US20130116404A1 - Targeted non-invasive imaging probes of egfr expressing cells - Google Patents

Targeted non-invasive imaging probes of egfr expressing cells Download PDF

Info

Publication number
US20130116404A1
US20130116404A1 US13/671,957 US201213671957A US2013116404A1 US 20130116404 A1 US20130116404 A1 US 20130116404A1 US 201213671957 A US201213671957 A US 201213671957A US 2013116404 A1 US2013116404 A1 US 2013116404A1
Authority
US
United States
Prior art keywords
probe
egfr
cells
moiety
linker
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/671,957
Inventor
James Basilion
Richard Agnes
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Case Western Reserve University
Original Assignee
Case Western Reserve 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 Case Western Reserve University filed Critical Case Western Reserve University
Priority to US13/671,957 priority Critical patent/US20130116404A1/en
Assigned to CASE WESTERN RESERVE UNIVERSITY reassignment CASE WESTERN RESERVE UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AGNES, RICHARD, BASILION, JAMES P.
Publication of US20130116404A1 publication Critical patent/US20130116404A1/en
Priority to US16/396,265 priority patent/US20190315805A1/en
Priority to US17/224,505 priority patent/US11708393B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/50Cyclic peptides containing at least one abnormal peptide link
    • C07K7/54Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring
    • C07K7/56Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring the cyclisation not occurring through 2,4-diamino-butanoic acid

Definitions

  • Non-invasive optical imaging technologies can be used for diagnosis and monitoring therapeutic response.
  • optical instrumentation advances e.g., tomographic imaging
  • NIRF near infrared fluorescence
  • fluorochrome reporter moiety a molecule labeled with near infrared fluorescence (NIRF) or fluorochrome reporter moiety.
  • Glioblastoma multiforme is the most common and most malignant of the glial tumors. In 40-50% of these tumors, mutations resulting to over expression or activation of epidermal growth factors are found.
  • Epidermal growth factors receptors are cell surface receptors that regulate growth and survival as well as adhesion, migration, differentiation and other cellular processes. The ability to differentially image EGFR expression levels might provide non-invasive means to identify tumor biomarker that can aide in the selection of treatment as well as means of targeted drug delivery.
  • the probe can include an EGFR targeting moiety, a reporter moiety, and a hydrophilic linker that links or couples the EGFR targeting moiety to the reporter moiety.
  • the hydrophilic linker can enhance solubility of the probe in an aqueous media as well as binding affinity of the probe to EGFR expressing cells.
  • the EGFR expressing cells can include EGFR expressing cancer cells.
  • the EGFR expressing cancer cells can include glioblastomas as well as solid tumors, such as breast, lung, ovarian, and colon cancers.
  • the probe upon systemic administration to subject is capable of crossing the blood brain barrier, targeting the EGFR expressing cancer cells, and imaging the EGFR expressing cancer cells.
  • the EGFR targeting moiety can include an EGF peptide ligand having an amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the reporter moiety can include a fluorochrome, and the linker comprising a polyethylene glycol linker.
  • the probe can include the following formula:
  • R is a reporter moiety, such as a NIRF reporter moiety or fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3.
  • a reporter moiety such as a NIRF reporter moiety or fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1)
  • fluorochrome e.g., Cy5, Cy5.5, or ZW800-1
  • n is an integer from 1 to 3.
  • the probe can have the following formula:
  • n is an integer from 1 to 3 (e.g., 1).
  • the probe can include the following formula:
  • R is a reporter moiety, such as a NIRF reporter moiety or a fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3.
  • a reporter moiety such as a NIRF reporter moiety or a fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3.
  • fluorochrome e.g., Cy5, Cy5.5, or ZW800-1
  • n is an integer from 1 to 3.
  • the probe can have the following formula:
  • n is an integer from 1 to 3 (e.g., 1).
  • FIG. 1 is a schematic illustration of an imaging probe in accordance with an embodiment of the application.
  • FIG. 2 illustrates a plot showing cell associated fluorescence measured using spectrofluorometry at the indicated times for Gli36 ⁇ 5 cells incubated with 1 ⁇ mol/L of compounds 1 to 4 [Cy5.5-(AEEA) n -EGF pep ].
  • FIGS. 3(A-B) illustrate images showing Gli36 ⁇ 5 (A) and U87-MG (B) cells incubated with 1 ⁇ mol/L compounds for the indicated times. Uptake of the compounds was assessed using epifluorescence microscopy. Images were taken at ⁇ 40 magnification. Scale bar, 50 ⁇ m.
  • FIG. 4 illustrates graphs showing the specificity of the targeting assessed using a scrambled peptide (6) via competition against unlabeled parent peptide compound (5).
  • Ex vivo analysis of brain tissues showed that compound 2 targets the tumor significantly (***, P>0.0001) more efficiently than the scrambled peptide 6.
  • Representative images of the brain tumors are shown above the corresponding bars (left graph).
  • FIG. 5 illustrates images showing the colocalization of EGFR-targeted probe with cells expressing high levels of EGFR. Scale bar, 20 mm
  • FIGS. 6(A-B) illustrate western blots showing (A) showing the relative levels of wild-type EGFR expressed in Gli36 ⁇ 5 and U87-MG glioblastoma cell lysates using an antibody specific to wild-type EGFR (DAKO, cat #M7289); and (B) comparing the mutant EGFRvIII content in Gli36 ⁇ 5 and U87-MG tumor cells lines using an EGFRvIII-specific antibody (Bioss Inc., cat #bs-2558R). ⁇ -Actin was used a loading control. Densitometry analysis of Western blotting by ImageJ comparing the levels of EGFRvIII in Gli36 ⁇ 5 and U87-MG cells show similar content of 1,800 and 2,400 arbitrary units, respectively.
  • FIG. 7 illustrates an image of FMT3-dimensional reconstruction showing the targeting of compound 2 to brain Gli36 ⁇ 5 orthotopic brain tumors. Sham, U87-MG, and Gli36 ⁇ 5 shows 272, 1,608, and 9,271 nmol/L amounts of fluorescence in the matching ROIs.
  • FIGS. 8(A-C) illustrate graphs showing the detected level of the fluorescent agent in (ppmol) administered to live animals (A) or ex vivo brains (B and C) with brain tumors, Gli36 ⁇ 5 and U87, using FMT imaging and Maestro imaging (C).
  • an element means one element or more than one element.
  • sample can refer to a specimen or culture obtained from any source, as well as clinical, research, biological and environmental samples.
  • Biological samples may be obtained from animals (including humans) and encompass cells, fluids, solids, tissues, and organs, and whole organisms.
  • the term “subject” can refer to any animal including, but not limited to, humans and non-human animals (e.g., rodents, arthropods, insects, fish (e.g., zebrafish)), non-human primates, ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, or canines felines, aves, etc.).
  • non-human animals e.g., rodents, arthropods, insects, fish (e.g., zebrafish)
  • non-human primates e.g., ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, or canines felines, aves, etc.
  • non-human animals e.g., rodents, arthropods, insects, fish (e.g., zebrafish)
  • non-human primates e.g., ovines, bovines, ruminants
  • the terms “cancer” or “tumor” refer to any neoplastic growth in a subject, including an initial tumor and any metastases.
  • the cancer can be of the liquid or solid tumor type.
  • Liquid tumors include tumors of hematological origin, including, e.g., myelomas (e.g., multiple myeloma), leukemias (e.g., Waldenstrom's syndrome, chronic lymphocytic leukemia, other leukemias), and lymphomas (e.g., B-cell lymphomas, non-Hodgkin's lymphoma).
  • Solid tumors can originate in organs and include cancers of the lungs, brain, breasts, prostate, ovaries, colon, kidneys and liver.
  • carcinomas such as squamous cell carcinoma, non-small cell carcinoma (e.g., non-small cell lung carcinoma), small cell carcinoma (e.g., small cell lung carcinoma), basal cell carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, adenocarcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, undifferentiated carcinoma, bronchogenic carcinoma, melanoma, renal cell carcinoma, hepatoma-liver cell carcinoma, bile duct carcinoma, cholangiocarcinoma, papillary carcinoma, transitional cell carcinoma, choriocarcinoma, semonoma, embryonal carcinoma, mammary carcinomas, gastrointestinal carcinoma, colonic carcinomas, bladder carcinoma, prostate carcinoma, and squamous
  • polypeptide or “peptide” are used interchangeably and can refer to an oligopeptide, peptide, polypeptide, or protein sequence, or to a fragment, portion, or subunit of any of these, and to naturally occurring or synthetic molecules.
  • polypeptide can also include amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain any type of modified amino acids.
  • the term can also include peptides and polypeptide fragments, motifs and the like, glycosylated polypeptides, and all “mimetic” and “peptidomimetic” polypeptide forms.
  • probe or “imaging probe” can refer to a biological moiety that may be used to detect, image, and/or monitor the presence and/or progression of a cell cycle, cell function/physiology, condition, pathological disorder and/or disease.
  • diagnosisally effective amount can refer to an amount of an imaging composition that is sufficient to enable imaging of at least one cell, tissue, or organism using an imaging modality.
  • targeting moiety can refer to a molecule or molecules that are able to bind to and complex with a biomarker.
  • the term can also refer to a functional group that serves to target or direct an imaging probe to a particular location, cell type, diseased tissue, or association.
  • a “targeting moiety” can be directed against a biomarker.
  • the term “antibody” refers to an immunoglobulin, derivatives thereof which maintain specific binding ability, and proteins having a binding domain which is homologous or largely homologous to an immunoglobulin binding domain. These proteins may be derived from natural sources, or partly or wholly synthetically produced.
  • An antibody may be monoclonal or polyclonal.
  • the antibody may be a member of any immunoglobulin class, including any of the human classes: IgG, IgM, IgA, IgD, and IgE.
  • antibodies used with the methods and compositions described herein are derivatives of the IgG class.
  • antibody fragment refers to any derivative of an antibody which is less than full-length. In exemplary embodiments, the antibody fragment retains at least a significant portion of the full-length antibody's specific binding ability. Examples of antibody fragments include, but are not limited to, Fab, Fab′, F(ab′) 2 , scFv, Fv, dsFv diabody, and Fd fragments.
  • the antibody fragment may be produced by any means. For instance, the antibody fragment may be enzymatically or chemically produced by fragmentation of an intact antibody, it may be recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced.
  • the antibody fragment may optionally be a single chain antibody fragment. Alternatively, the fragment may comprise multiple chains which are linked together, for instance, by disulfide linkages. The fragment may also optionally be a multimolecular complex.
  • a functional antibody fragment will typically comprise at least about 50 amino acids and more typically will comprise at least about 200 amino acids.
  • diabodies refers to dimeric scFvs.
  • the components of diabodies typically have shorter peptide linkers than most scFvs and they show a preference for associating as dimers.
  • epitope refers to a physical structure on a molecule that interacts with a selective component.
  • epitope refers to a desired region on a target molecule that specifically interacts with a selectivity component.
  • Fab refers to an antibody fragment that is essentially equivalent to that obtained by digestion of immunoglobulin (typically IgG) with the enzyme papain.
  • the heavy chain segment of the Fab fragment is the Fd piece.
  • Such fragments may be enzymatically or chemically produced by fragmentation of an intact antibody, recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced.
  • Fab′ refers to an antibody fragment that is essentially equivalent to that obtained by reduction of the disulfide bridge or bridges joining the two heavy chain pieces in the F(ab′) 2 fragment. Such fragments may be enzymatically or chemically produced by fragmentation of an intact antibody, recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced.
  • F(ab′) 2 refers to an antibody fragment that is essentially equivalent to a fragment obtained by digestion of an immunoglobulin (typically IgG) with the enzyme pepsin at pH 4.0-4.5. Such fragments may be enzymatically or chemically produced by fragmentation of an intact antibody, recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced.
  • Fv refers to an antibody fragment that consists of one V H and one V L domain held together by noncovalent interactions.
  • dsFv is used herein to refer to an Fv with an engineered intermolecular disulfide bond to stabilize the V H -V L pair.
  • single-chain Fvs and “scFvs” refers to recombinant antibody fragments consisting of only the variable light chain (V L ) and variable heavy chain (V H ) covalently connected to one another by a polypeptide linker.
  • V L or V H may be the NH 2 -terminal domain.
  • the polypeptide linker may be of variable length and composition so long as the two variable domains are bridged without serious steric interference.
  • the linkers are comprised primarily of stretches of glycine and serine residues with some glutamic acid or lysine residues interspersed for solubility.
  • This application generally relates to compositions and methods for imaging cells, and more particularly to imaging probes for detection or imaging ex vivo or in vivo epidermal growth factor (EGFR) expressing cells.
  • EGFR plays important roles in cell growth, differentiation, and migration. Its positive signaling was found to cause increased proliferation, decreased apoptosis, and enhanced tumor cell motility and angiogenesis.
  • EGFR expression is frequently found in wide spectrum of human tumors of epithelial origin, including non-small cell lung cancer (NSCLC), breast, head and neck (SCCHN), gastric, colorectal, esophageal, prostate, bladder, renal, pancreatic, ovarian, and brain cancers.
  • NSCLC non-small cell lung cancer
  • SCCHN head and neck
  • gastric colorectal, esophageal
  • prostate prostate
  • renal pancreatic, ovarian
  • brain cancers can thus be used as a tumor or cancer specific target for receptor-mediated delivery systems of therapeutic agents and imaging probes.
  • FIG. 1 is a schematic illustration of an imaging probe 10 that can target EGFR expressing cells, such as EGFR expressing cancer cells (e.g., glioblastomas) in accordance with an embodiment described herein.
  • the imaging probe can include an EGFR targeting moiety 12 , a reporter moiety 14 , and a hydrophilic linker 16 that links or couples the EGFR targeting moiety 12 to the reporter moiety 14 .
  • the EGFR targeting moiety can be hydrophobic and include a peptide that selectively or specifically binds to EGFR.
  • the peptide can be identified using phage display technology.
  • Phage display technology provides a means for expressing a diverse population of random or selectively randomized peptides.
  • Various methods of phage display and methods for producing diverse populations of peptides are well known in the art. For example, methods for preparing diverse populations of binding domains on the surface of a phage have been described in U.S. Pat. No. 5,223,409.
  • phage vectors useful for producing a phage display library as well as methods for selecting potential binding domains and producing randomly or selectively mutated binding domains are also provided in U.S.
  • phage peptide display libraries including vectors and methods of diversifying the population of peptides that are expressed, are also described in Smith et al., 1993, Meth. Enzymol., 217:228-257, Scott et al., Science, 249:386-390, and two PCT publications WO 91/07141 and WO 91/07149.
  • Phage display technology can be particularly powerful when used, for example, with a codon based mutagenesis method, which can be used to produce random peptides or randomly or desirably biased peptides (see, e.g., U.S. Pat. No. 5,264,563).
  • These or other well-known methods can be used to produce a phage display library, which can be subjected to the in vivo phage display method in order to identify a peptide that homes to one or a few selected tissues.
  • the EGFR targeting moiety can include an EGF peptide ligand that selectively or specifically binds to EGFR expressed by cancer cells.
  • the EGF peptide ligand can have an amino acid sequence of YHWYGYTPQNVI (SEQ ID NO: 1) (referred to herein as GE 11).
  • GE 11 is a peptide discovered through phage display screening against EGFR.
  • the EGFR targeting moiety can be a cyclic analogue of an EGF peptide ligand (e.g., GE 11) that includes a stabilized conformational turn in the peptide.
  • the cyclic analogue can have an amino acid sequence of YHWY-cyclo[KYTPQE]VI (SEQ ID NO: 2).
  • the EGFR targeting moiety can include any other molecule, or complex of molecules, which is/are capable of interacting and/or binding with EGFR of the cell.
  • An example of another EGFR targeting moiety is an antibody that selectively or specifically binds to EGFR.
  • the antibody can be a monoclonal antibody, a polyclonal antibody, or a humanized antibody including without limitation: Fv fragments, single chain Fv (scFv) fragments, Fab′ fragments, F(ab′)2 fragments, single domain antibodies, camelized antibodies and antibody fragments, humanized antibodies and antibody fragments, and multivalent versions of the foregoing; multivalent targeting moieties including without limitation: monospecific or bispecific antibodies, such as disulfide stabilized Fv fragments, scFv tandems ((scFv) 2 fragments), diabodies, tribodies or tetrabodies, which typically are covalently linked or otherwise stabilized (i.e., leucine zipper or helix stabilized) scFv fragments; and receptor molecules, which naturally interact with a desired target molecule.
  • scFv single chain Fv
  • Fab′ fragments fragments
  • F(ab′)2 fragments single domain antibodies
  • camelized antibodies and antibody fragments came
  • Preparation of antibodies may be accomplished by any number of well-known methods for generating monoclonal antibodies. These methods typically include the step of immunization of animals, typically mice, with a desired immunogen (e.g., a desired target molecule or fragment thereof). Once the mice have been immunized, and preferably boosted one or more times with the desired immunogen(s), monoclonal antibody-producing hybridomas may be prepared and screened according to well known methods. See, for example, Kuby, Janis, Immunology, Third Edition, pp. 131-139, W.H. Freeman & Co. (1997), for a general overview of monoclonal antibody production, that portion of which is incorporated herein by reference.
  • a desired immunogen e.g., a desired target molecule or fragment thereof.
  • the reporter moiety linked to the EGFR targeting moiety by the linker region can include any reporter moiety or contrast agent that allows the probe to be detectable upon in vivo, in vitro, or ex vivo administration to a EGFR expressing cell and particularly allows the EGFR expressing cell to be optically imaged in vivo.
  • reporter moieties or contrast agents can include isotopic labels, such as a naturally non-abundant heavy isotope or radioactive isotope; optically detectable moieties, such as a chromophore, luminophore, fluorophore, quantum dot or nanoparticle light scattering label; electromagnetic spin label; calorimetric agent; magnetic substance; electron-rich material such as a metal; electrochemiluminescent label; moiety that can be detected based on a nuclear magnetic, paramagnetic, electrical, charge to mass, or thermal characteristic; light scattering or plasmon resonant materials, such as gold or silver particles; or multielement reporter systems, such as affinity tags including but not limited to enzyme and substrate reporter groups, and dinitrophenyl (DNP) reporter group and fluorophore labeled anti-DNP antibody and the like.
  • isotopic labels such as a naturally non-abundant heavy isotope or radioactive isotope
  • optically detectable moieties such as
  • Fluorophores that can potentially be used include, for example, fluorescent lanthanide complexes, including those of Europium and Terbium, fluorescein, fluorescein isothiocyanate, dichlorotriazinylamine fluorescein, rhodamine, tetramethylrhodamine, umbelliferone, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, Cy3, Cy5, Cy5.5, ZW800-1 (i.e., N-hydroxysuccinimide (NHS) ester), stilbene, Lucifer Yellow, Cascade Blue, Texas Red, alexa dyes, dansyl chloride, phycoerythin, luciferin, green fluorescent protein and its wavelength shifted variants, bodipy, and others known in the art such as those described in Haugland, Molecular Probes Handbook, (Eugene, Oreg.) 6th Edition; The Synthegen catalog (Houston,
  • the reporter moiety can include a near infrared fluorescence (NIRF) imaging moiety or fluorochrome, such as Cy3, Cy5, Cy5.5, or ZW800-1, that can be linked to the EGFR targeting moiety with the hydrophilic linker.
  • NIRF near infrared fluorescence
  • the hydrophilic linker that links or couples the EGFR targeting moiety to the reporter moiety can include any linker, chemical, and/or or biological moiety that enhances solubility of the probe in an aqueous media and enhances the binding affinity of the EGFR targeting moiety to EGFR.
  • the linker can be of any suitable length and contain any suitable number of atoms and/or subunits.
  • the linker region is sufficiently long and hydrophilic to offset the hydrophobicity of the EGFR targeting moiety.
  • the linker can comprise a monomer, dimer, or oligomer PEG linker.
  • the PEG linker can be attached to a hydrophobic EGFR targeting moiety and reporter moiety in various lengths.
  • a probe for imaging and/or detecting an EGFR expressing cell can include a near infrared fluorescence (NIRF) or fluorochrome reporter moiety coupled to a EGF peptide ligand (e.g., GE11) by a linker.
  • NIRF near infrared fluorescence
  • fluorochrome can be, for example, Cy5, Cy5.5, ZW800-1
  • the linker can be a PEG linker, such as an amino-ethoxy-ethoxy-acid linker.
  • the probe can include the following formula:
  • R is a reporter moiety, such as a NIRF reporter moiety or fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3.
  • a reporter moiety such as a NIRF reporter moiety or fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1)
  • fluorochrome e.g., Cy5, Cy5.5, or ZW800-1
  • n is an integer from 1 to 3.
  • the probe can have the following formula:
  • n is an integer from 1 to 3 (e.g., 1).
  • a probe for imaging and/or detecting an EGFR expressing cell can include a near infrared fluorescence (NIRF) or fluorochrome reporter moiety coupled to a cyclic analogue of EGF peptide ligand by a linker.
  • the NIRF reporter moiety or fluorochrome can be, for example, Cy5, Cy5.5, ZW800-1, and the linker can be a PEG linker, such as an amino-ethoxy-ethoxy-acid linker.
  • the probe can include the following formula:
  • R is a reporter moiety, such as a NIRF reporter moiety or a fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3.
  • a reporter moiety such as a NIRF reporter moiety or a fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3.
  • fluorochrome e.g., Cy5, Cy5.5, or ZW800-1
  • n is an integer from 1 to 3.
  • the probe can have the following formula:
  • n is an integer from 1 to 3 (e.g., 1).
  • the probe can be used to detect cancer cells expressing EGFR at relatively short time scale and distinguish between cancer cells with high and low expression levels.
  • the cell may be in vivo or ex vivo.
  • Ex vivo cells can be collected as part of one or more samples using one or a combination of known techniques (e.g., biopsy) and, if needed, further processed (e.g., centrifuged) prior to culture, analysis, etc.
  • the probe can be applied (e.g., topically) to the collected cells and used, for example, in immunohistochemistry assays to determine whether the collected cells express EGFR.
  • a diagnostically effective amount of the probe can be administered to a subject.
  • Methods of introduction include, but are not limited to, topical, local, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, rectal, vaginal, and oral routes.
  • the probe may be administered by any convenient route, such as by infusion or bolus injection or by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, vaginal, rectal and intestinal mucosa, etc.), and may be administered together with other biologically active agents.
  • the probe may be introduced into the central nervous system by any suitable route, including intraventricular injection, intrathecal injection, or intraventricular injection via an intraventricular catheter that is attached to a reservoir.
  • the probe can be delivered systematically (e.g., intra-venously), regionally, or locally (e.g., intra- or peri-tumoral injection) by, for example, intra-arterial, intra-tumoral, intra-venous, parenteral, intra-pneural cavity, topical, oral or local administration, as well as subcutaneous, intra-zacheral (e.g., by aerosol), or transmucosal (e.g., voccal, bladder, vaginal, uterine, rectal, nasal, mucosal).
  • probe can be injected into an artery of the carotid system of arteries (e.g., occipital artery, auricular artery, temporal artery, cerebral artery, maxillary artery etc.).
  • arteries e.g., occipital artery, auricular artery, temporal artery, cerebral artery, maxillary artery etc.
  • the amount of the probe sufficient to provide the desired result(s) will depend on a variety of factors, such as the amount required to avoid undesirable physiological results.
  • the precise dose to be employed can also depend on the route of administration, and should be decided according to the judgment of a medical practitioner and each subject's circumstances.
  • known in vitro and in vivo assays may optionally be employed to help identify optimal dosage ranges. Effective doses may be extrapolated from dose-response curves derived from in vitro or in vivo test systems.
  • the probe can be administered in a variety of unit dosage forms, depending upon the particular cell or tissue being imaged, the general medical condition of each subject, the method of administration, and the like. Details on dosages are well described in the scientific literature. The exact amount and concentration of the imaging composition, or the “effective dose”, can be routinely determined (e.g., by a medical practitioner).
  • the “dosing regimen” will depend upon a variety of factors, such as whether the cell or tissue to be imaged is disseminated or local, the general state of the subject's health, the subject's age, and the like. Using guidelines describing alternative dosing regimens, e.g., from the use of other imaging agents and compositions, the skilled artisan can readily determine by routine trials the optimal effective concentrations of the imaging composition.
  • the probe can be used as part of an intra-operative molecular imaging (IOI) procedure, such as disclosed in U.S. patent application Ser. No. 11/811,818 to Basilion, to detect abnormal EGFR expressing cells and/or diseased tissue.
  • IOI intra-operative molecular imaging
  • microscopic IOI techniques can be combined with topically applied probes described herein.
  • the probe can be delivered via topical application, as opposed to intravenous injection, which facilitates localized diffusion of the imaging probes.
  • the imaging probe can be combined with IOI to identify malignant cells that have infiltrated and/or are beginning to infiltrate at a tumor brain margin.
  • the method can be performed in real-time during brain or other surgery.
  • the method can include topical application of an imaging probe.
  • An imaging modality can then be used to detect and subsequently gather image data.
  • the resultant image data may be used to determine, at least in part, a radiological treatment.
  • this image data may be used to control, at least in part, an automated surgical device (e.g., laser, scalpel, and micromachine) or to aid in manual guidance of surgery.
  • the image data may be used to plan and/or control the delivery of a therapeutic agent (e.g., by a micro-electronic machine or micro-machine).
  • the present application provides a method for treating a disease in a subject.
  • One step of the method can include contacting the EGFR expressing cell with a diagnostically effective amount of the imaging probe.
  • one or more imaging modalities can be used to detect the reporter moiety to produce a molecular signature of the cell or differentiate the cell. Based on the molecular signature of the cell, the subject can then be appropriately treated for the particular disease by, for example, administering an effective amount of a therapeutic composition.
  • an “effective amount” can refer to that amount of a therapeutic agent that results in amelioration of symptoms or a prolongation of survival in the subject and relieves, to some extent, one or more symptoms of the disease or returns to normal (either partially or completely) one or more physiological or biochemical parameters associated with or causative of the disease.
  • Therapeutic agents can include any agent (e.g., molecule, drug, pharmaceutical composition, etc.) capable of preventing, inhibiting, or arresting the symptoms and/or progression of a disease.
  • a molecular signal of a cell can indicate that a subject has a particular cancer. Because the type, severity, and stage of the cancer can be determined based on the molecular signature of the cell, the subject can then be administered an effective amount of a chemotherapy agent to treat the cancer.
  • Chemotherapy agents are well known in the art and can include, for example, alkylating agents (e.g., cyclophosphamide, ifosfamide), antibiotics that affect nucleic acids (e.g., doxorubicin, bleomycin), platinum compounds (e.g., cisplatin), mitotic inhibitors (e.g., vincristine), antimetabolites (e.g., 5-fluorouracil), camptothecin derivatives (e.g., topotecan), biological response modifiers (e.g., interferon), and hormone therapies (e.g., tamoxifen).
  • alkylating agents e.g., cyclophosphamide, ifosfamide
  • antibiotics that affect nucleic acids e.g., doxorubicin, bleomycin
  • platinum compounds e.g., cisplatin
  • mitotic inhibitors e.g., vincristine
  • the imaging probe can be used in a method to identify cells that are treatable by a particular therapeutic agent or therapeutic intervention. Additionally, the imaging probe can be used to the progress of therapy by monitoring a decreasing signal over time.
  • peptides were synthesized manually using protocols previously described. Peptide was labeled in solution with monoreactive Cy5.5 NHS ester (CyDye, GE Healthcare). Crude fluorophore-labeled peptides were purified by reversed phase high-performance liquid chromatography (RP-HPLC). The isolated peak was lyophilized and characterized matrix-assisted laser desorption/ionization (MALDI) mass spectrometry was done under positive mode. Compounds are characterized by RPHPLC under 2 different conditions and by thin layer chromatography with 3 different solvent conditions.
  • MALDI matrix-assisted laser desorption/ionization
  • glioblastoma astrocytoma an epithelial-like cell line U87-MG, and human glioblastoma cell line stably over expressing the EGFRvIII-mutant form of the egfr gene, Gli36 ⁇ 5, were used in these studies.
  • U87-MG and A431 cells were recently obtained from American Type Culture Collection.
  • Gli36 ⁇ 5 cells were obtained from E.A. Chiocca and were authenticated by Research Animal Diagnostic Laboratory at the University of Missouri (Columbia, Mo.) for interspecies and mycoplasma contamination by PCR analysis.
  • DMSO was added before reading fluorescence (excitation 670 nm, emission 700 nm) with a Tecan Infinite M200 plate reader. Saturation binding assay data were in quadruplicates and analyzed using one site binding classical equation for nonlinear regression analysis with the GraphPad Prism version 4.0.
  • Gli36 ⁇ 5 or U87-MG cell lines plated in 96-well culture plates (20,000 cells per well; 3 wells per condition) were incubated over indicated time periods with 1 mmol/L of Cy5.5-labeled EGF peptides 2, 3, and 4 at 37° C. and 5% CO 2 .
  • Cells were briefly rinsed with HEPES buffer and imaged with a Tecan Infinite M200 fluorescence plate reader (excitation 670 nm, emission 700 nm).
  • Gli36 ⁇ 5 or U87-MG cancer cell lines were plated on coverslips and incubated overnight to promote adherence. The cells were then fixed with 4% paraformaldehyde, rinsed with PBS, and blocked with 1% host serum for 30 minutes at room temperature. Coverslips were incubated with primary antibody at room temperature for 2 hours. Antibodies used were mouse anti-human wild-type EGFR (1:100 dilution; clone DAK-H1-WT, Dako cat. no. M7289). The coverslips were then rinsed with PBS and counterstained with 40,6-diamidino-2-phenylindole (DAPI) for 10 minutes at room temperature to visualize the nuclei. After a final rinse with PBS, the coverslips were mounted using Fluor-Mount aqueous media, sealed with nail polish, and observed using epifluorescence microscopy.
  • DAPI 40,6-diamidino-2-phenylindole
  • NIH athymic nude female mice (5-8 weeks and 20-25 g upon arrival, NCI-NIH) were maintained at the Animal Resource Center at Case Western Reserve University (Cleveland, Ohio) according to institutional policies.
  • Cell for brain implantation were harvested with 1 mL 0.05% trypsin-EDTA (Gibco) and briefly washed with PBS. Trypsin was inactivated by the addition of serum-containing media. The resulting cell suspension was centrifuged at 1,000 ⁇ g for 3 minutes. The cell suspension was centrifuged and the supernatant was removed after 2 washes in PBS. Finally, the cells were resuspended in 2 mL PBS for brain implants per animal (250,000 cells per animal for brain implants).
  • mice were inoculated.
  • mice were anesthetized by intraperitoneal (i.p.) injection of 50 mg/kg ketamine/xylazine and fitted into a stereotaxic rodent frame (David Kopf Instruments).
  • a stereotaxic rodent frame (David Kopf Instruments).
  • a small incision was made just lateral to midline to expose bregma suture.
  • Glioblastoma cells were slowly deposited at a rate of 1 ⁇ L/min in the right striatum at a depth of 3 mm from dura with a 10- ⁇ L syringe (23-G needle).
  • mice bearing brain tumors derived from Gli36 ⁇ 5 or U87-MG cells were administered with compounds at 1 nmol/g via tail vein injection. In vivo competition assays were done with mixture of compound 2 and 10-fold concentration of nonlabeled probe 5. Before injection, mice were anesthetized with isoflurane and subjected to tomographic and spectral fluorescence imaging. One hour after injection, animals were re-imaged. Brains were then extracted and imaged. The excised brains were embedded in Tissue-Tek optimum cutting temperature for cryosections for immunohistochemistry (IHC).
  • IHC immunohistochemistry
  • Fluorescence-mediated molecular tomography (FMT) images were obtained using FMT2500 (Perkin-Elmer), and 3-dimensional reconstructions of fluorescent signals were acquired using the accompanying software TrueQuant. Quantitative fluorescent signals for Cy5.5 of compound 2 were calibrated as per manufacturer's instructions using the 680-channel. Region of interest (ROI) assigned on the basis of the precise placement of cells during implantation at 3 to 4 mm into the brain. ROI was corroborated with fluorescent signals from ex vivo imaging. Fluorescent multispectral images were obtained using the Maestro In-Vivo Imaging System (CRi, Inc.). The yellow filter set appropriate for Cy5.5 was used for emission and excitation light.
  • ROI Region of interest
  • the tunable filter was automatically stepped in 10-nm increments whereas the camera captured images at a constant exposure of 200 ms. Fluorescence images were acquired before treatment, immediately after and 1.5 hours after treatment. To compare signal intensities, ROIs were selected over the tumor or nontumor areas, and the change in fluorescence signal over baseline was determined. The spectral fluorescent images consisting of autofluorescence spectra and imaging probe were captured and unmixed on the basis of their spectral patterns. The total signal in the ROI defined in photons measured at the surface of the animal was divided by the area (in pixels). Spectral libraries were generated by assigning spectral peaks to background and fluorescence probe on tissue. The spectral libraries were manually computed using the Maestro software, with each tissue used as its own background control.
  • Sections (2 mm) of whole mouse brains implanted with Gli36 ⁇ 5 cells were fixed with 4% paraformaldehyde, cryosectioned onto microscope slides, rinsed with PBS, and blocked with 1% host serum for 30 minutes at room temperature. Sections were incubated with primary antibody at room temperature for 2 hours. Antibodies used were mouse anti-human wild-type EGFR (1:100 dilution; clone DAK-H1-WT, Dako cat. no. M7289). The coverslips were then rinsed with PBS and counterstained with DAPI for 10 minutes at room temperature to visualize the nuclei. After a final rinse with PBS, the slides were mounted with coverslips using Fluor-Mount aqueous media, sealed with nail polish, and observed using epifluorescence microscopy.
  • Cell extracts (50 ⁇ g) were fractionated using SDS-PAGE and transferred onto a nitrocellulose membrane. Immunoblotting was done using a 1:500 dilution of an antibody against wild-type EGFR (DAKO, DAK-H1-WT) or a 1:500 dilution of a specific antibody against mutant EGFRvIII (Bioss Inc., cat #bs-2558R). Horseradish peroxidase-conjugated secondary antibodies against mouse IgG (Chemicon) or rabbit IgG (Amersham) were used. Bands were detected using an enhanced chemiluminescence detection system (Pierce).
  • ANOVA analysis at 95% confidence interval (CI) was used to compare treatments (*, P>0.05; **, P>0.01; confidence interval, ***, P>0.001).
  • nonparametric one-way ANOVA analyses Karl-Wallis tests
  • mice were orthotopically implanted with Gli36 ⁇ 5 cells. Approximately 10 days after implantation, the animals were administered 1 nmol/g via tail vein injection and sacrificed 1 hour later. Brains were harvested and imaged ex vivo for accumulation of the imaging probe.
  • a Cy5.5-labeled scrambled peptide, compound 6 using the amino acid residues of the parent peptide (compound 5) in random order. Compound 2 targeted the tumor efficiently, accumulating 1.1% of injected dose ( FIG. 3A , left graph).
  • compound 6 did not target the tumor with delivery of only 0.006% of the injected dose.
  • animals that bore Gli36 ⁇ 5 brain tumors were administered compound 2 alone or in the presence of a 10-fold excess of unlabeled peptide (compound 5). In animals that received only compound 2, there was significant tumor-associated fluorescence. In contrast, when a 10-fold excess of competitor peptide was coadministered with compound 2, there was approximately a 60% decrease in accumulation of the probe, ( FIG. 4A , right graph). Imaging and quantification of identical ROIs taken on the contralateral brain showed little uptake of the probe (data not shown).
  • mice were orthotopically implanted with either Gli36 ⁇ 5 cells, which express high EGFR, or U87-MG cells, which express relatively low EGFR levels ( FIG. 6A ). Following implantation, the tumors were allowed to grow approximately 10 to 12 days and then mice were administered 1 nmol/g of compound 2 via tail vein injection. One hour after injection, the mice were anesthetized and the intensity of Cy5.5 fluorescence from the tumor was noninvasively quantified using FMT ( FIG. 7 ).
  • the tumors formed from Gli36D5 cells had approximately 4-fold more fluorescence than either tumors formed with U87-MG cells or control sham surgeries ( FIG. 8A ).
  • these fluorescence signals corresponded to injected doses of 1.1% and 0.27%, respectively.
  • Statistical analyses showed that the median fluorescence signals between the 2 groups were significantly different ( FIG. 8A ).
  • tumors were excised and subjected to ex vivo FMT and Maestro fluorescence imaging analyses ( FIGS. 8B and 8C ). These measurements were in good agreement with those measurements made during the live animal imaging.
  • Peptides have conformational structures based on their sequences.
  • Tyr 1 -His 2 -Trp 3 -Tyr 4 -Gly 5 -Tyr 6 -Thr 7 -Pro 8 -Gln 9 -Asn 10 -Val 11 -Ile 12 sequence contains a turn at the Thr-Pro for binding to EGF receptors. Since linear peptides are flexible, we designed and synthesized a peptide that will stabilize that conformational turn. Gly 5 and Asn 10 sites were replaced with Lys and Glu, respectively, to form a side chain to side chain lactam cyclic bridge. Cy5 NIR label and miniPEG linker were also added as shown below.
  • the cyclic peptide was generated using Fmoc chemistry on solid support on a Rink amide resin.
  • Fmoc-Glu(OAllyl)-OH and Fmoc-Lys(Alloc)-OH were used for the lactam bridge cyclization on resin.
  • the side chain groups OAllyl and Alloc were removed using palladium catalysts and the subsequent ring closure was achieved by HCTU coupling. Standard cleavage procedures were subsequently used to isolate the pure peptide. Cy5.5 conjugation was also a standard procedure.

Abstract

A probe for imaging EGFR expressing cells includes an EGFR targeting moiety, a reporter moiety, and a hydrophilic linker that links the EGFR targeting moiety to the reporter moiety. The hydrophilic linker enhances solubility of the probe in an aqueous media as well as binding affinity of the probe to EGFR expressing cells.

Description

    RELATED APPLICATION
  • This application claims priority from U.S. Provisional Application No. 61/557,025, filed Nov. 8, 2011, the subject matter of which is incorporated herein by reference in its entirety.
  • BACKGROUND
  • Non-invasive optical imaging technologies can be used for diagnosis and monitoring therapeutic response. As optical instrumentation advances (e.g., tomographic imaging), the development of optical imaging agents that are selective for tumors for in vivo or in vitro studies is fast becoming an important field for cancer research. One type of such agents is a molecule labeled with near infrared fluorescence (NIRF) or fluorochrome reporter moiety.
  • Glioblastoma multiforme (GBM) is the most common and most malignant of the glial tumors. In 40-50% of these tumors, mutations resulting to over expression or activation of epidermal growth factors are found. Epidermal growth factors receptors (EGFR) are cell surface receptors that regulate growth and survival as well as adhesion, migration, differentiation and other cellular processes. The ability to differentially image EGFR expression levels might provide non-invasive means to identify tumor biomarker that can aide in the selection of treatment as well as means of targeted drug delivery.
  • SUMMARY
  • This application relates to a probe for imaging EGFR expressing cells of a subject. The probe can include an EGFR targeting moiety, a reporter moiety, and a hydrophilic linker that links or couples the EGFR targeting moiety to the reporter moiety. The hydrophilic linker can enhance solubility of the probe in an aqueous media as well as binding affinity of the probe to EGFR expressing cells.
  • In some aspects, the EGFR expressing cells can include EGFR expressing cancer cells. The EGFR expressing cancer cells can include glioblastomas as well as solid tumors, such as breast, lung, ovarian, and colon cancers. The probe upon systemic administration to subject is capable of crossing the blood brain barrier, targeting the EGFR expressing cancer cells, and imaging the EGFR expressing cancer cells.
  • In other aspects, the EGFR targeting moiety can include an EGF peptide ligand having an amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2. The reporter moiety can include a fluorochrome, and the linker comprising a polyethylene glycol linker.
  • In one embodiment, the probe can include the following formula:
  • Figure US20130116404A1-20130509-C00001
  • where R is a reporter moiety, such as a NIRF reporter moiety or fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3. By way of example the probe can have the following formula:
  • Figure US20130116404A1-20130509-C00002
  • where n is an integer from 1 to 3 (e.g., 1).
  • In other embodiments, the probe can include the following formula:
  • Figure US20130116404A1-20130509-C00003
  • where R is a reporter moiety, such as a NIRF reporter moiety or a fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3. By way of example the probe can have the following formula:
  • Figure US20130116404A1-20130509-C00004
  • where n is an integer from 1 to 3 (e.g., 1).
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic illustration of an imaging probe in accordance with an embodiment of the application.
  • FIG. 2 illustrates a plot showing cell associated fluorescence measured using spectrofluorometry at the indicated times for Gli36Δ5 cells incubated with 1 μmol/L of compounds 1 to 4 [Cy5.5-(AEEA)n-EGFpep].
  • FIGS. 3(A-B) illustrate images showing Gli36Δ5 (A) and U87-MG (B) cells incubated with 1 μmol/L compounds for the indicated times. Uptake of the compounds was assessed using epifluorescence microscopy. Images were taken at ×40 magnification. Scale bar, 50 μm.
  • FIG. 4 illustrates graphs showing the specificity of the targeting assessed using a scrambled peptide (6) via competition against unlabeled parent peptide compound (5). Subjects bearing orthotopic brain tumors derived from Gli36Δ5 cells over expressing EGFR were treated with (1 nmol/g mice) of compound 6 (n=3) or compound 2 (n=5). Ex vivo analysis of brain tissues showed that compound 2 targets the tumor significantly (***, P>0.0001) more efficiently than the scrambled peptide 6. Representative images of the brain tumors are shown above the corresponding bars (left graph). When subjects bearing orthotopic brain tumors derived from Gli36Δ5 cells over expressing EGFR were cotreated with compound 2 and a 10-fold excess of nonlabeled parent compound 5, the signal over the tumor region was significantly reduced. This suggests that the fluorescence labeling of tumor is specific to the peptide sequence of 2.
  • FIG. 5 illustrates images showing the colocalization of EGFR-targeted probe with cells expressing high levels of EGFR. Scale bar, 20 mm
  • FIGS. 6(A-B) illustrate western blots showing (A) showing the relative levels of wild-type EGFR expressed in Gli36Δ5 and U87-MG glioblastoma cell lysates using an antibody specific to wild-type EGFR (DAKO, cat #M7289); and (B) comparing the mutant EGFRvIII content in Gli36Δ5 and U87-MG tumor cells lines using an EGFRvIII-specific antibody (Bioss Inc., cat #bs-2558R). β-Actin was used a loading control. Densitometry analysis of Western blotting by ImageJ comparing the levels of EGFRvIII in Gli36Δ5 and U87-MG cells show similar content of 1,800 and 2,400 arbitrary units, respectively.
  • FIG. 7 illustrates an image of FMT3-dimensional reconstruction showing the targeting of compound 2 to brain Gli36Δ5 orthotopic brain tumors. Sham, U87-MG, and Gli36Δ5 shows 272, 1,608, and 9,271 nmol/L amounts of fluorescence in the matching ROIs.
  • FIGS. 8(A-C) illustrate graphs showing the detected level of the fluorescent agent in (ppmol) administered to live animals (A) or ex vivo brains (B and C) with brain tumors, Gli36Δ5 and U87, using FMT imaging and Maestro imaging (C).
  • DETAILED DESCRIPTION
  • All scientific and technical terms used in this application have meanings commonly used in the art unless otherwise specified. The definitions provided herein are to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the application.
  • The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.
  • In the context of the application, the term “sample” can refer to a specimen or culture obtained from any source, as well as clinical, research, biological and environmental samples. Biological samples may be obtained from animals (including humans) and encompass cells, fluids, solids, tissues, and organs, and whole organisms.
  • As used herein, the term “subject” can refer to any animal including, but not limited to, humans and non-human animals (e.g., rodents, arthropods, insects, fish (e.g., zebrafish)), non-human primates, ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, or canines felines, aves, etc.).
  • As used herein, the terms “cancer” or “tumor” refer to any neoplastic growth in a subject, including an initial tumor and any metastases. The cancer can be of the liquid or solid tumor type. Liquid tumors include tumors of hematological origin, including, e.g., myelomas (e.g., multiple myeloma), leukemias (e.g., Waldenstrom's syndrome, chronic lymphocytic leukemia, other leukemias), and lymphomas (e.g., B-cell lymphomas, non-Hodgkin's lymphoma). Solid tumors can originate in organs and include cancers of the lungs, brain, breasts, prostate, ovaries, colon, kidneys and liver.
  • As used herein, the terms “cancer cell” or “tumor cell” can refer to cells that divide at an abnormal (i.e., increased) rate. Cancer cells include, but are not limited to, carcinomas, such as squamous cell carcinoma, non-small cell carcinoma (e.g., non-small cell lung carcinoma), small cell carcinoma (e.g., small cell lung carcinoma), basal cell carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, adenocarcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, undifferentiated carcinoma, bronchogenic carcinoma, melanoma, renal cell carcinoma, hepatoma-liver cell carcinoma, bile duct carcinoma, cholangiocarcinoma, papillary carcinoma, transitional cell carcinoma, choriocarcinoma, semonoma, embryonal carcinoma, mammary carcinomas, gastrointestinal carcinoma, colonic carcinomas, bladder carcinoma, prostate carcinoma, and squamous cell carcinoma of the neck and head region; sarcomas, such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordosarcoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, synoviosarcoma and mesotheliosarcoma; hematologic cancers, such as myelomas, leukemias (e.g., acute myelogenous leukemia, chronic lymphocytic leukemia, granulocytic leukemia, monocytic leukemia, lymphocytic leukemia), lymphomas (e.g., follicular lymphoma, mantle cell lymphoma, diffuse large B-cell lymphoma, malignant lymphoma, plasmocytoma, reticulum cell sarcoma, or Hodgkin's disease), and tumors of the nervous system including glioma, meningoma, medulloblastoma, schwannoma and epidymoma.
  • As used herein, the terms “polypeptide” or “peptide” are used interchangeably and can refer to an oligopeptide, peptide, polypeptide, or protein sequence, or to a fragment, portion, or subunit of any of these, and to naturally occurring or synthetic molecules. The term “polypeptide” can also include amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain any type of modified amino acids. The term can also include peptides and polypeptide fragments, motifs and the like, glycosylated polypeptides, and all “mimetic” and “peptidomimetic” polypeptide forms.
  • As used herein, the terms “probe” or “imaging probe” can refer to a biological moiety that may be used to detect, image, and/or monitor the presence and/or progression of a cell cycle, cell function/physiology, condition, pathological disorder and/or disease.
  • As used herein, the term “diagnostically effective amount” can refer to an amount of an imaging composition that is sufficient to enable imaging of at least one cell, tissue, or organism using an imaging modality.
  • As used herein, the term “targeting moiety” can refer to a molecule or molecules that are able to bind to and complex with a biomarker. The term can also refer to a functional group that serves to target or direct an imaging probe to a particular location, cell type, diseased tissue, or association. In general, a “targeting moiety” can be directed against a biomarker.
  • As used herein, the term “antibody” refers to an immunoglobulin, derivatives thereof which maintain specific binding ability, and proteins having a binding domain which is homologous or largely homologous to an immunoglobulin binding domain. These proteins may be derived from natural sources, or partly or wholly synthetically produced. An antibody may be monoclonal or polyclonal. The antibody may be a member of any immunoglobulin class, including any of the human classes: IgG, IgM, IgA, IgD, and IgE. In exemplary embodiments, antibodies used with the methods and compositions described herein are derivatives of the IgG class.
  • As used herein, the term “antibody fragment” refers to any derivative of an antibody which is less than full-length. In exemplary embodiments, the antibody fragment retains at least a significant portion of the full-length antibody's specific binding ability. Examples of antibody fragments include, but are not limited to, Fab, Fab′, F(ab′)2, scFv, Fv, dsFv diabody, and Fd fragments. The antibody fragment may be produced by any means. For instance, the antibody fragment may be enzymatically or chemically produced by fragmentation of an intact antibody, it may be recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced. The antibody fragment may optionally be a single chain antibody fragment. Alternatively, the fragment may comprise multiple chains which are linked together, for instance, by disulfide linkages. The fragment may also optionally be a multimolecular complex. A functional antibody fragment will typically comprise at least about 50 amino acids and more typically will comprise at least about 200 amino acids.
  • As used herein, the term “diabodies” refers to dimeric scFvs. The components of diabodies typically have shorter peptide linkers than most scFvs and they show a preference for associating as dimers.
  • As used herein, the term “epitope” refers to a physical structure on a molecule that interacts with a selective component. In exemplary embodiments, epitope refers to a desired region on a target molecule that specifically interacts with a selectivity component.
  • As used herein, the term “Fab” refers to an antibody fragment that is essentially equivalent to that obtained by digestion of immunoglobulin (typically IgG) with the enzyme papain. The heavy chain segment of the Fab fragment is the Fd piece. Such fragments may be enzymatically or chemically produced by fragmentation of an intact antibody, recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced.
  • As used herein, the term “Fab′” refers to an antibody fragment that is essentially equivalent to that obtained by reduction of the disulfide bridge or bridges joining the two heavy chain pieces in the F(ab′)2 fragment. Such fragments may be enzymatically or chemically produced by fragmentation of an intact antibody, recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced.
  • As used herein, the term “F(ab′)2” refers to an antibody fragment that is essentially equivalent to a fragment obtained by digestion of an immunoglobulin (typically IgG) with the enzyme pepsin at pH 4.0-4.5. Such fragments may be enzymatically or chemically produced by fragmentation of an intact antibody, recombinantly produced from a gene encoding the partial antibody sequence, or it may be wholly or partially synthetically produced.
  • As used herein, the term “Fv” refers to an antibody fragment that consists of one VH and one VL domain held together by noncovalent interactions. The term “dsFv” is used herein to refer to an Fv with an engineered intermolecular disulfide bond to stabilize the VH-VL pair.
  • As used herein, the terms “single-chain Fvs” and “scFvs” refers to recombinant antibody fragments consisting of only the variable light chain (VL) and variable heavy chain (VH) covalently connected to one another by a polypeptide linker. Either VL or VH may be the NH2-terminal domain. The polypeptide linker may be of variable length and composition so long as the two variable domains are bridged without serious steric interference. In exemplary embodiments, the linkers are comprised primarily of stretches of glycine and serine residues with some glutamic acid or lysine residues interspersed for solubility.
  • This application generally relates to compositions and methods for imaging cells, and more particularly to imaging probes for detection or imaging ex vivo or in vivo epidermal growth factor (EGFR) expressing cells. EGFR plays important roles in cell growth, differentiation, and migration. Its positive signaling was found to cause increased proliferation, decreased apoptosis, and enhanced tumor cell motility and angiogenesis. EGFR expression is frequently found in wide spectrum of human tumors of epithelial origin, including non-small cell lung cancer (NSCLC), breast, head and neck (SCCHN), gastric, colorectal, esophageal, prostate, bladder, renal, pancreatic, ovarian, and brain cancers. EGFR can thus be used as a tumor or cancer specific target for receptor-mediated delivery systems of therapeutic agents and imaging probes.
  • FIG. 1 is a schematic illustration of an imaging probe 10 that can target EGFR expressing cells, such as EGFR expressing cancer cells (e.g., glioblastomas) in accordance with an embodiment described herein. The imaging probe can include an EGFR targeting moiety 12, a reporter moiety 14, and a hydrophilic linker 16 that links or couples the EGFR targeting moiety 12 to the reporter moiety 14.
  • In some embodiments, the EGFR targeting moiety can be hydrophobic and include a peptide that selectively or specifically binds to EGFR. The peptide can be identified using phage display technology. Phage display technology provides a means for expressing a diverse population of random or selectively randomized peptides. Various methods of phage display and methods for producing diverse populations of peptides are well known in the art. For example, methods for preparing diverse populations of binding domains on the surface of a phage have been described in U.S. Pat. No. 5,223,409. In particular, phage vectors useful for producing a phage display library as well as methods for selecting potential binding domains and producing randomly or selectively mutated binding domains are also provided in U.S. Pat. No. 5,223,409. Similarly, methods of producing phage peptide display libraries, including vectors and methods of diversifying the population of peptides that are expressed, are also described in Smith et al., 1993, Meth. Enzymol., 217:228-257, Scott et al., Science, 249:386-390, and two PCT publications WO 91/07141 and WO 91/07149. Phage display technology can be particularly powerful when used, for example, with a codon based mutagenesis method, which can be used to produce random peptides or randomly or desirably biased peptides (see, e.g., U.S. Pat. No. 5,264,563). These or other well-known methods can be used to produce a phage display library, which can be subjected to the in vivo phage display method in order to identify a peptide that homes to one or a few selected tissues.
  • In vitro screening of phage libraries has previously been used to identify peptides that bind to antibodies or cell surface receptors (see, e.g., Smith, et al., 1993, Meth. Enzymol., 217:228-257). For example, in vitro screening of phage peptide display libraries has been used to identify novel peptides that specifically bind to integrin adhesion receptors (see, e.g., Koivunen et al., 1994, J. Cell Biol. 124:373-380), and to the human urokinase receptor (Goodson, et al., 1994, Proc. Natl. Acad. Sci., USA 91:7129-7133).
  • In certain embodiments, the EGFR targeting moiety can include an EGF peptide ligand that selectively or specifically binds to EGFR expressed by cancer cells. In one example, the EGF peptide ligand can have an amino acid sequence of YHWYGYTPQNVI (SEQ ID NO: 1) (referred to herein as GE 11). GE 11 is a peptide discovered through phage display screening against EGFR.
  • In other embodiments, the EGFR targeting moiety can be a cyclic analogue of an EGF peptide ligand (e.g., GE 11) that includes a stabilized conformational turn in the peptide. In one example, the cyclic analogue can have an amino acid sequence of YHWY-cyclo[KYTPQE]VI (SEQ ID NO: 2).
  • The EGFR targeting moiety can include any other molecule, or complex of molecules, which is/are capable of interacting and/or binding with EGFR of the cell. An example of another EGFR targeting moiety is an antibody that selectively or specifically binds to EGFR. The antibody can be a monoclonal antibody, a polyclonal antibody, or a humanized antibody including without limitation: Fv fragments, single chain Fv (scFv) fragments, Fab′ fragments, F(ab′)2 fragments, single domain antibodies, camelized antibodies and antibody fragments, humanized antibodies and antibody fragments, and multivalent versions of the foregoing; multivalent targeting moieties including without limitation: monospecific or bispecific antibodies, such as disulfide stabilized Fv fragments, scFv tandems ((scFv)2 fragments), diabodies, tribodies or tetrabodies, which typically are covalently linked or otherwise stabilized (i.e., leucine zipper or helix stabilized) scFv fragments; and receptor molecules, which naturally interact with a desired target molecule.
  • Preparation of antibodies may be accomplished by any number of well-known methods for generating monoclonal antibodies. These methods typically include the step of immunization of animals, typically mice, with a desired immunogen (e.g., a desired target molecule or fragment thereof). Once the mice have been immunized, and preferably boosted one or more times with the desired immunogen(s), monoclonal antibody-producing hybridomas may be prepared and screened according to well known methods. See, for example, Kuby, Janis, Immunology, Third Edition, pp. 131-139, W.H. Freeman & Co. (1997), for a general overview of monoclonal antibody production, that portion of which is incorporated herein by reference.
  • The reporter moiety linked to the EGFR targeting moiety by the linker region can include any reporter moiety or contrast agent that allows the probe to be detectable upon in vivo, in vitro, or ex vivo administration to a EGFR expressing cell and particularly allows the EGFR expressing cell to be optically imaged in vivo. Examples of such reporter moieties or contrast agents can include isotopic labels, such as a naturally non-abundant heavy isotope or radioactive isotope; optically detectable moieties, such as a chromophore, luminophore, fluorophore, quantum dot or nanoparticle light scattering label; electromagnetic spin label; calorimetric agent; magnetic substance; electron-rich material such as a metal; electrochemiluminescent label; moiety that can be detected based on a nuclear magnetic, paramagnetic, electrical, charge to mass, or thermal characteristic; light scattering or plasmon resonant materials, such as gold or silver particles; or multielement reporter systems, such as affinity tags including but not limited to enzyme and substrate reporter groups, and dinitrophenyl (DNP) reporter group and fluorophore labeled anti-DNP antibody and the like. Fluorophores that can potentially be used include, for example, fluorescent lanthanide complexes, including those of Europium and Terbium, fluorescein, fluorescein isothiocyanate, dichlorotriazinylamine fluorescein, rhodamine, tetramethylrhodamine, umbelliferone, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, Cy3, Cy5, Cy5.5, ZW800-1 (i.e., N-hydroxysuccinimide (NHS) ester), stilbene, Lucifer Yellow, Cascade Blue, Texas Red, alexa dyes, dansyl chloride, phycoerythin, luciferin, green fluorescent protein and its wavelength shifted variants, bodipy, and others known in the art such as those described in Haugland, Molecular Probes Handbook, (Eugene, Oreg.) 6th Edition; The Synthegen catalog (Houston, Tex.), Lakowicz, Principles of Fluorescence Spectroscopy, 2nd Ed., Plenum Press New York (1999), or WO 98/59066.
  • In a particular embodiment, the reporter moiety can include a near infrared fluorescence (NIRF) imaging moiety or fluorochrome, such as Cy3, Cy5, Cy5.5, or ZW800-1, that can be linked to the EGFR targeting moiety with the hydrophilic linker.
  • The hydrophilic linker that links or couples the EGFR targeting moiety to the reporter moiety can include any linker, chemical, and/or or biological moiety that enhances solubility of the probe in an aqueous media and enhances the binding affinity of the EGFR targeting moiety to EGFR. The linker can be of any suitable length and contain any suitable number of atoms and/or subunits. Advantageously, the linker region is sufficiently long and hydrophilic to offset the hydrophobicity of the EGFR targeting moiety. In one example, the linker can comprise a monomer, dimer, or oligomer PEG linker. The PEG linker can be attached to a hydrophobic EGFR targeting moiety and reporter moiety in various lengths.
  • In some embodiments, a probe for imaging and/or detecting an EGFR expressing cell can include a near infrared fluorescence (NIRF) or fluorochrome reporter moiety coupled to a EGF peptide ligand (e.g., GE11) by a linker. The NIRF reporter moiety or fluorochrome can be, for example, Cy5, Cy5.5, ZW800-1, and the linker can be a PEG linker, such as an amino-ethoxy-ethoxy-acid linker.
  • In one example, the probe can include the following formula:
  • Figure US20130116404A1-20130509-C00005
  • where R is a reporter moiety, such as a NIRF reporter moiety or fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3. By way of example the probe can have the following formula:
  • Figure US20130116404A1-20130509-C00006
  • where n is an integer from 1 to 3 (e.g., 1).
  • In other embodiments, a probe for imaging and/or detecting an EGFR expressing cell can include a near infrared fluorescence (NIRF) or fluorochrome reporter moiety coupled to a cyclic analogue of EGF peptide ligand by a linker. The NIRF reporter moiety or fluorochrome can be, for example, Cy5, Cy5.5, ZW800-1, and the linker can be a PEG linker, such as an amino-ethoxy-ethoxy-acid linker.
  • In one example, the probe can include the following formula:
  • Figure US20130116404A1-20130509-C00007
  • where R is a reporter moiety, such as a NIRF reporter moiety or a fluorochrome (e.g., Cy5, Cy5.5, or ZW800-1) and n is an integer from 1 to 3. By way of example the probe can have the following formula:
  • Figure US20130116404A1-20130509-C00008
  • where n is an integer from 1 to 3 (e.g., 1).
  • In some embodiments, the probe can be used to detect cancer cells expressing EGFR at relatively short time scale and distinguish between cancer cells with high and low expression levels. Depending upon the particular application, the cell may be in vivo or ex vivo. Ex vivo cells can be collected as part of one or more samples using one or a combination of known techniques (e.g., biopsy) and, if needed, further processed (e.g., centrifuged) prior to culture, analysis, etc. The probe can be applied (e.g., topically) to the collected cells and used, for example, in immunohistochemistry assays to determine whether the collected cells express EGFR.
  • To detect an EGFR expressing cell in vivo, a diagnostically effective amount of the probe can be administered to a subject. Methods of introduction include, but are not limited to, topical, local, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, rectal, vaginal, and oral routes. The probe may be administered by any convenient route, such as by infusion or bolus injection or by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, vaginal, rectal and intestinal mucosa, etc.), and may be administered together with other biologically active agents. The probe may be introduced into the central nervous system by any suitable route, including intraventricular injection, intrathecal injection, or intraventricular injection via an intraventricular catheter that is attached to a reservoir.
  • The probe can be delivered systematically (e.g., intra-venously), regionally, or locally (e.g., intra- or peri-tumoral injection) by, for example, intra-arterial, intra-tumoral, intra-venous, parenteral, intra-pneural cavity, topical, oral or local administration, as well as subcutaneous, intra-zacheral (e.g., by aerosol), or transmucosal (e.g., voccal, bladder, vaginal, uterine, rectal, nasal, mucosal). If delivery of the probe to the brain is desired, probe can be injected into an artery of the carotid system of arteries (e.g., occipital artery, auricular artery, temporal artery, cerebral artery, maxillary artery etc.).
  • The amount of the probe sufficient to provide the desired result(s) will depend on a variety of factors, such as the amount required to avoid undesirable physiological results. The precise dose to be employed can also depend on the route of administration, and should be decided according to the judgment of a medical practitioner and each subject's circumstances. In addition, known in vitro and in vivo assays may optionally be employed to help identify optimal dosage ranges. Effective doses may be extrapolated from dose-response curves derived from in vitro or in vivo test systems.
  • The probe can be administered in a variety of unit dosage forms, depending upon the particular cell or tissue being imaged, the general medical condition of each subject, the method of administration, and the like. Details on dosages are well described in the scientific literature. The exact amount and concentration of the imaging composition, or the “effective dose”, can be routinely determined (e.g., by a medical practitioner). The “dosing regimen” will depend upon a variety of factors, such as whether the cell or tissue to be imaged is disseminated or local, the general state of the subject's health, the subject's age, and the like. Using guidelines describing alternative dosing regimens, e.g., from the use of other imaging agents and compositions, the skilled artisan can readily determine by routine trials the optimal effective concentrations of the imaging composition.
  • In another example, the probe can be used as part of an intra-operative molecular imaging (IOI) procedure, such as disclosed in U.S. patent application Ser. No. 11/811,818 to Basilion, to detect abnormal EGFR expressing cells and/or diseased tissue. To identify and facilitate removal of abnormal EGFR expressing cells, for example, microscopic IOI techniques can be combined with topically applied probes described herein. Thus, the probe can be delivered via topical application, as opposed to intravenous injection, which facilitates localized diffusion of the imaging probes. In one example, the imaging probe can be combined with IOI to identify malignant cells that have infiltrated and/or are beginning to infiltrate at a tumor brain margin. The method can be performed in real-time during brain or other surgery. The method can include topical application of an imaging probe. An imaging modality can then be used to detect and subsequently gather image data. The resultant image data may be used to determine, at least in part, a radiological treatment. Alternatively, this image data may be used to control, at least in part, an automated surgical device (e.g., laser, scalpel, and micromachine) or to aid in manual guidance of surgery. Further, the image data may be used to plan and/or control the delivery of a therapeutic agent (e.g., by a micro-electronic machine or micro-machine).
  • In another aspect, the present application provides a method for treating a disease in a subject. One step of the method can include contacting the EGFR expressing cell with a diagnostically effective amount of the imaging probe. Upon contacting the imaging probe with the cell, one or more imaging modalities can be used to detect the reporter moiety to produce a molecular signature of the cell or differentiate the cell. Based on the molecular signature of the cell, the subject can then be appropriately treated for the particular disease by, for example, administering an effective amount of a therapeutic composition. An “effective amount” can refer to that amount of a therapeutic agent that results in amelioration of symptoms or a prolongation of survival in the subject and relieves, to some extent, one or more symptoms of the disease or returns to normal (either partially or completely) one or more physiological or biochemical parameters associated with or causative of the disease. Therapeutic agents can include any agent (e.g., molecule, drug, pharmaceutical composition, etc.) capable of preventing, inhibiting, or arresting the symptoms and/or progression of a disease.
  • In one example of the application, a molecular signal of a cell can indicate that a subject has a particular cancer. Because the type, severity, and stage of the cancer can be determined based on the molecular signature of the cell, the subject can then be administered an effective amount of a chemotherapy agent to treat the cancer. Chemotherapy agents are well known in the art and can include, for example, alkylating agents (e.g., cyclophosphamide, ifosfamide), antibiotics that affect nucleic acids (e.g., doxorubicin, bleomycin), platinum compounds (e.g., cisplatin), mitotic inhibitors (e.g., vincristine), antimetabolites (e.g., 5-fluorouracil), camptothecin derivatives (e.g., topotecan), biological response modifiers (e.g., interferon), and hormone therapies (e.g., tamoxifen).
  • Alternatively, the imaging probe can be used in a method to identify cells that are treatable by a particular therapeutic agent or therapeutic intervention. Additionally, the imaging probe can be used to the progress of therapy by monitoring a decreasing signal over time.
  • The following examples are for the purpose of illustration only and are not intended to limit the scope of the claims, which are appended hereto.
  • EXAMPLE 1
  • In this Example, we synthesized different versions of an imaging agent by varying the length of polyethylene glycol (PEG) linker between a 12-residue linear peptide sequence, GE11 (YHWYGYTPQNVI) (SEQ ID NO: 1) and a fluorochrome, Cy5.5. These imaging compounds were then tested in tissue culture cells lines expressing different levels of the EGFR and in orthotopic brain tumors generated from the cell lines. It was determined that the length of the linker critically affected the efficacy of the agent both in tissue culture and in the in vivo setting. Furthermore, these agents were capable of discriminating tumors expressing different levels of EGFR in orthotopic GBM models.
  • Materials and Methods Probe Synthesis and Analysis
  • The peptides were synthesized manually using protocols previously described. Peptide was labeled in solution with monoreactive Cy5.5 NHS ester (CyDye, GE Healthcare). Crude fluorophore-labeled peptides were purified by reversed phase high-performance liquid chromatography (RP-HPLC). The isolated peak was lyophilized and characterized matrix-assisted laser desorption/ionization (MALDI) mass spectrometry was done under positive mode. Compounds are characterized by RPHPLC under 2 different conditions and by thin layer chromatography with 3 different solvent conditions. Concentration of stock Cy5.5-labeled peptide solutions in dimethyl sulfoxide (DMSO) was determined by UV-Vis spectrometry [Cy5.5 molar extinction coefficient is 250,000 (mol/L)−1 cm−1 at 675 nm].
  • Cell Culture
  • Human glioblastoma astrocytoma, an epithelial-like cell line U87-MG, and human glioblastoma cell line stably over expressing the EGFRvIII-mutant form of the egfr gene, Gli36Δ5, were used in these studies. U87-MG and A431 cells were recently obtained from American Type Culture Collection. Gli36Δ5 cells were obtained from E.A. Chiocca and were authenticated by Research Animal Diagnostic Laboratory at the University of Missouri (Columbia, Mo.) for interspecies and mycoplasma contamination by PCR analysis. Cell lines were maintained in RPMI or Dulbecco's Modified Eagle's Medium (DMEM; Gibco), respectively, and supplemented with 10% FBS and 1% penicillin-streptomycin. The cells were incubated at 37° C. in a humidified 5% CO2 atmosphere. For Gli36Δ5 cells, puromycin was also included to maintain the expression of the EGFRvIII plasmid.
  • Saturation Binding Assays
  • To determine binding affinities of the Cy5.5-labeled peptides 1 to 4 to EGFR, a saturation binding assay protocol using labeled fluorescence ligand against cell surface receptors was adapted as previously reported. Briefly, Gli36Δ5 cells were plated 20,000 cells per well were plated on black Costar 96-well plates (cat. no. 3603). Probes were diluted to range of concentration (0-25 mmol/L) in growing media with 0.3% bovine serum albumin (BSA) and added into wells and incubated at 37° C. for 90 minutes at 5% CO2. Cells were then washed with PBS twice and dried. DMSO was added before reading fluorescence (excitation 670 nm, emission 700 nm) with a Tecan Infinite M200 plate reader. Saturation binding assay data were in quadruplicates and analyzed using one site binding classical equation for nonlinear regression analysis with the GraphPad Prism version 4.0.
  • Receptor Uptake Immunofluorescence
  • Gli36Δ5 or U87-MG cell lines plated in 96-well culture plates (20,000 cells per well; 3 wells per condition) were incubated over indicated time periods with 1 mmol/L of Cy5.5-labeled EGF peptides 2, 3, and 4 at 37° C. and 5% CO2. Cells were briefly rinsed with HEPES buffer and imaged with a Tecan Infinite M200 fluorescence plate reader (excitation 670 nm, emission 700 nm).
  • Immunocytochemistry of Cells Grown on Coverslips
  • Gli36Δ5 or U87-MG cancer cell lines were plated on coverslips and incubated overnight to promote adherence. The cells were then fixed with 4% paraformaldehyde, rinsed with PBS, and blocked with 1% host serum for 30 minutes at room temperature. Coverslips were incubated with primary antibody at room temperature for 2 hours. Antibodies used were mouse anti-human wild-type EGFR (1:100 dilution; clone DAK-H1-WT, Dako cat. no. M7289). The coverslips were then rinsed with PBS and counterstained with 40,6-diamidino-2-phenylindole (DAPI) for 10 minutes at room temperature to visualize the nuclei. After a final rinse with PBS, the coverslips were mounted using Fluor-Mount aqueous media, sealed with nail polish, and observed using epifluorescence microscopy.
  • Orthotopic Brain Tumor Model
  • NIH athymic nude female mice (5-8 weeks and 20-25 g upon arrival, NCI-NIH) were maintained at the Animal Resource Center at Case Western Reserve University (Cleveland, Ohio) according to institutional policies. Cell for brain implantation were harvested with 1 mL 0.05% trypsin-EDTA (Gibco) and briefly washed with PBS. Trypsin was inactivated by the addition of serum-containing media. The resulting cell suspension was centrifuged at 1,000 μg for 3 minutes. The cell suspension was centrifuged and the supernatant was removed after 2 washes in PBS. Finally, the cells were resuspended in 2 mL PBS for brain implants per animal (250,000 cells per animal for brain implants). Immediately following the cell harvesting procedure, animals were inoculated. For brain tumor implantation, mice were anesthetized by intraperitoneal (i.p.) injection of 50 mg/kg ketamine/xylazine and fitted into a stereotaxic rodent frame (David Kopf Instruments). A small incision was made just lateral to midline to expose bregma suture. A small (1.0 mm) burr hole was drilled at anterior (AP)=+1, lateral (ML)=−2.5 from the bregma. Glioblastoma cells were slowly deposited at a rate of 1 μL/min in the right striatum at a depth of 3 mm from dura with a 10-μL syringe (23-G needle). The needle was slowly withdrawn and the incision was closed with 2 to 3 sutures. Brain tumors grew for 10 to 12 days as per IACUC protocols at which point in vivo imaging studies were conducted. As control for the effects of surgical intervention, animals were also subjected to the implantation procedure but Optical Imaging of Tumors with EGFR Overexpression received 2 μL PBS (sham animals). Animals were fed exclusively on a special rodent diet (Harlan Laboratories, Inc.; Tekland 2018S) to reduce autofluorescence.
  • In Vivo and Ex Vivo Fluorescence Imaging
  • Mice bearing brain tumors derived from Gli36Δ5 or U87-MG cells were administered with compounds at 1 nmol/g via tail vein injection. In vivo competition assays were done with mixture of compound 2 and 10-fold concentration of nonlabeled probe 5. Before injection, mice were anesthetized with isoflurane and subjected to tomographic and spectral fluorescence imaging. One hour after injection, animals were re-imaged. Brains were then extracted and imaged. The excised brains were embedded in Tissue-Tek optimum cutting temperature for cryosections for immunohistochemistry (IHC). Fluorescence-mediated molecular tomography (FMT) images were obtained using FMT2500 (Perkin-Elmer), and 3-dimensional reconstructions of fluorescent signals were acquired using the accompanying software TrueQuant. Quantitative fluorescent signals for Cy5.5 of compound 2 were calibrated as per manufacturer's instructions using the 680-channel. Region of interest (ROI) assigned on the basis of the precise placement of cells during implantation at 3 to 4 mm into the brain. ROI was corroborated with fluorescent signals from ex vivo imaging. Fluorescent multispectral images were obtained using the Maestro In-Vivo Imaging System (CRi, Inc.). The yellow filter set appropriate for Cy5.5 was used for emission and excitation light. The tunable filter was automatically stepped in 10-nm increments whereas the camera captured images at a constant exposure of 200 ms. Fluorescence images were acquired before treatment, immediately after and 1.5 hours after treatment. To compare signal intensities, ROIs were selected over the tumor or nontumor areas, and the change in fluorescence signal over baseline was determined The spectral fluorescent images consisting of autofluorescence spectra and imaging probe were captured and unmixed on the basis of their spectral patterns. The total signal in the ROI defined in photons measured at the surface of the animal was divided by the area (in pixels). Spectral libraries were generated by assigning spectral peaks to background and fluorescence probe on tissue. The spectral libraries were manually computed using the Maestro software, with each tissue used as its own background control.
  • IHC of Cryosection of Brain Tissue
  • Sections (2 mm) of whole mouse brains implanted with Gli36Δ5 cells were fixed with 4% paraformaldehyde, cryosectioned onto microscope slides, rinsed with PBS, and blocked with 1% host serum for 30 minutes at room temperature. Sections were incubated with primary antibody at room temperature for 2 hours. Antibodies used were mouse anti-human wild-type EGFR (1:100 dilution; clone DAK-H1-WT, Dako cat. no. M7289). The coverslips were then rinsed with PBS and counterstained with DAPI for 10 minutes at room temperature to visualize the nuclei. After a final rinse with PBS, the slides were mounted with coverslips using Fluor-Mount aqueous media, sealed with nail polish, and observed using epifluorescence microscopy.
  • Western Blotting
  • Cell extracts (50 μg) were fractionated using SDS-PAGE and transferred onto a nitrocellulose membrane. Immunoblotting was done using a 1:500 dilution of an antibody against wild-type EGFR (DAKO, DAK-H1-WT) or a 1:500 dilution of a specific antibody against mutant EGFRvIII (Bioss Inc., cat #bs-2558R). Horseradish peroxidase-conjugated secondary antibodies against mouse IgG (Chemicon) or rabbit IgG (Amersham) were used. Bands were detected using an enhanced chemiluminescence detection system (Pierce).
  • Statistical Analysis
  • Analyses of data were achieved with GraphPad Prism version 4.00, GraphPad Software. Binding affinity was determined with nonlinear regression analysis with one site binding hyperbola with an equation: Y=Bmax×X (Kd+X), where Bmax is the maximal binding site and the Kd is the concentration required to reach half-maximal binding. ANOVA analysis at 95% confidence interval (CI) was used to compare treatments (*, P>0.05; **, P>0.01; confidence interval, ***, P>0.001). To compare live animal FMT, nonparametric one-way ANOVA analyses (Kruskal-Wallis tests) and the median differences were considered significant with P=0.0220.
  • Results
  • The goal of these studies was to develop a peptide based NIRF probe that would cross the BBTB and selectively bind to brain tumor cells over expressing EGFR. For these studies, we used a peptide discovered through phage display screening against purified human EGFR. The peptide was modified to include linkers and a NIRF dye. To determine the optimal space between the NIRF dye and the peptide, we designed and synthesized a series of peptides to include increasing numbers of discrete ethylene glycol units to serve as linkers between a Cy5.5 and the N-terminal end of the peptide. Cy5.5 and EGFpep were either directly linked or linked via 1, 2, or 3 units of discrete ethylene glycol (AEEA) moieties (Table 1). To determine which of the compounds optimally interacts with cells expressing EGFR, the apparent binding for each bioconjugate was fluorometrically determined from a saturation binding assay in vitro using a human GBM cell line overexpressing EGFR and Gli36Δ5 (Table 1). Compounds 1, 2, 3, and 4 all bound to the cells with affinities in the micromolar range. Compound 2, which had one linker, had the highest apparent affinity with a Kd at least 2-fold better than compound 1, which had no ethylene linker, 8.9 to 18.5 μmol/L respectively. Compounds 3 and 4 have weaker affinities with Kd of 64.4 and 123.0 μmol/L, respectively.
  • TABLE 1
    Table 1. The sequences of the peptides studied and the corresponding
    apparent binding affinities (Kd) from saturation binding assays
    Compound Peptide (EGFpep) Kd, μmol/L
    1 Cy5.5-YHWYGYTPQNVI-amide 18.5 ± 3.9 
    2 Cy5.5-(AEEA)1-YHWYGYTPQNVI-amide 8.9 ± 3.7
    3 Cy5.5-(AEEA)2-YHWYGYTPQNVI-amide 64.4 ± 24.6
    4 Cy5.5-(AEEA)3-YHWYGYTPQNVI-amide 123.0 ± 174.0
    5 YHWYGYTPQNVI-amide (GE11-amide) ND
    6 Cy5.5-(AEEA)1-NYQTPVYGWIYH-amide scrambled ND
    Abbreviation: ND, not determined
  • We next used immunofluorescence microscopy to determine the fate of the peptide complexes once they bound to glioblastoma cells expressing EGFR. As predicted from the affinity measurements compound 2, which had the highest affinity, also showed the greatest accumulation of fluorescence after incubation with Gli36Δ5 cells (FIG. 2). Neither compound 1 (no linker) nor either of the molecules with greater linker numbers (compounds 3 and 4) was taken up by the cells to the same extent as compound 2. Interestingly, the peptide with the longest linker and the worst binding affinity, compound 4, was taken up by cells better than compound 3 (FIG. 3A). When tested against U87-MG cells, which express much lower levels of the EGFR, no cellular uptake for any of the compounds was observed (FIG. 3B).
  • We next tested the ability of these compounds to target EGFR-expressing tumors implanted within the brains of mice. For these studies, mice were orthotopically implanted with Gli36Δ5 cells. Approximately 10 days after implantation, the animals were administered 1 nmol/g via tail vein injection and sacrificed 1 hour later. Brains were harvested and imaged ex vivo for accumulation of the imaging probe. As a control for specificity, we synthesized a Cy5.5-labeled scrambled peptide, compound 6, using the amino acid residues of the parent peptide (compound 5) in random order. Compound 2 targeted the tumor efficiently, accumulating 1.1% of injected dose (FIG. 3A, left graph). In contrast, compound 6 did not target the tumor with delivery of only 0.006% of the injected dose. To further assess specificity, animals that bore Gli36Δ5 brain tumors were administered compound 2 alone or in the presence of a 10-fold excess of unlabeled peptide (compound 5). In animals that received only compound 2, there was significant tumor-associated fluorescence. In contrast, when a 10-fold excess of competitor peptide was coadministered with compound 2, there was approximately a 60% decrease in accumulation of the probe, (FIG. 4A, right graph). Imaging and quantification of identical ROIs taken on the contralateral brain showed little uptake of the probe (data not shown).
  • To show that the uptake was associated with human EGFR expression on the Gli36Δ5 cells, the resected brains were fixed and subjected to IHC with monoclonal antibodies specific for human EGFR (FIG. 5B). These results have shown that only cells that expressed human EGFR were associated with Cy5.5 fluorescence. Notably, no Cy5.5 signal was associated with surrounding mouse brain.
  • Our final test for compound 2 was to determine whether it would be useful for noninvasive in vivo detection and discrimination of tumors differentially expressing EGFR. For these studies, mice were orthotopically implanted with either Gli36Δ5 cells, which express high EGFR, or U87-MG cells, which express relatively low EGFR levels (FIG. 6A). Following implantation, the tumors were allowed to grow approximately 10 to 12 days and then mice were administered 1 nmol/g of compound 2 via tail vein injection. One hour after injection, the mice were anesthetized and the intensity of Cy5.5 fluorescence from the tumor was noninvasively quantified using FMT (FIG. 7). The tumors formed from Gli36D5 cells had approximately 4-fold more fluorescence than either tumors formed with U87-MG cells or control sham surgeries (FIG. 8A). For Gli36Δ5 and U87-MG brain tumors, these fluorescence signals corresponded to injected doses of 1.1% and 0.27%, respectively. Statistical analyses showed that the median fluorescence signals between the 2 groups were significantly different (FIG. 8A). To corroborate these data, tumors were excised and subjected to ex vivo FMT and Maestro fluorescence imaging analyses (FIGS. 8B and 8C). These measurements were in good agreement with those measurements made during the live animal imaging.
  • We also examined the expression of EGFRvIII in the cell lines. Western blots for the EGFRvIII on both U87-MG and Gli36D5 cells showed that the mutant receptor expression is similar for both cell lines (FIG. 6B). In addition, we examined A431 cells, a squamous carcinoma cell line, that expresses high levels of wild-type EGFR. Saturation binding studies indicated that the Kd for binding was similar to that measured with Gli36Δ5 cells. Furthermore, incubation of compound 2 with A431 cells displayed increasing fluorescence in the presence of the EGF ligand, which increases cycling of wild-type EGFR in cells.
  • We surmised that conjugation of the GE11 peptide via its amine resulted in lower serum stability as well as poor in vivo targeting and drug delivery due in part to the free carboxylate at the C-terminus. We aimed to develop compound 5 into a fluorescence imaging probe for noninvasive detection of tumors overexpressing EGFR and, therefore, incorporated lessons from prior studies into our probe design by conjugating the peptide via its free carboxyl-terminal end.
  • EXAMPLE 2 Cyclic Analogues of EGF Peptide Ligands
  • Peptides have conformational structures based on their sequences. In the EGF peptide ligand, Tyr1-His2-Trp3-Tyr4-Gly5-Tyr6-Thr7-Pro8-Gln9-Asn10-Val11-Ile12 sequence contains a turn at the Thr-Pro for binding to EGF receptors. Since linear peptides are flexible, we designed and synthesized a peptide that will stabilize that conformational turn. Gly5 and Asn10 sites were replaced with Lys and Glu, respectively, to form a side chain to side chain lactam cyclic bridge. Cy5 NIR label and miniPEG linker were also added as shown below.
  • Figure US20130116404A1-20130509-C00009
  • Cy5.5-miniPEG-YHWY-cyclo[KYTPQE]VI-amide.
  • The cyclic peptide was generated using Fmoc chemistry on solid support on a Rink amide resin. Fmoc-Glu(OAllyl)-OH and Fmoc-Lys(Alloc)-OH were used for the lactam bridge cyclization on resin. After linear sequence was coupled, the side chain groups OAllyl and Alloc were removed using palladium catalysts and the subsequent ring closure was achieved by HCTU coupling. Standard cleavage procedures were subsequently used to isolate the pure peptide. Cy5.5 conjugation was also a standard procedure.
  • From the above description, those skilled in the art will perceive improvements, changes and modifications. Such improvements, changes and modifications within the skill of the art are intended to be covered by the appended claims. All references, publications, and patents cited in the present application are herein incorporated by reference in their entirety.

Claims (20)

Having described the invention, we claim:
1. A probe for imaging EGFR expressing cells, comprising:
an EGFR targeting moiety,
a reporter moiety, and
a hydrophilic linker that links the EGFR targeting moiety to the reporter moiety, the hydrophilic linker enhancing solubility of the probe in an aqueous media as well as binding affinity of the probe to EGFR expressing cells.
2. The probe of claim 1, EGFR targeting moiety comprising a peptide having SEQ ID NO: 1 or SEQ ID NO: 2.
3. The probe of claim 1, the reporter moiety comprising a fluorochrome.
4. The probe of claim 1, the linker comprising a polyethylene glycol linker.
5. The probe of claim 1, upon systemic administration to subject being capable of crossing the blood brain barrier.
6. The probe of claim 1, the EGFR expressing cells comprising EGFR expressing cancer cells.
7. The probe of claim 1, comprising the following formula:
Figure US20130116404A1-20130509-C00010
where R is a near infrared fluorescence moiety and n is an integer from 1-3.
8. A probe for imaging EGFR expressing cells, comprising:
a hydrophobic EGFR targeting moiety,
a reporter moiety, and
a hydrophilic linker that links the hydrophobic EGFR targeting moiety to the reporter moiety, the hydrophilic linker enhancing solubility of the probe in an aqueous media as well as binding affinity of the probe to EGFR expressing cells.
9. The probe of claim 8, EGFR targeting moiety comprising a peptide having SEQ ID NO: 1 or SEQ ID NO: 2.
10. The probe of claim 9, the reporter moiety comprising a fluorochrome.
11. The probe of claim 9, the linker comprising a polyethylene glycol linker.
12. The probe of claim 9, upon systemic administration to subject being capable of crossing the blood brain barrier.
13. The probe of claim 9, the EGFR expressing cells comprising EGFR expressing cancer cells.
14. The probe of claim 8, comprising the following formula:
Figure US20130116404A1-20130509-C00011
where R is a near infrared fluorescence moiety and n is an integer from 1-3.
15. A probe for imaging EGFR expressing cells, comprising:
a hydrophobic EGFR targeting moiety that includes a peptide having SEQ ID NO: 1 or SEQ ID NO: 2,
a fluorochrome reporter moiety, and
a hydrophilic linker that links the hydrophobic EGFR targeting moiety to the reporter moiety, the hydrophilic linker enhancing solubility of the probe in an aqueous media as well as binding affinity of the probe to EGFR expressing cells.
16. The probe of claim 15, the linker comprising a polyethylene glycol linker.
17. The probe of claim 15, upon systemic administration to subject being capable of crossing the blood brain barrier.
18. The probe of claim 15, the EGFR expressing cells comprising EGFR expressing cancer cells.
19. The probe of claim 15, comprising the following formula:
Figure US20130116404A1-20130509-C00012
where R is a near infrared fluorescence moiety and n is an integer from 1-3.
20. The probe of claim 15, selected from the group consisting of:
Figure US20130116404A1-20130509-C00013
where n is an integer from 1 to 3.
US13/671,957 2011-11-08 2012-11-08 Targeted non-invasive imaging probes of egfr expressing cells Abandoned US20130116404A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/671,957 US20130116404A1 (en) 2011-11-08 2012-11-08 Targeted non-invasive imaging probes of egfr expressing cells
US16/396,265 US20190315805A1 (en) 2011-11-08 2019-04-26 Targeted noninvasive imaging probes of egfr expressing cells
US17/224,505 US11708393B2 (en) 2011-11-08 2021-04-07 Targeted non-invasive imaging probes of EGFR expressing cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161557025P 2011-11-08 2011-11-08
US13/671,957 US20130116404A1 (en) 2011-11-08 2012-11-08 Targeted non-invasive imaging probes of egfr expressing cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/396,265 Continuation US20190315805A1 (en) 2011-11-08 2019-04-26 Targeted noninvasive imaging probes of egfr expressing cells

Publications (1)

Publication Number Publication Date
US20130116404A1 true US20130116404A1 (en) 2013-05-09

Family

ID=48224120

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/671,957 Abandoned US20130116404A1 (en) 2011-11-08 2012-11-08 Targeted non-invasive imaging probes of egfr expressing cells
US16/396,265 Abandoned US20190315805A1 (en) 2011-11-08 2019-04-26 Targeted noninvasive imaging probes of egfr expressing cells
US17/224,505 Active 2033-07-18 US11708393B2 (en) 2011-11-08 2021-04-07 Targeted non-invasive imaging probes of EGFR expressing cells

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/396,265 Abandoned US20190315805A1 (en) 2011-11-08 2019-04-26 Targeted noninvasive imaging probes of egfr expressing cells
US17/224,505 Active 2033-07-18 US11708393B2 (en) 2011-11-08 2021-04-07 Targeted non-invasive imaging probes of EGFR expressing cells

Country Status (1)

Country Link
US (3) US20130116404A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7038078B2 (en) * 2000-11-27 2006-05-02 University Of Marlyland, Baltimore Methods of synthesizing and using derivatives of [2-(2-aminoethoxy)ethoxy] acetic acid
US20100048602A1 (en) * 2007-03-12 2010-02-25 Nektar Therapeutics Oligomer-Opioid Agonist Conjugates
US20100260686A1 (en) * 2009-04-09 2010-10-14 Washington, University Of Nanoparticles for brain tumor imaging
US20100297120A1 (en) * 2007-05-29 2010-11-25 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US20100329983A1 (en) * 2009-06-26 2010-12-30 Soricimed Biopharma Inc. Compounds and methods for the detection of trpv-6 cancers and drug delivery
US20110165079A1 (en) * 2009-12-30 2011-07-07 Lu Maggie J M Peptide for transmigration across blood brain barrier and delivery systems comprising the same

Family Cites Families (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE383251B (en) 1973-04-26 1976-03-08 K B K A Lundberg CABINET DEVICE INCLUDING ATMINSTONE A EXTENSIBLE PART
US6528499B1 (en) 2000-04-27 2003-03-04 Georgetown University Ligands for metabotropic glutamate receptors and inhibitors of NAALADase
US6479470B1 (en) 1999-04-28 2002-11-12 Georgetown University Ligands for metabotropic glutamate receptors and inhibitors of NAALAdase
US7408079B2 (en) 2002-01-10 2008-08-05 The Johns Hopkins University Imaging agents and methods of imaging NAALADase or PSMA
US20060222595A1 (en) 2005-03-31 2006-10-05 Priyabrata Mukherjee Nanoparticles for therapeutic and diagnostic applications
WO2006133271A2 (en) 2005-06-06 2006-12-14 The General Hospital Corporation Compositions and methods relating to target-specific photodynamic therapy
AU2006262231B2 (en) 2005-06-20 2013-01-24 Psma Development Company, Llc PSMA antibody-drug conjugates
US20090011004A1 (en) 2005-12-30 2009-01-08 Philadelphia Health & Education Corp., D/B/A/ Drexel University Of College Of Medicine Improved carriers for delivery of nucleic acid agents to cells and tissues
US8078264B2 (en) 2006-07-11 2011-12-13 Case Western Reserve University Intra-operative molecular imaging
SI2502938T1 (en) 2006-10-27 2015-05-29 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
WO2008073856A2 (en) 2006-12-08 2008-06-19 Massachusetts Institute Of Technology Delivery of nanoparticles and/or agents to cells
US9271653B2 (en) 2007-06-12 2016-03-01 Case Western Reserve University Intra-operative molecular imaging
WO2009045579A2 (en) 2007-06-14 2009-04-09 The Regents Of The University Of California Multimodal imaging probes for in vivo targeted and non-targeted imaging and therapeutics
EP2921482B1 (en) 2007-06-26 2018-09-05 The Johns Hopkins University Labeled inhibitors of prostate-specific membrane antigen (psma), biological evaluation, and use as imaging agents
JP2010536790A (en) 2007-08-17 2010-12-02 パーデュー・リサーチ・ファウンデーション PSMA binding ligand-linker conjugate and method of use
WO2009070302A1 (en) 2007-11-30 2009-06-04 The Johns Hopkins University Prostate specific membrane antigen (psma) targeted nanoparticles for therapy of prostate cancer
US20090297615A1 (en) 2008-05-27 2009-12-03 The Chinese University Of Hong Kong Nanoparticles, methods of making same and cell labeling using same
KR101014456B1 (en) 2008-07-31 2011-02-14 기아자동차주식회사 Tilting Apparatus for Seat Cushion in Vehicle
HUE059436T2 (en) 2008-08-01 2022-11-28 Univ Johns Hopkins Psma-binding agents and uses thereof
GB0814960D0 (en) 2008-08-15 2008-09-24 Ge Healthcare As Method for detecting dysplasia
US10717750B2 (en) 2009-03-19 2020-07-21 The Johns Hopkins University 68Ga-labeled NOTA-chelated PSMA-targeted imaging and therapeutic agents
HUE059796T2 (en) 2009-03-19 2022-12-28 Univ Johns Hopkins Psma-targeting compounds and uses thereof
CN102791827B (en) 2009-11-09 2016-11-16 华盛顿大学商业化中心 Functionalization chromophoric polymers point and bioconjugate body thereof
KR101510413B1 (en) 2010-02-08 2015-04-08 어젠시스 인코포레이티드 Antibody drug conjugates (adc) that bind to 161p2f10b proteins
US9951324B2 (en) 2010-02-25 2018-04-24 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
EP2555802A1 (en) 2010-04-08 2013-02-13 Sanford-Burnham Medical Research Institute Methods and compositions for enhanced delivery of compounds
US20130289520A1 (en) 2010-04-23 2013-10-31 Children's Hospital Boston Targeted and light-activated cytosolic drug delivery
BR112012029866A2 (en) 2010-06-03 2017-03-07 Genentech Inc method for determining the presence of a steap-1 protein
SG189034A1 (en) 2010-09-24 2013-05-31 Agency Science Tech & Res A nanoprobe comprising gold colloid nanoparticles for multimodality optical imaging of cancer and targeted drug delivery for cancer
PT2621526T (en) 2010-09-29 2018-08-02 Seattle Genetics Inc Antibody drug conjugates (adc) that bind to 191p4d12 proteins
WO2012135592A2 (en) 2011-03-31 2012-10-04 The Johns Hopkins University Theranostic imaging agents and methods of use
US9517277B2 (en) 2011-07-15 2016-12-13 University Of Georgia Research Foundation, Inc. Immune-stimulating photoactive hybrid nanoparticles
WO2013078091A1 (en) 2011-11-22 2013-05-30 The Johns Hopkins University Collagen mimetic peptides for targeting collagen strands for in vitro and in vivo imaging and therapeutic use
CN113149921A (en) 2011-11-30 2021-07-23 约翰霍普金斯大学 Homogenic and xenogenic multivalent inhibitors of Prostate Specific Membrane Antigen (PSMA) and uses thereof
CN112587658A (en) 2012-07-18 2021-04-02 博笛生物科技有限公司 Targeted immunotherapy for cancer
WO2014127365A1 (en) 2013-02-15 2014-08-21 Case Western Reserve University Psma ligands and uses thereof
US10207005B2 (en) 2013-02-15 2019-02-19 Case Western Reserve University Photodynamic therapy composition
JP2016519070A (en) 2013-03-15 2016-06-30 アッヴィ・インコーポレイテッド Purification of antibody-drug conjugate (ADC)
SI2968588T1 (en) 2013-03-15 2019-05-31 AbbVie Deutschland GmbH & Co. KG Anti-egfr antibody drug conjugate formulations
US10188754B2 (en) 2013-05-16 2019-01-29 The Johns Hopkins University Compositions and methods for chemical exchange saturation transfer (CEST) based magnetic resonance imaging (MRI)
JP6510518B2 (en) 2013-08-01 2019-05-08 アジェンシス,インコーポレイテッド Antibody-drug conjugate (ADC) that binds to the CD37 protein
US10232058B2 (en) 2013-10-14 2019-03-19 The Johns Hopkins University Prostate-specific membrane antigen-targeted photosensitizers for photodynamic therapy
US10548985B2 (en) 2014-01-10 2020-02-04 Birdie Biopharmaceuticals, Inc. Compounds and compositions for treating EGFR expressing tumors
EP3536345A1 (en) 2014-05-06 2019-09-11 The Johns Hopkins University Metal/radiometal-labeled psma inhibitors for psma-targeted imaging and radiotherapy
WO2016040683A1 (en) 2014-09-12 2016-03-17 The Regents Of The University Of California Macropinocytosing human anti-cd46 antibodies and targeted cancer therapeutics
US10736974B2 (en) 2014-10-22 2020-08-11 The Johns Hopkins University Scaffolds and multifunctional intermediates for imaging PSMA and cancer therapy
SG11201703446RA (en) 2014-10-31 2017-05-30 Abbvie Biotherapeutics Inc Anti-cs1 antibodies and antibody drug conjugates
US10596259B2 (en) 2015-05-20 2020-03-24 The Regents Of The University Of California Tumor radiosensitization with monomethyl auristatin E (MMAE) and derivatives thereof
PL412787A1 (en) 2015-06-22 2017-01-02 Magdalena Król Macrophage-based aimed system for delivery of ferritin related compounds
US10688198B2 (en) 2015-09-30 2020-06-23 The Johns Hopkins University Salicylic acid-based polymeric CEST contrast agents targeting prostate-specific membrane antigen and uses thereof
US10722593B2 (en) 2015-10-02 2020-07-28 Sirenas Llc Anti-cancer compounds and conjugates thereof
WO2017095808A1 (en) 2015-11-30 2017-06-08 Abbvie Inc. ANTI-huLRRC15 ANTIBODY DRUG CONJUGATES AND METHODS FOR THEIR USE
TW201836647A (en) 2017-04-06 2018-10-16 美商艾伯維有限公司 Anti-prlr antibody-drug conjugates (adc) and uses thereof
CN110831976A (en) 2017-05-23 2020-02-21 斯索恩生物制药有限公司 Dual conjugation process for preparing antibody-drug conjugates
LT3691692T (en) 2017-10-14 2021-05-10 Abbvie Inc. Anti-cd71 activatable antibody drug conjugates and methods of use thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7038078B2 (en) * 2000-11-27 2006-05-02 University Of Marlyland, Baltimore Methods of synthesizing and using derivatives of [2-(2-aminoethoxy)ethoxy] acetic acid
US20100048602A1 (en) * 2007-03-12 2010-02-25 Nektar Therapeutics Oligomer-Opioid Agonist Conjugates
US20100297120A1 (en) * 2007-05-29 2010-11-25 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US20100260686A1 (en) * 2009-04-09 2010-10-14 Washington, University Of Nanoparticles for brain tumor imaging
US20100329983A1 (en) * 2009-06-26 2010-12-30 Soricimed Biopharma Inc. Compounds and methods for the detection of trpv-6 cancers and drug delivery
US20110165079A1 (en) * 2009-12-30 2011-07-07 Lu Maggie J M Peptide for transmigration across blood brain barrier and delivery systems comprising the same

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Agnes et al., An Optical Probe for Noninvasive Molecular Imaging of Orthotopic Brain Tumors Overexpressing Epidermal Growth Factor Receptor, Mol Cancer Ther, Vol 11(10): OF1-OF10, (published online July 17, 2012). *
Bennike (Development of a Novel Peptide Based Probe for Tumour Imaging, Aalborg University, Thesis (June 2011), 92 pages *
DeJesus, Synthesis of [64Cu]Cu-NOTA-Bn-GE11 for PET Imaging of EGFR-Rich Tumors, Current Radiopharmaceuticals, Vol 5(1):15-18 (Jan 2012). *
Li et al., FASEB Journal, Vol. 19:1978-1985 (Dec. 2005) *
Song et al. (Peptide Ligand-mediated liposome distribution and targeting to EGFR expressing tumor in vivo, International Journal of Pharmaceutics, vol 363:155-161 (pub online July 23, 2008). *

Also Published As

Publication number Publication date
US11708393B2 (en) 2023-07-25
US20210221846A1 (en) 2021-07-22
US20190315805A1 (en) 2019-10-17

Similar Documents

Publication Publication Date Title
AU2016318949B2 (en) PSMA-targeted NIR dyes and their uses
Joshi et al. Targeted optical imaging agents in cancer: focus on clinical applications
EP2968335B1 (en) Synthesis and composition of amino acid linking groups conjugated to compounds used for the targeted imaging of tumors
CN110199195B (en) PSMA-targeted NIR dyes and uses thereof
EP3193945B1 (en) Upar targeting peptide for use in peroperative optical imaging of invasive cancer
US10682427B2 (en) Intra-operative imaging
US20210275694A1 (en) Synthesis and composition of amino acid linking groups conjugated to compounds used for the targeted imaging of tumors
CN110869057A (en) Ultra-small nanoparticles labeled with zirconium-89 and methods thereof
US20190175745A1 (en) Photodynamic therapy composition
US11484607B2 (en) PSMA-targeted NIR dyes and their uses
US20180140703A1 (en) Tumor radiosensitization with antibody conjugates
US11708393B2 (en) Targeted non-invasive imaging probes of EGFR expressing cells
US10413621B2 (en) Composition and methods for imaging cells
US20170340698A1 (en) Cyclic peptide specifically binding to apoptotic cells and use thereof
JP2017510547A (en) Methods and pharmaceutical compositions for the treatment of hepatocellular carcinoma
Olive Preparation of near-infrared-labeled targeted contrast agents for clinical translation
KR20100104370A (en) Quantum dot based-tracer for multimodal imaging of epidermal growth factor receptors
Chen et al. Detection of Barrett's Neoplasia with Near-Infrared Fluorescent Heterodimeric Peptide: Feasibility Results from a Phase 1 Study

Legal Events

Date Code Title Description
AS Assignment

Owner name: CASE WESTERN RESERVE UNIVERSITY, OHIO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BASILION, JAMES P.;AGNES, RICHARD;SIGNING DATES FROM 20130115 TO 20130116;REEL/FRAME:029748/0902

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION