa 546 donkey anti-mouse antibody (Thermo Fisher Scientific, Waltham, MA, USA) for 1 hour at 37 ºC respectively. The cells treated with LS301 were washed twice and incubated with the first and second antibodies complexes for another hour at 4 ºC. Secondary antibody treatment alone served as controls. After washing twice, the cells were imaged using an FV1000 confocal microscope. FV1000 software determined the mean fluorescence intensities (Ex/Em = 488/510–530 nm; Ex/Em = 546/555– 600 nm, 633/645–700 nm; Ex/Em = 780/805–830 nm). [0112] For LS301 cell internalization studies involving insulin treatment, cells were grown on Lab-Tek slides overnight. HEK 293T cells (or in some cases 4T1 cells) were maintained in DMEM medium supplemented with 10% fetal bovine serum and 100 units/mL penicillin. After DNA transfection, the HEK 293TAnxa2-WT-GFP and HEK 293TAnxa2-Y23A-GFP cells were maintained in Dulbecco’s Modified Eagle’s medium (DMEM) medium with 500 μg/mL G418, 10% fetal bovine serum (FBS), and 100 units/mL penicillin. The medium was changed, and cells were grown in serum-starved conditions overnight. Cells were then cultivated with or without 100 nM insulin for 4 h. Then 1 μM of LS301 was added to each sample to incubated for 4 hours or 24 hours at 37 °C. Cells were imaged using an FV1000 confocal microscope, and the mean fluorescence intensities (Ex/Em = 488/495 nm; Ex/Em = 590/617 nm; Ex/Em = 780/805–830 nm) were determined using the FV1000 software. [0113] For Western blot analysis of insulin/PP2-induced change in cellular pANXA2 levels, 4T1 cells were treated with insulin or PP2 as above, lysed, and subjected to SDS-PAGE analysis. For mechanistic LS301 cell internalization experiments with endocytosis pathway inhibitors, 4T1 murine breast cancer cells were seeded in an 8-well plate at a density of 5000 cells/well with DMEM media supplemented with 10% FBS and penicillin/streptomycin and 33 KC0004-401-PC 019279/WO incubated overnight at 37 °C. The next day, culture media was removed, followed by pre- treatment of cells with chlorpromazine hydrochloride at 13 μg/mL for 30 min, filipin complex at 3.2 μg/mL for 1 hour, or amiloride hydrochloride at 300 μM for 1 hour in a 300-μL volume of cell culture media. Next, the drug-containing media was removed after incubation and replaced with fresh culture media containing 6 μM or 0.9 μg/mL of LS301 in complete DMEM for 1 hour at 37°C. [0114] For Pitstop 2 studies, 4T1 cells were incubated with Pitstop 2 reagent at 25 μM for 20 minutes, followed by incubation with LS301 at 2 μM for 1 or 3 hours. After LS301 treatment, the culture media was removed, followed by adding culture media containing SYTO 17 nuclear dye for 40 minutes at 37 °C. Finally, nuclear stain media was removed. Cells were washed 2 times with PBS. They were later re-suspended in culture media for live-cell confocal microscopy imaging using Ex/Em = 780/805–830 nm for LS301 and Ex/Em = 621/634 nm for SYTO 17. ImageJ software corrected total cell fluorescence (CTCF) value from multiple cells per condition and summarized data with a box plot. Background signal was maintained at zero background for the analysis. [0115] For LS301 intracellular trafficking experiments, 4T1 cells were cultured overnight using similar parameters as above. The next day culture media was removed, followed by incubation of cells with 200 μL of 4 μM LS301 in complete culture media for 5 hours at 37 °C. At the end of this internalization process, LS301-containing media was removed, followed by three PBS washes. Fluorescence organelle staining and imaging were performed per manufacturers’ protocols using VectaCell Rhodamine 123 for mitochondria, Lysotracker for lysosomes, BODIPY TR Ceramide for Golgi apparatus and CellLight Peroxisome-GFP, BacMam 2.0 for peroxisomes. [0116] Immunocytochemistry was performed with 4% paraformaldehyde fixation, permeabilization with 0.1% Triton X- 100 solution, and blocking with 5% normal goat serum and 1% BSA in 1X PBS blocking solution. Primary monoclonal antibodies against Rab5 (1:333) for early endosomes and Rab7 (1:333) for late endosomes were added to cells overnight at 4 °C. The next day, wells were washed three times in PBS and then incubated with secondary Ab (goat anti-rabbit Dylight 550 nm (1:500)) for 1 hour. Finally, wells were washed three times and then imaged without coverslipping with a water immersion lens at 60x. ImageJ performed colocalization analyses to obtain the Pearson’s correlation coefficient (PCC) and the scatterplot inset. [0117] For in vitro blocking studies of LS301, 4T1 luc/GFP cells were treated for 24 h with 34 KC0004-401-PC 019279/WO LS301 alone, blocking peptide LCKLSL plus LS301, scrambled blocking peptide LGKLSL plus LS301, or unlabeled LS301 analog LS637 plus LS301. Cells were prepared for confocal microscopy. [0118] For co-culture experiments on intercellular pANXA2/ANXA2 transfer, fibroblasts (FB) (5 × 104 cells/well) were seeded on a 35-mm glass-bottom Petri dish (MarTek Cor. Ashland, MA, USA) overnight before seeding of 5×104 of cancer cells and further culture overnight before the experiment. LS301 (1 μM) was incubated with co-cultures of FB and ANXA2-YFP transfected HL60 cells, co-cultures of FB and wild-type HL60 cells, or co- cultures of FB and 4T1-GFP cells at 37 °C for 3 hours and then washed twice with 0.01 M PBS (pH 7.4). Then, the cells were fixed with 4% paraformaldehyde for 10 minutes. [0119] For immunocytochemistry, slides were blocked with appropriate serum for 35 minutes or 5% non-fat milk PBS (pH 7.4) overnight at 4 °C. Then, the slides were incubated with 1:500 monoclonal rabbit anti-ANXA2 antibody (Cell signaling Technology. Inc. Danvers, MA, USA), 1:500 monoclonal mouse anti-pANXA2 antibody (Santa Cruz Biotechnology, Inc. Santa Cruz, CA, USA), and/or 1:250 monoclonal mouse anti-vimentin antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA) for 1 hour at 37 °C. After washing twice with PBS, the tissue sections were incubated with 1:1000 Alexa 488 anti-rabbit antibody (Thermo Fisher Scientific, Waltham, MA) and/or 1: 1000 TRITC labeled donkey anti-mouse IgG in MPBS (Jackson ImmunoResearch Lab, West Grove, PA, USA), for 1 hour at 25ºC. Finally, slides were imaged under a confocal microscope as described in the Fluorescence Microscopy methods section. Fluorescence microscopy was performed using either an Olympus FV1000 confocal microscope (Olympus Corp., Tokyo, Japan) with filters/channels as follows: Ex/Em = 488/510–530 nm; Ex/Em = 546/555–600 nm, 633/645–700 nm; Ex/Em = 785–825 nm /805–830 nm; or epifluorescence microscope with filters/channels as follows: 4′,6-diamidino-2-phenylindole, (DAPI, Ex/Em = 330–385/420 nm), FITC (Ex/Em = 460– 500/510–560 nm), Texas Red (Ex/Em = 542–582/604–644 nm), and cypate (Ex/Em = 750– 800/818–873 nm), using exposure times 1 to 30 seconds and sensitivity settings ISO200- ISO1600. The same parameters were used for control and treatment groups unless otherwise indicated. ImageJ software (National Institutes of Health, Bethesda, MD, USA) was used for image processing. [0120] In general, cells were fixed in Lab-Tek 8-chamber slides with 3% paraformaldehyde in PBS solution for 15 minutes. Nonspecific binding sites were blocked by PBS containing 10% normal donkey serum (blocking solution) for 1 hour. Cells were incubated overnight at 4 35
Figure imgf000036_0001
KC0004-401-PC 019279/WO °C with the appropriate primary antibodies after three 5-minute PBS washes. Cells were then incubated at room temperature for 1 hour with diluted secondary donkey anti-rabbit antibody (Alexa Fluor 594) in blocking solution. Slides were counterstained and mounted with coverslips for imaging on an FV1000 confocal microscope. Mean fluorescence intensities (Ex/Em = 590/617 nm) on the slide were determined using the FV1000 software. Cell transfection and transduction ANXA2-YFP expressing HL60 cells were generated by lentiviral transduction as follows. Human full-length ANXA2 cDNA in the pCMV6-AC-GFP vector was purchased from OriGene Technologies (Rockville, MD, USA). BamHI, XhoI, and AgeI restriction enzymes and Quick Ligation kit were purchased from New England Biolabs (Ipswich, MA, USA). An FCIV lentiviral vector containing IRES-Venus YFP under the ubiquitin promoter was obtained as a generous gift from Dr. Mingjie Lee at the Hope Center, Washington University School of Medicine (St Louis, MO, USA). The ANXA2 cDNA was subcloned into the FCIV lentiviral vector by restriction digest of the ANXA2 cDNA vector with BamHI and XhoI enzymes, restriction digest of the FCIV vector with BamHI and AgeI enzymes, and ligation using Quick Ligation per manufacturer’s instructions. Klenow fragment and dNTP nucleotide mix (New England Biolabs) was used to fill sticky ends. [0121] HEK 293T cells (ATCC) were cultured overnight in DMEM with 10% fetal bovine serum, 100 units/mL penicillin 100 μg/mL streptomycin before transfection. DNA constructs were transfected into HEK 293T cells using Genejuice (EMD Millipore, Burlington, MA) per manufacturer’s instructions. Culture supernatants were collected 40 hours after transfection, mixed 1:1 with Iscove’s Modified Dulbecco’s medium (IMDM) (Thermo Fisher Scientific) supplemented with 10% fetal bovine serum (FBS) and 1% Streptomycin, and added to HL60 cells in 6-well plates. At 72 hours post-transduction, the HL60 cells were checked under a confocal microscope for fluorescence. Populations of HL60 cells strongly expressing ANXA2-YFP were analyzed and collected by flow cytometry using a BD FACS AriaII Cell Sorter at a core laboratory at Washington University in St. Louis School of Medicine (St. Louis, MO, USA). Cell viability was checked by luciferase MTS assay as described in the MTS Assay section of methods. Transwell culture and extracellular vesicle transfer studies For transwell studies, human dermal fibroblasts (300,000 cells/well) were cultured alone in 6- well plates, or transwell with 4T1 cells (100,000 cells/insert) suspended within transwell inserts (3 μm pore polycarbonate membrane, MilliporeSigma, St. Louis, MO, USA). Cells were plated and grown in extracellular vesicle-depleted media for 72 hours and subsequently stained for pANXA2 using rabbit anti-pANXA2 primary antibody (15 min. at 4 °C) and 36 KC0004-401-PC 019279/WO donkey anti-rabbit AlexaFluor 594 conjugated secondary antibody (15 min. at 4 °C) at 1:500 dilutions, and visualized by fluorescence microscopy using the Texas Red filter set (Ex/Em = 542-582/604–644 nm). [0122] Extracellular vesicle-depleted media were generated by collecting flow-through after centrifugal filtration of complete DMEM media using Amicon Ultra-100K (100 kDa molecular weight cutoff membrane) devices (MilliporeSigma, St. Louis, MO). For extracellular vesicle transfer studies, supernatants from 4T1 cells cultured in extracellular vesicle-depleted media for 16 hours were harvested. The 4T1-derived extracellular vesicle fraction was concentrated by serial centrifugal filtration on Amicon Ultra-100K devices. The 4T1-derived extracellular vesicle fraction was then added to fibroblasts (60 μg/well) for 24 h, with untreated fibroblasts as control. LS301 was then added at 5 μM for 1 hour, and LS301 uptake was visualized by fluorescent microscopy using the cypate filter set (Ex/Em 750–800 nm/818– 873 nm). A separate group of fibroblasts with or without added 4T1-derived extracellular vesicles was stained for pANXA2 using rabbit anti-pANXA2 primary antibody and donkey anti-rabbit AlexaFluor 594 conjugated secondary antibody at 1:500 dilutions and visualized by fluorescence microscopy as above. [0123] For the MTS Assay, 6,0004T1 or fibroblast cells (n = 3) were seeded in 96-well plates and cultured in DMEM with 10% FBS at 37 °C overnight before treatment with different concentrations of LS301 for 48 hours or 72 hours. Cell viability was evaluated using CellTiterGlo Luminescent cell viability assay (Promega, USA) per manufacturer’s instructions by standard plate reader for luminescence. The luminescence intensity from cells was normalized to reagent alone. [0124] Overnight cultured 4T1 tumor cells in 96-well plates were treated with 0.5 μg/ml of doxorubicin (DOX) for 3 hours as a positive control. After treatment with different concentrations of LS301 for 48 hours or 72 hours, the cells were trypsinized, centrifuged, and washed twice with PBS (pH 7.4) before cell cytometry. Apoptotic cell death was determined using the FITC Annexin V Apoptosis Detection kit (Biolegend, USA) per manufacturer’s instructions. Data were acquired and analyzed using BD FACS AriaII Cell Sorter (Beckman Coulter, USA) at the core laboratory at Washington University in St. Louis School of Medicine. Example 8 - Tissue analysis [0125] In general, tissues of interest were harvested and frozen at -80°C in Optimal Cutting Temperature (OCT) media (storage at ≤-20°C). Frozen sections were cut at 10-μm thickness, 37 KC0004-401-PC 019279/WO and slides were stored at -40 °C. The LS301 fluorescence signal was then assessed by fluorescence microscopy using Cy7 filters or confocal microscope. Sections were subsequently fixed with 4% paraformaldehyde for 10 minutes. [0126] For immunohistochemistry, slides were blocked with appropriate serum for 35 minutes or with 5% non-fat milk PBS (pH 7.4) overnight at 4 °C and incubated with primary antibody overnight at 4 °C or 1 hour at 37 °C. For ANXA2/pANXA2 studies, tissue sections were incubated with 1:250 monoclonal rabbit anti-ANXA2 antibody (Cell signaling Technology. Inc. Danvers, MA) or 1:250 monoclonal mouse anti-pANXA2 antibody (Santa Cruz Biotechnology, Inc. Santa Cruz, CA). After washing twice with PBS, the tissue sections were incubated with 1:1000 AlexaFluor 488 anti-rabbit antibody (Thermo Fisher Scientific, Waltham, MA) and 1:800 TRITC labeled donkey anti-mouse IgG in MPBS (Jackson ImmunoResearch Lab, West Grove, PA, USA) for 1 hour at 25 ºC respectively. Slides were washed again and stained with Hoechst or DAPI nuclear stains for 5 min or stained with 1:4000 nucleus dye ToPro3 (Thermo Fisher Scientific, Waltham, MA) for 45 minutes at 37 °C. After final washes, a coverslip with aqueous fluorescence-saving mounting media was applied before imaging. Images were co-registered with others from the same sections to allow co-localization analysis. [0127] For H&E staining, the same section or an adjacent frozen section was fixed for 10 min in 4% paraformaldehyde solution (Sigma, St. Louis, MO, USA) and stained with Harris hematoxylin for 90 seconds and with eosin (Sigma, St . Louis, MO) for 15 seconds, and then washed with tap water for 5 min. The tissue sections were mounted with Richard-Allan Scientific mounting medium (Thermo Fisher Scientific) and coverslips for microscopic examination. [0128] For Z-stack imaging, the frozen 4T1 tumor tissue sections (10 μm thickness) were warmed in PBS (pH 7.4) for 5 min and then stained with SYTO-13 green fluorescent nuclear stain (Thermo Fisher Scientific, Waltham, MA) for 45 min at 37 ºC. Slides were imaged under a confocal microscope at Ex/Em = 488/510–530 nm for SYTO-13 and Ex/Em = 780/805–830 nm for detection of LS301. Each step of the Z-stack imaging ranged from 0.8 μm to 1.6 μm. [0129] For quantitative correlation of pANXA2 expression to LS301 fluorescence in 4T1 luc-GFP, HT1080, and BxPC-3 tumors, the ImageJ plugin Colocalization Finder was applied to three representative tumor areas for each tumor type and determined the Pearson’s correlation coefficients for pANXA2/LS301 signal overlap. For quantitative analysis of 38
Figure imgf000040_0001
pANXA2 or ANXA2 signal within human tissues, FV1000 software was applied to calculate the fluorescence intensity at different areas in tumor tissues (n = 3 mice /group, n = 15 view spots). Ten lines were drawn in each view site, three view sites were chosen in tissue sections using 10x and 60x objective lens to do quantitative analysis. All values are the mean + S.D., and Student’s two-tailed unpaired t-test compared the difference. P < 0.05 was accepted as statistically different. [0130] For quantitative analysis of intratumoral LS301 levels in 4T1 luc/GFP, HT1080, and BxPC-3 tumors, mean fluorescence was measured for six independent views, including peripheral and core tumor tissues for each tumor type using ImageJ software (Analysis>Measure function). For quantitation of intratumoral distribution (periphery vs. core) of LS301 in 4T1 luc/GFP tumors, mean fluorescence was measured for three independent views of peripheral tumor tissues versus three independent views of core tumor tissues using ImageJ. [0131] To establish the various tumor models, nude, Balb/c, or C57BL/6 mice were injected with 105–107 tumor cells subcutaneously on unilateral or bilateral flanks. In some cases, spontaneous tumor models were used. Animals injected subcutaneously with saline served as sham tumor controls. For near-infrared imaging, 100 μL volumes of 60 μM LS301 or cypate in PBS were injected intravenously by lateral tail vein, and animals were imaged using a Pearl Small Animal Imager (LI-COR Biotechnology, Lincoln, NE, USA) at the indicated time points. In some experiments, probes were formulated in 1% albumin in PBS. Animals were shaved using commercially available hair removal equipment and products before imaging. For experiments involving further tissue analysis, animals were anesthetized with isoflurane and euthanized by cervical dislocation under anesthesia. Organs were harvested by surgical dissection. For quantitative organ biodistribution studies, individual regions of interest were drawn around each organ of interest, and fluorescence was quantified using the Pearl Small Animal Imager software. [0132] For flow cytometric analysis of cells from LS301-labeled 4T1 tumor xenografts, after injecting LS301 into 4T1 luc/GFP tumor-bearing mice at the time points indicated, the mice were imaged, euthanized, and tumors were excised and kept at -80 °C until analysis. Tumors were thawed, cut into small pieces, and incubated in trypsin for 1 to 2 hours at 37 °C. The tissue was dissociated and filtered with a 40-μm cell strainer tube (Falcon 352235) to collect the sample and washed with PBS (pH 7.4) three times and further centrifuged for collection. The cells were sorted using a BD FACS AriaII Cell Sorter at a core laboratory at Washington 39 KC0004-401-PC 019279/WO University in St. Louis School of Medicine (St. Louis, MO, USA). Untreated cells, cells treated with LS301 (4 μM for 24h), wild-type 4T1 cells (without GFP), and 4T1 luc/GFP cells were used for compensation and gating. The sorted cells were classified and collected in fresh IMDM supplemented with 10% FBS and 1% streptomycin. Example 9 - Fluorescence imaging-guided surgical margin assessment [0133] Three 6-week old Balb/c mice were implanted with 100,0004T1-Luc/GFP murine breast cancer cells orthotopically in the mammary fat pad below the right front leg. Seven to ten days post-implantation, all three mice were injected with 100 μL of calcium-formulated LS301 (5 mM Ca/60 μM LS301) via tail vein injection. Twenty-four hours post-injection, these mice were subjected to image-guided surgery using the CancerVision™ Goggle (CVG) system. [0134] Mice were maintained under anesthesia using a continuous 5% isoflurane gas inhalation. A midline incision was made from the sternum to the base of the right front leg to create a skin flap along the tumor guided by LS301 fluorescence visualized using the CVG. The skin flap was deflected to expose and resect the tumor under fluorescence guidance. The surgical bed was surveyed for residual fluorescence, and additional fluorescent tissue identified using the CVG was resected. [0135] In vivo confirmatory closed-field fluorescence images were obtained using the Pearl small animal imaging system (Li-Cor Biosciences, Lincoln, NE, USA) to ensure no residual fluorescence was left behind. Additional in vivo validation of complete tumor resection was obtained using quantitative bioluminescence imaging using the IVIS imaging system (PerkinElmer, Waltham, MA, USA) after injecting the mice with luciferin intraperitoneally before the start of surgical resection. All resected tissues were embedded in Tissue-Tek OCT (Sakura Finetek, Torrance, CA, USA) and frozen for preservation. All preserved tissues were sectioned into 10 µM thick slices using a cryo-microtome and immediately mounted onto microscope slides. Consecutive tissue section slides were observed for LS301 fluorescence using the epifluorescence microscope (Olympus B61). These sections were then stained using hematoxylin and eosin to identify cancerous tissue and imaged under brightfield conditions using the epifluorescence microscope to find correspondence between the fluorescence and cancerous regions. [0136] Fluorescence quantification of images obtained using the CVG was done using ImageJ. Tumor-to-background ratio (TBR) calculations and statistical analyses were done using Origin Pro 8.0 software (OriginLabs). Means and standard errors denoted TBR average 40 KC0004-401-PC 019279/WO values and errors, respectively. Paired student’s t-tests compared the TBRs before and after tumor resection to measure complete tumor resection and lack of residual fluorescence in the surgical bed. A p-value < 0.05 was statistically significant. Example 10 - In vivo LS301-doxorubicin drug conjugate therapy [0137] Athymic nude mice (n=3 per group) were subcutaneously implanted with HT1080 human fibrosarcoma cells. Tumor growth was measured by a caliper twice weekly. On days 8, 11, and 15 post tumor implantation, mice were intravenously injected with either control (no treatment), doxorubicin alone, or LS301-doxorubicin drug conjugate at a dose of 0.5 μmol/kg. Mice were euthanized when tumors reached 2 cm in diameter in any direction. Survival was plotted using Kaplan-Meier curves, and statistical significance was assessed by a Logrank test of untreated control vs. LS301-doxorubicin treatment. Example 11 – DMSO-Based Formulation [0138] At room temperature (~25 °C), 200 µL of DMSO was added to 0.5 mg LS301 and vortexed for about 1 minute to homogenize. The solution was centrifuged to remove any non- dissolved components. The concentration was measured via absorbance. [0139] Generally, solution concentrations ranged between 290 and 330 µM. This concentrated solution was diluted dropwise into 800 µL phosphate-buffered saline (PBS) calculated such that the final concentration was 60 µM LS301 and that the final concentration was 20% DMSO/ 80% PBS by volume (“20% DMSO”). Example 12 – Ethanol-Based Formulation [0140] A 1:1 PEG400/Ethanol solution (v/v) was prepared by mixing 4 mL of PEG400 and 4 mL of ethanol and vortexing at high speed for 1 minute.200 µL of this 1:1 solution was added to about 0.5 mg of LS301 to produce a transparent solution (>300 µM). No precipitate was observed after centrifugation. The solution was then added dropwise to a PBS/Ethanol solution, such that the final solution had a concentration of 60 µM, 10% PEG400, 10% ethanol solution, 80% PBS (“1:1 PEG400/Ethanol”). Example 13 - NIR Contrast Evaluation For DMSO and Ethanol-Based Formulations [0141] LS301 exhibited variable contrast when placed in different media, with demonstrable significantly increased contrast in 20% PEG400 (contrast in vivo over time) and 3e (contrast ex vivo). LS301 cleared via renal and hepatic mechanisms. [0142] LS301 placed in 20% PEG400 solutions demonstrated increased tumor uptake compared to renal or hepatic clearance. Compared to DMSO based solvents, all the ethanol- 41 KC0004-401-PC 019279/WO based solvents demonstrated a more rapid increase in bladder fluorescence in vivo, suggesting increased renal clearance by ethanol-based solvents. No difference in histology between the various solvents was seen. Example 14 - Performance Test of PEG/Ethanol Formulation in Different Tumor Models [0143] LS301’s solubility in ethanol ranged near the cutoff for producing a 20% ethanol/PBS solution. Besides, although 20% PEG400 + 20% ethanol had a higher contrast than the other solvent basis, efforts were made to reduce the non-aqueous component and increase the ease in producing the stock solution. The 1:1 PEG400/ethanol solution could maintain stability up to about 1-to-16 dilution. In vivo, a screening test was done by IV injection of 60 µM solution into a Balb/c mouse with two 4T1luc tumors dorsally injected. The data demonstrated increased contrast compared to 20% DMSO. To confirm LS301’s multitumor affinity in the new solvent, solutions were tested against HT1080, A431, and DBT tumor-bearing mice. LS301 in 20% 1:1 PEG400 demonstrates a tumor-to-muscle ratio of greater than 10-fold in all three models and visible over time and ex vivo studies. Example 15 - HSA/MSA Formulation [0144] One percent human serum albumin (HSA) or mouse serum albumin (MSA) was prepared by diluting 100 mL 25% HSA to 2.5 L with sterile water or dissolving 1 g MSA in sterile water. One gram LS301 was suspended in 10 L of 1% HSA or 1% MSA solution. The solution vortexed on a shaker for 30 minutes. The resulting LS301-HSA or LS301-MSA formulation was sterile filtered through Steriflip™ filter. Aliquots of the filtered sample were taken. The concentration of LS301 and HSA or MSA was confirmed via absorbance. The sample vials containing filtered LS301-HSA or LS301-MSA formulation were placed on dry ice to quick-freeze the sample. The frozen sample vials were placed onto the lyophilizer’s tray dryer, then loaded into the lyophilizer to dry overnight (at least 12 hours). After lyophilization, the sample vials were removed from the lyophilizer’s tray dryer, then crimp sealed. The sample vials were labeled and stored at -20 °C. The resulting vial comprised 2 mg active LS301 and about 100 mg of HSA or MSA. LS301-HSA or LS301-MSA lyophilized product was reconstituted by injecting 10 mL of saline into vials to form a 0.2 mg/mL injection solution. [0145] LS301-HSA formulation was evaluated in multiple cancer models with an injection dose of 0.35 mg/kg. NIR fluorescent images were monitored over time to confirm tumor accumulation. Without wishing to be bound by theory, formulating LS301 improved uptake 42 KC0004-401-PC 019279/WO into cancer cells, which use albumin for energy and express cell surface receptors, such as secreted protein acidic and rich in cysteine (SPARC), to internalize the LS301-albumin complex. Example 16 - HSA/MSA Formulation With Metal Ions [0146] Five mM of metal chloride (Na, K, Ca, Mg, Mn, Al) solutions were prepared. To the LS301-HSA or LS301-MSA lyophilized product, 10 mL of 5 mM metal chloride solution were added to reconstitute the solution, which was then incubated at room temperature for at least 10 minutes before administration. [0147] In vitro cell internalization studies were performed on a 4T1/luc cancer cell lines with 1 µM LS301-MSA formulation comprising 5 mM metal ions. Fluorescent confocal images were taken over time to investigate the internalization pattern difference between different metal ions. Adding divalent ions enhanced cell uptake of LS301-MSA formulation, whereas trivalent ions inhibited the cell uptake of LS301-MSA formulation. [0148] In vivo NIR fluorescent imaging showed biodistribution of the formulations in 4T1luc tumor mice. Like the in vitro study, LS301-MSA with divalent metal ions enhanced tumor accumulation in a shorter time and maintained good tumor retention. [0149] A colorectal cancer mice model was dosed via oral gavage. Fluorescence imaging and histology analysis both proved LS301-MSA targeted cancer cells through the GI system. Example 17 - Pluronic Gel Formulation For Topical Application [0150] To formulate LS301 in a sol-gel mixture for topical administration, Pluronic F-127 (1 g; Sigma Aldrich, St. Louis, MO) was dissolved in phosphate-buffered saline (PBS) (10 mL; PBS) to obtain a 10% (w/v) stock solution. A solution of LS301 (0.5 mL; 60 μM) was dissolved in 10% w/v aqueous Pluronic solution to form the sol-gel system, which was stored on ice before administration to maintain its liquid state. [0151] To evaluate the Pluronic gel-LS301 formulation’s performance, a topical approach of the formulation in-vivo study was conducted using a colon cancer HT29 mice model.10 µL of the Pluronic gel-LS301 formulation was applied to the skin where the tumor was located. In-vivo NIR fluorescent images were taken at different time points post-application. Example 18 - Thermal Sensitive Hydrogel Formulation For Topical Application [0152] PLGA-b-PEG-b-PLGA triblock copolymers (1500:1000:1500, transition temperature ~30 °C, 250 mg) dissolved in 1 mL of cold water (5 °C) were mixed with 44 μg of LS301 in 1 mL tert-butanol at 60 °C and lyophilized for 24 hours. The lyophilized cake was then 43 KC0004-401-PC 019279/WO rehydrated with 1 mL of cold DI water at 4 °C and gently stirred for at least 6 hours in the cold room at 300 rpm. The rehydrated solution was passed through a 0.2-μm regenerated cellulose filter to remove unincorporated precursors. Example 19 - Liposomal Formulation of LS301 [0153] General preparation used DPPC (1,2-dipalmitoyl-sn-glycero-3-phosphocholine), DSPE-PEG (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-5000 amine) and cholesterol (phosphocholine 67.5%, PEGylated phosphocholine 2.5%, cholesterol 30%) for the liposome. Lipid stock solutions were prepared by dissolving each lipid in chloroform in different glass vials. Aliquots from the stock solutions were mixed in a different glass vial to give 2 mL of solution with 30 mM lipid concentration. LS301 (0.005 mmol%, 2.205 mg) was dissolved in 20 µL dimethyl sulfoxide (DMSO) and added to the 2 mL lipid mixture. The solvent from the lipid mixture was evaporated off with a nitrogen stream to obtain a lipid film. The lipid film was further dried under vacuum at -100 kPa for 2 hours. The lipid film was then hydrated with 2 mL 120 mM ammonium sulfate. The lipid suspension thus formed was shaken at 60 °C for 30 minutes, followed by freeze-though cycles. [0154] This suspension was extruded through a 50-nm polycarbonate membrane to form liposomes, purified through size exclusion chromatography to remove any unbound LS301. LS301 containing liposomes were prepared by extrusion in total lipid concentration of 30 mM. Size analysis of the liposomal-LS301 showed that the particle is around 30 nm. [0155] Components of the liposome can and will vary, depending on the application. Different phosphocholine class lipids, PEGylated derivatives of phosphatidylcholine class lipids and cholesterol was used in different ratios to form the main core of the liposome. The phosphocholine class lipids that were used comprised: 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC); 1,2-didodecanoyl-sn-glycero-3-phosphocholine (DLPC); 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC); 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC); 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC); 1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE); 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE); 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE); 1,2-dimyristoyl-sn-glycero-3-phosphate (DMPA); 44 KC0004-401-PC 019279/WO 1,2-dipalmitoyl-sn-glycero-3-phosphatidic acid (DPPA); and 1,2-dioleoyl-sn-glycero-3-phosphate. [0156] The PEGylated phosphocholine class lipids used comprised: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)- 5000 [DSPE- PEG (5000) Amine]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)- 2000 [DSPE- PEG (2000) Amine]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)- 5000 [DSPE- PEG (5000) carboxylic acid]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)- 2000 [DSPE- PEG (2000) carboxylic acid]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[succinyl(polyethylene glycol)- 5000 [DSPE- PEG (5000) succinyl]; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[succinyl(polyethylene glycol)- 2000 [DSPE-PEG (2000) succinyl]; and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[maleimide(polyethylene glycol)-5000 [DSPE-PEG (5000) maleimide]. [0157] In vivo performance of liposomal-LS301 formulation was investigated in a PyMT BO1 tumor-bearing C57/B6 mice. Ex-vivo fluorescence imaging of biodistribution showed excellent tumor uptake of LS301. [0158] The preceding description is given for clearness of understanding only. No unnecessary limitations should be understood from it, as modifications within the disclosure scope may be apparent to those having ordinary skill in the art. Throughout the specification, where compositions are described as including components or materials, it is contemplated that the compositions can also consist essentially of, or consist of, any combination of the recited components or materials, unless described otherwise. Likewise, where methods are described as including steps, it is contemplated that the methods can also consist essentially of, or consist of, any combination of the recited steps, unless described otherwise. The disclosure illustratively disclosed herein suitably may be practiced in the absence of any element or step which is not specifically disclosed herein. [0159] The practice of a method disclosed herein, and individual steps thereof, can be performed manually and/or with the aid of or automation provided by electronic equipment. Although processes have been described concerning embodiments, a person of ordinary skill 45 KC0004-401-PC 019279/WO in the art will readily appreciate that other ways of performing the acts for the methods may be used. For example, the order of various of the steps may be changed without departing from the scope or spirit of the method unless described otherwise. Also, some of the individual steps can be combined, omitted, or further subdivided into additional steps. [0160] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. All combinations of the embodiments of the chemical groups represented by the variables contained within the generic chemical formulae described herein are specifically embraced by the present invention just as if each combination was individually explicitly recited, to the extent that such combinations embrace stable compounds (i.e., compounds that can be isolated, characterized and tested for biological activity). Also, all subcombinations of the chemical groups listed in the embodiments describing such variables, as well as all subcombinations of uses and medical indications described herein, are specifically embraced by the present invention just as if each subcombination of chemical groups and subcombination of uses and medical indications was individually and explicitly recited herein. [0161] All patents, publications, and references cited herein are fully incorporated by reference. In case of conflict between the present disclosure and incorporated patents, publications, and references, the present disclosure should control. 46

Claims

KC0004-401-PC 019279/WO CLAIMS What Is Claimed Is: 1. A pharmaceutical composition comprising an effective amount of cypate-Cyclo(Cys- Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)- Tyr-OH (LS838) or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration; a divalent metal ion; and a pharmaceutically acceptable carrier. 2. The pharmaceutical composition of claim 1, wherein the divalent metal ion is chosen from Ca2+, Mg2+, and Mn2+. 3. The pharmaceutical composition of claim 2, wherein the divalent metal ion is Ca2+. 4. The pharmaceutical composition of any preceding claim, wherein the divalent metal ion is present at a concentration between 1 mM and 10 mM. 5. The pharmaceutical composition of claim 4, wherein the divalent metal ion is present at a concentration of 5 mM. 6. The pharmaceutical composition of any preceding claim, wherein the cypate is . 7. The pharmaceuti
Figure imgf000048_0001
, herein at least one of the Cys amino acid residues is D-Cys. 8. The pharmaceutical composition to claim 1, wherein the compound is LS301 comprising the structural formula 47 KC0004-401-PC 019279/WO 1
Figure imgf000049_0001
comprising the structural formula
Figure imgf000049_0002
comprising the structural formula 48 KC0004-401-PC 019279/WO 11
Figure imgf000050_0001
comprising the structural formula 1
Figure imgf000050_0002
13. The pharmaceutical composition of claim 12, wherein the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin. 49 KC0004-401-PC 019279/WO 14. The pharmaceutical composition of any preceding claim, wherein the pharmaceutically acceptable carrier comprises phosphate-buffered saline. 15. The pharmaceutical composition of any preceding claim formulated for intravenous administration. 16. The pharmaceutical composition of any one of claims 1–14 formulated for topical administration. 17. The pharmaceutical composition of any preceding claim, wherein the effective amount is 0.2 µmol/kg. 18. The pharmaceutical composition of any one of claims 1–16, wherein the effective amount is 0.4 µmol/kg. 19. The pharmaceutical composition of any one of claims 1–16, wherein the effective amount is 0.6 µmol/kg. 20. A lyophilized product comprising a dye-conjugate chosen from cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Lys-OH (LS301), cypate-Cyclo(Cys-Gly-Arg-Asp-Ser-Pro-Cys)-Tyr-OH (LS838) or pharmaceutically acceptable salts thereof, wherein each amino acid residue is independently in a D or L configuration, and albumin, wherein the dye-conjugate and the albumin are in a 1:50 (w/w) ratio. 21. The lyophilized product of claim 20, wherein the albumin is chosen from human serum albumin, mouse serum albumin, and bovine serum albumin 22. The lyophilized product of claim 20 or 21, formed by a method comprising suspending the dye-conjugate in 1% albumin solution at a ratio of 1 gram of dye- conjugate per 10 liters of the albumin solution; mixing the suspension; filtering the mixed suspension; and lyophilizing the filtered suspension to form the lyophilized product. 23. The lyophilized product of claim 22, wherein the suspension is mixed on a shaker of at least about 30 minutes. 24. The lyophilized product of claim 22, the dye-conjugate and albumin concentration in the suspension are measured via absorbance. 25. A vial comprising about 102 mg of the lyophilized product of any one of claims 20– 24. 50 KC0004-401-PC 019279/WO 26. An injectable solution comprising about 102 mg of the lyophilized product of any one of claims 20 to 24 and about 10 mL of phosphate-buffered saline. 27. The injectable solution of claim 26, comprising about 0.2 mg/mL dye-conjugate. 28. A method of preparing an injectable solution, the method comprising mixing about 10 mL of phosphate-buffered saline into a vial comprising about 102 mg of the lyophilized product of any one of claims 20–24 to form an injectable solution comprising about 0.2 mg/mL dye-conjugate. 29. A method for identifying compromised fibroblasts, comprising administering an effective amount of the pharmaceutical composition of any one of claims 1–19 or a lyophilized product of any one of claims 20–24 or an injectable solution of claim 26 or 27 to a subject in need thereof. 30. The method of claim 29, wherein the compromised fibroblasts are proximal to dormant cancer cells. 31. The method of claim 30, wherein the dormant cancer cells are chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (primary), cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, cervical cancer, childhood cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, desmoplastic small round cell tumor, endometrial cancer, ependymoma, esophageal cancer, Ewing’s sarcoma in the Ewing family of tumors, extracranial germ cell tumor (childhood), extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancers (intraocular melanoma, retinoblastoma), gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumors (childhood extracranial, extragonadal, ovarian), gestational trophoblastic tumor, gliomas (adult, childhood brain stem, childhood cerebral astrocytoma, childhood visual pathway and hypothalamic), gastric carcinoid, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, hypothalamic and 51 KC0004-401-PC 019279/WO visual pathway glioma (childhood), intraocular melanoma, islet cell carcinoma, Kaposi sarcoma, kidney cancer (renal cell cancer), laryngeal cancer, leukemias (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, hairy cell), lip and oral cavity cancer, liver cancer (primary), lung cancers (non-small cell, small cell), lymphomas (AIDS- related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, primary central nervous system), macroglobulinemia (Waldenström), malignant fibrous histiocytoma of bone/osteosarcoma, medulloblastoma (childhood), melanoma, intraocular melanoma, Merkel cell carcinoma, mesotheliomas (adult malignant, childhood), metastatic squamous neck cancer with occult primary, mouth cancer, multiple endocrine neoplasia syndrome (childhood), multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, myelogenous leukemia (chronic), myeloid leukemias (adult acute, childhood acute), multiple myeloma, myeloproliferative disorders (chronic), nasal cavity and paranasal sinus cancer, nasopharyngeal carcinoma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma/malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer (surface epithelial-stromal tumor), ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, pancreatic cancer (islet cell), paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pineal astrocytoma, pineal germinoma, pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood), pituitary adenoma, plasma cell neoplasia, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell carcinoma (kidney cancer), renal pelvis and ureter transitional cell cancer, retinoblastoma, rhabdomyosarcoma (childhood), salivary gland cancer, sarcoma (Ewing family of tumors, Kaposi, soft tissue, uterine), Sézary syndrome, skin cancers (nonmelanoma, melanoma), skin carcinoma (Merkel cell), small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer with occult primary (metastatic), stomach cancer, supratentorial primitive neuroectodermal tumor (childhood), T- Cell lymphoma (cutaneous), testicular cancer, throat cancer, thymoma (childhood), thymoma and thymic carcinoma, thyroid cancer, thyroid cancer (childhood), transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor (gestational), unknown primary site (adult, childhood), ureter and renal pelvis transitional cell cancer, urethral cancer, uterine cancer (endometrial), uterine sarcoma, vaginal cancer, visual pathway and hypothalamic glioma (childhood), vulvar cancer, Waldenström macroglobulinemia, and Wilms tumor (childhood). 52 KC0004-401-PC 019279/WO 32. The method of claim 30, wherein the dormant cancer cells are pancreatic cancer. 33. The method of claim 30, wherein the pharmaceutical composition is administered intravenously. 34. The method of claim 30, wherein the pharmaceutical composition is administered topically. 35. The method of claim 30, wherein the topical administration is to the subject’s colon. 36. A method of binding phosphorylated annexin A2 (pANXA2) protein in a biological sample comprising contacting the biological sample with a pharmaceutical composition of any one of claims 1–19 or a lyophilized product of any one of claims 20–24 or an injectable solution of claim 26 or 27. 37. The method of claim 36, wherein the binding is selective over annexin A1 (ANXA1), non-activated ANXA2, and annexin A3 (ANXA3). 38. The method of claim 36, wherein tumor margins of cancer are defined. 39. The method of claim 36, wherein the accuracy of cancer resection is improved during surgery. 40. The method of claim 36, wherein cancer is treated simultaneously from the periphery and interior core of a tumor. 41. The method of any one of claims 36–40, wherein the cancer is chosen from acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas, breast cancer, bronchial adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (primary), cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, cervical cancer, childhood cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, desmoplastic small round cell tumor, endometrial cancer, ependymoma, esophageal cancer, Ewing’s sarcoma in the Ewing family of tumors, extracranial germ cell tumor (childhood), extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancers (intraocular melanoma, retinoblastoma), gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell 53 KC0004-401-PC 019279/WO tumors (childhood extracranial, extragonadal, ovarian), gestational trophoblastic tumor, gliomas (adult, childhood brain stem, childhood cerebral astrocytoma, childhood visual pathway and hypothalamic), gastric carcinoid, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, hypothalamic and visual pathway glioma (childhood), intraocular melanoma, islet cell carcinoma, Kaposi sarcoma, kidney cancer (renal cell cancer), laryngeal cancer, leukemias (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, hairy cell), lip and oral cavity cancer, liver cancer (primary), lung cancers (non-small cell, small cell), lymphomas (AIDS- related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, primary central nervous system), macroglobulinemia (Waldenström), malignant fibrous histiocytoma of bone/osteosarcoma, medulloblastoma (childhood), melanoma, intraocular melanoma, Merkel cell carcinoma, mesotheliomas (adult malignant, childhood), metastatic squamous neck cancer with occult primary, mouth cancer, multiple endocrine neoplasia syndrome (childhood), multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, myelogenous leukemia (chronic), myeloid leukemias (adult acute, childhood acute), multiple myeloma, myeloproliferative disorders (chronic), nasal cavity and paranasal sinus cancer, nasopharyngeal carcinoma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma/malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer (surface epithelial-stromal tumor), ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, pancreatic cancer (islet cell), paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pineal astrocytoma, pineal germinoma, pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood), pituitary adenoma, plasma cell neoplasia, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell carcinoma (kidney cancer), renal pelvis and ureter transitional cell cancer, retinoblastoma, rhabdomyosarcoma (childhood), salivary gland cancer, sarcoma (Ewing family of tumors, Kaposi, soft tissue, uterine), Sézary syndrome, skin cancers (nonmelanoma, melanoma), skin carcinoma (Merkel cell), small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer with occult primary (metastatic), stomach cancer, supratentorial primitive neuroectodermal tumor (childhood), T- Cell lymphoma (cutaneous), testicular cancer, throat cancer, thymoma (childhood), thymoma and thymic carcinoma, thyroid cancer, thyroid cancer (childhood), transitional cell cancer of 54 KC0004-401-PC 019279/WO the renal pelvis and ureter, trophoblastic tumor (gestational), unknown primary site (adult, childhood), ureter and renal pelvis transitional cell cancer, urethral cancer, uterine cancer (endometrial), uterine sarcoma, vaginal cancer, visual pathway and hypothalamic glioma (childhood), vulvar cancer, Waldenström macroglobulinemia, and Wilms tumor (childhood). 55
PCT/US2020/064506 2019-12-13 2020-12-11 Near infrared fluorescent dyes, formulations and related methods WO2021119423A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP20897916.1A EP4072598A4 (en) 2019-12-13 2020-12-11 Near infrared fluorescent dyes, formulations and related methods

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201962947974P 2019-12-13 2019-12-13
US62/947,974 2019-12-13
US202062976702P 2020-02-14 2020-02-14
US62/976,702 2020-02-14

Publications (1)

Publication Number Publication Date
WO2021119423A1 true WO2021119423A1 (en) 2021-06-17

Family

ID=76316518

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/064506 WO2021119423A1 (en) 2019-12-13 2020-12-11 Near infrared fluorescent dyes, formulations and related methods

Country Status (3)

Country Link
US (2) US11712482B2 (en)
EP (1) EP4072598A4 (en)
WO (1) WO2021119423A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11765340B2 (en) 2012-01-23 2023-09-19 Washington University Goggle imaging systems and methods

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140023700A1 (en) * 2011-02-22 2014-01-23 Leo Pharma A/S Cyclic peptide and conjugate thereof for binding to keratinocytes
US20180289842A1 (en) * 2015-05-06 2018-10-11 Washington University Compounds having rd targeting motifs and methods of use thereof

Family Cites Families (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3706266A1 (en) 1986-02-28 1987-09-03 Olympus Optical Co RECORD CARRIER FOR OPTICAL INFORMATION
US5268486A (en) 1986-04-18 1993-12-07 Carnegie-Mellon Unversity Method for labeling and detecting materials employing arylsulfonate cyanine dyes
US5972890A (en) 1988-05-02 1999-10-26 New England Deaconess Hospital Corporation Synthetic peptides for arterial imaging
US5107063A (en) 1990-10-31 1992-04-21 E. I. Du Pont De Nemours And Company Aqueous soluble infrared antihalation dyes
IT1251083B (en) 1991-07-19 1995-05-04 Ivano Delprato SILVER HALIDE PHOTOGRAPHIC ELEMENTS
JP2787742B2 (en) 1992-03-30 1998-08-20 富士写真フイルム株式会社 Silver halide photographic material
US5254852A (en) 1992-05-28 1993-10-19 Night Vision General Partnership Helmet-mounted night vision system and secondary imager
JPH06145539A (en) 1992-11-04 1994-05-24 Fuji Photo Film Co Ltd Cyanine compound
EP0620703B1 (en) 1993-04-12 1997-12-29 Ibiden Co, Ltd. Resin compositions and printed circuit boards using the same
JPH0772446A (en) 1993-09-01 1995-03-17 Sharp Corp Display system
US5453505A (en) 1994-06-30 1995-09-26 Biometric Imaging, Inc. N-heteroaromatic ion and iminium ion substituted cyanine dyes for use as fluorescence labels
US5518934A (en) 1994-07-21 1996-05-21 Trustees Of Princeton University Method of fabricating multiwavelength infrared focal plane array detector
DE4445065A1 (en) 1994-12-07 1996-06-13 Diagnostikforschung Inst Methods for in-vivo diagnostics using NIR radiation
US6032070A (en) 1995-06-07 2000-02-29 University Of Arkansas Method and apparatus for detecting electro-magnetic reflection from biological tissue
US5959705A (en) 1996-03-15 1999-09-28 Osd Envizion, Inc. Welding lens with integrated display, switching mechanism and method
US20020028474A1 (en) 1996-09-19 2002-03-07 Daiichi Pure Chemical Co., Ltd. Composition for immunohistochemical staining
DE19649971A1 (en) 1996-11-19 1998-05-28 Diagnostikforschung Inst Optical diagnostics for the diagnosis of neurodegenerative diseases using near-infrared radiation (NIR radiation)
US6027709A (en) 1997-01-10 2000-02-22 Li-Cor Inc. Fluorescent cyanine dyes
WO1998048845A1 (en) 1997-04-29 1998-11-05 Nycomed Imaging As Method of demarcating tissue
JP2001526650A (en) 1997-04-29 2001-12-18 ニユコメド・イメージング・アクシエセルカペト Optical contrast agent
US5955224A (en) 1997-07-03 1999-09-21 E. I. Du Pont De Nemours And Company Thermally imageable monochrome digital proofing product with improved near IR-absorbing dye(s)
US6180084B1 (en) 1998-08-25 2001-01-30 The Burnham Institute NGR receptor and methods of identifying tumor homing molecules that home to angiogenic vasculature using same
US6232287B1 (en) 1998-03-13 2001-05-15 The Burnham Institute Molecules that home to various selected organs or tissues
US6472367B1 (en) 1998-05-05 2002-10-29 Adherex Technologies, Inc. Compounds and methods for modulating OB-cadherin mediated cell adhesion
US7547721B1 (en) 1998-09-18 2009-06-16 Bayer Schering Pharma Ag Near infrared fluorescent contrast agent and fluorescence imaging
JP2000095758A (en) 1998-09-18 2000-04-04 Schering Ag Near-infrared, fluorescent contrast medium, and its production
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
US6217848B1 (en) 1999-05-20 2001-04-17 Mallinckrodt Inc. Cyanine and indocyanine dye bioconjugates for biomedical applications
WO2001005161A1 (en) 1999-07-13 2001-01-18 Surgivision Ltd. Stereoscopic video observation and image magnification system
EA200200220A1 (en) 1999-07-29 2002-08-29 Эпикс Медикал, Инк. AIMING MULTIMERAL VISUALIZING AGENTS WITH A MULTI-CUSCLE BINDING MECHANISM
US6944493B2 (en) 1999-09-10 2005-09-13 Akora, Inc. Indocyanine green (ICG) compositions and related methods of use
BR9917587A (en) 1999-12-15 2002-08-06 Schering Ag Fluorescent contrast agent near infrared and fluorescence imaging
US20110213625A1 (en) 1999-12-18 2011-09-01 Raymond Anthony Joao Apparatus and method for processing and/or for providing healthcare information and/or helathcare-related information
US20020041898A1 (en) 2000-01-05 2002-04-11 Unger Evan C. Novel targeted delivery systems for bioactive agents
US6554444B2 (en) 2000-03-13 2003-04-29 Kansai Technology Licensing Organization Co., Ltd. Gazing point illuminating device
JP2001299676A (en) 2000-04-25 2001-10-30 Fuji Photo Film Co Ltd Method and system for detecting sentinel lymph node
US7345277B2 (en) 2000-08-09 2008-03-18 Evan Zhang Image intensifier and LWIR fusion/combination system
US6487428B1 (en) 2000-08-31 2002-11-26 Trustees Of The University Of Pennsylvania Extravasation detection apparatus and method based on optical sensing
CA2424454C (en) 2000-09-19 2010-07-20 Li-Cor, Inc. Cyanine dyes
EP1221465A1 (en) 2001-01-03 2002-07-10 Innosense S.r.l. Symmetric, monofunctionalised polymethine dyes labelling reagents
US6585660B2 (en) 2001-05-18 2003-07-01 Jomed Inc. Signal conditioning device for interfacing intravascular sensors having varying operational characteristics to a physiology monitor
US20030105299A1 (en) 2001-10-17 2003-06-05 Mallinckrodt Inc. Carbocyanine dyes for tandem, photodiagnostic and therapeutic applications
JP2003261464A (en) 2002-03-07 2003-09-16 Fuji Photo Film Co Ltd Near infrared fluorescent contrast agent and fluorescent contrast radiography
US7134994B2 (en) 2002-05-20 2006-11-14 Volcano Corporation Multipurpose host system for invasive cardiovascular diagnostic measurement acquisition and display
ES2311736T3 (en) 2003-01-24 2009-02-16 Bayer Schering Pharma Aktiengesellschaft HYDROPHILIC DYES OF CYANINE REAGENT TIOL AND CONJUGATES OF THE SAME WITH BIOMOLECULES FOR FLUORESCENCE DIAGNOSIS.
US7172907B2 (en) 2003-03-21 2007-02-06 Ge Healthcare Bio-Sciences Corp. Cyanine dye labelling reagents with meso-substitution
US20040215081A1 (en) 2003-04-23 2004-10-28 Crane Robert L. Method for detection and display of extravasation and infiltration of fluids and substances in subdermal or intradermal tissue
WO2005000218A2 (en) 2003-05-31 2005-01-06 Washington University Macrocyclic cyanine and indocyanine bioconjugates provide improved biomedical applications
US8675059B2 (en) 2010-07-29 2014-03-18 Careview Communications, Inc. System and method for using a video monitoring system to prevent and manage decubitus ulcers in patients
US20060173351A1 (en) 2005-01-03 2006-08-03 Ronald Marcotte System and method for inserting a needle into a blood vessel
US20060173360A1 (en) 2005-01-07 2006-08-03 Kalafut John F Method for detection and display of extravasation and infiltration of fluids and substances in subdermal or intradermal tissue
US8053415B2 (en) 2005-01-21 2011-11-08 Washington University In St. Louis Compounds having RD targeting motifs
US8227621B2 (en) 2005-06-30 2012-07-24 Li-Cor, Inc. Cyanine dyes and methods of use
WO2007014293A1 (en) 2005-07-25 2007-02-01 The Regents Of The University Of California Digital imaging system and method to produce mosaic images
US7211778B1 (en) 2005-10-07 2007-05-01 Itt Manufacturing Enterprises, Inc. Night vision goggle with separate camera and user output paths
US7826890B1 (en) 2005-12-06 2010-11-02 Wintec, Llc Optical detection of intravenous infiltration
US8199189B2 (en) 2006-04-07 2012-06-12 Novarix Ltd. Vein navigation device
US20100040547A1 (en) 2006-08-03 2010-02-18 Frangioni John V Dyes and precursors and conjugates thereof
US9229230B2 (en) 2007-02-28 2016-01-05 Science Applications International Corporation System and method for video image registration and/or providing supplemental data in a heads up display
CN102017147B (en) 2007-04-18 2014-01-29 因维萨热技术公司 Materials, systems and methods for optoelectronic devices
WO2009023813A1 (en) 2007-08-15 2009-02-19 Applied Soil Water Technologies Llc Metals recovery from mining heap leach ore
US20090074672A1 (en) 2007-09-17 2009-03-19 Sri International Tumor Boundary Imaging
US20090093761A1 (en) 2007-10-05 2009-04-09 Sliwa John W Medical-procedure assistance device and method with improved optical contrast, and new practitioner-safety, device-fixation, electrode and magnetic treatment and lumen-dilation capabilities
US8849380B2 (en) 2007-11-26 2014-09-30 Canfield Scientific Inc. Multi-spectral tissue imaging
WO2009089543A2 (en) 2008-01-10 2009-07-16 The Ohio State University Research Foundation Fluorescence detection system
US20100113940A1 (en) 2008-01-10 2010-05-06 The Ohio State University Research Foundation Wound goggles
US20090214436A1 (en) 2008-02-18 2009-08-27 Washington University Dichromic fluorescent compounds
US20090242797A1 (en) 2008-03-31 2009-10-01 General Electric Company System and method for multi-mode optical imaging
US8840249B2 (en) 2008-10-31 2014-09-23 Christie Digital Systems, Inc. Method, system and apparatus for projecting visible and non-visible images
ES2684697T3 (en) 2009-02-06 2018-10-04 Beth Israel Deaconess Medical Center, Inc. Agents for obtaining balanced load images
US20100240988A1 (en) 2009-03-19 2010-09-23 Kenneth Varga Computer-aided system for 360 degree heads up display of safety/mission critical data
CN102459469A (en) 2009-04-17 2012-05-16 利康股份有限公司 Fluorescent imaging with substituted cyanine dyes
KR101190265B1 (en) 2009-06-30 2012-10-12 고려대학교 산학협력단 Head mouted operating magnifying apparatus
US8498694B2 (en) 2009-07-13 2013-07-30 Entrotech, Inc. Subcutaneous access device and related methods
US8586924B2 (en) 2010-09-13 2013-11-19 Lawrence Livermore National Security, Llc Enhancement of the visibility of objects located below the surface of a scattering medium
US9105548B2 (en) 2011-06-22 2015-08-11 California Institute Of Technology Sparsely-bonded CMOS hybrid imager
CA2844135A1 (en) 2011-08-01 2013-02-07 Infrared Imaging Systems, Inc. Disposable light source for enhanced visualization of subcutaneous structures
WO2013028963A1 (en) 2011-08-24 2013-02-28 Volcano Corporation Medical communication hub and associated methods
US20130129835A1 (en) 2011-09-06 2013-05-23 Allergan, Inc. Crosslinked hyaluronic acid-collagen gels for improving tissue graft viability and soft tissue augmentation
EP2806781B1 (en) 2012-01-23 2018-03-21 Washington University Goggle imaging systems and methods
US20130338512A1 (en) 2012-03-12 2013-12-19 Ivwatch, Llc System and Method for Mitigating the Effects of Tissue Blood Volume Changes to Aid in Diagnosing Infiltration or Extravasation in Animalia Tissue
US20140046291A1 (en) 2012-04-26 2014-02-13 Evena Medical, Inc. Vein imaging systems and methods
US20140039309A1 (en) 2012-04-26 2014-02-06 Evena Medical, Inc. Vein imaging systems and methods
US20140200438A1 (en) 2012-12-21 2014-07-17 Volcano Corporation Intraluminal imaging system
JP6479678B2 (en) 2012-12-21 2019-03-06 ボルケーノ コーポレイション Display control for multi-sensor medical devices
US20140180316A1 (en) 2012-12-21 2014-06-26 Volcano Corporation Imaging and removing biological material
EP2934305B1 (en) 2012-12-21 2018-02-21 Volcano Corporation System for multi-site intravascular measurement
US20140194704A1 (en) 2012-12-21 2014-07-10 Volcano Corporation Intraluminal imaging system
US9486143B2 (en) 2012-12-21 2016-11-08 Volcano Corporation Intravascular forward imaging device
WO2014100207A1 (en) 2012-12-21 2014-06-26 Paul Hoseit Imaging guidewire with photoactivation capabilities
US10058284B2 (en) 2012-12-21 2018-08-28 Volcano Corporation Simultaneous imaging, monitoring, and therapy
US20140275950A1 (en) 2013-03-13 2014-09-18 Volcano Corporation Imaging guidewire with pressure sensing
US20140276110A1 (en) 2013-03-14 2014-09-18 Volcano Corporation Imaging guidewire system with flow visualization
WO2014151841A1 (en) 2013-03-15 2014-09-25 Volcano Corporation Universal patient interface module and associated devices, systems, and methods
WO2015066296A1 (en) 2013-10-31 2015-05-07 Beth Israel Deaconess Medical Center Near-infrared fluorescent nerve contrast agents and methods of use thereof
US10209242B2 (en) 2015-05-21 2019-02-19 Emit Imaging, Inc. Fluorescence histo-tomography (FHT) systems and methods

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140023700A1 (en) * 2011-02-22 2014-01-23 Leo Pharma A/S Cyclic peptide and conjugate thereof for binding to keratinocytes
US20180289842A1 (en) * 2015-05-06 2018-10-11 Washington University Compounds having rd targeting motifs and methods of use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP4072598A4 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11765340B2 (en) 2012-01-23 2023-09-19 Washington University Goggle imaging systems and methods

Also Published As

Publication number Publication date
EP4072598A4 (en) 2024-02-21
US20230321283A1 (en) 2023-10-12
US20210177993A1 (en) 2021-06-17
US11712482B2 (en) 2023-08-01
EP4072598A1 (en) 2022-10-19

Similar Documents

Publication Publication Date Title
ES2500340T3 (en) Annexins, derivatives thereof, and annexin-Cys variants, as well as diagnostic and therapeutic uses thereof
ES2389828T3 (en) Compositions comprising SPARC polypeptides
ES2700198T3 (en) PH-sensitive liposome nucleolin binding
ES2529660T3 (en) Treatment methods in which albumin binding proteins are used as targets
US8828925B2 (en) Etoposide and doxorubicin conjugates for drug delivery
Shen et al. Selective imaging of solid tumours via the calcium-dependent high-affinity binding of a cyclic octapeptide to phosphorylated Annexin A2
ES2687076T3 (en) SPARC and methods of using it
US8394758B2 (en) Tumor-targeting peptides and uses thereof
US9163063B2 (en) Peptides and nanoparticles for therapeutic and diagnostic applications
WO2017063542A1 (en) Stabilized a7r polypeptides, and use thereof in constructing tumor targeted therapeutic drug delivery system
US20230321283A1 (en) Near infrared fluorescent dyes, formulations and related methods
CN110114367B (en) VAP polypeptide and application thereof in preparation of targeted tumor diagnosis and treatment medicines
US20100076175A1 (en) Molecules that selectively home to vasculature of premalignant or malignant lesions of the pancreas and other organs
Shimizu et al. Development of tissue factor-targeted liposomes for effective drug delivery to stroma-rich tumors
Guo et al. Multicomponent thermosensitive lipid complexes enhance desmoplastic tumor therapy through boosting anti-angiogenesis and synergistic strategy
CN103038248B (en) Rhamm binding peptide
Santos-Pereira et al. Lactoferrin perturbs intracellular trafficking, disrupts cholesterol-rich lipid rafts and inhibits glycolysis of highly metastatic cancer cells harbouring plasmalemmal V-ATPase
ES2765037T3 (en) Peptides derived from CD44V6 to treat metastatic cancers
US11890352B2 (en) Plectin-targeted liposomes/PARP inhibitor in the treatment of cancer
US20120258038A1 (en) Uses of apoptotic cell-targeting peptides, label substances and liposomes containing a therapeutic agent for preventing, treating or therapeutically diagnosing apoptosis-related diseases
TWI630910B (en) Peptide-conjugated nanoparticles for targeting, imaging, and treatment of prostate cancer
Vári et al. SREKA-targeted liposomes for highly metastatic breast cancer therapy
IT202000013042A1 (en) LIPOSOMAL SYSTEM WITH KILLER TNF-APOPTOSIS INDUCED LIGAND (KILLERTRAIL), PRO-APOPTOTIC-DIRECTING.
JP7370598B2 (en) Peptides and their uses
JP2019509298A (en) Targeting tumor cells with chemotherapeutic agents conjugated to anti-matriptase antibodies via a cleavable linking moiety in vivo

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: 20897916

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020897916

Country of ref document: EP

Effective date: 20220713