US20110097742A1 - Contrast agents, methods for preparing contrast agents, and methods of imaging - Google Patents

Contrast agents, methods for preparing contrast agents, and methods of imaging Download PDF

Info

Publication number
US20110097742A1
US20110097742A1 US12/935,413 US93541309A US2011097742A1 US 20110097742 A1 US20110097742 A1 US 20110097742A1 US 93541309 A US93541309 A US 93541309A US 2011097742 A1 US2011097742 A1 US 2011097742A1
Authority
US
United States
Prior art keywords
contrast agent
protein
metal ion
agent
metal
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
US12/935,413
Inventor
Jenny Jie Yang
Zhiren Liu
Shunyi Li
Yubin Zhou
Jie Jiang
Shenghui Xue
Jinjuan Qiao
Lixia Wei
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.)
Georgia State University Research Foundation Inc
Original Assignee
Georgia State University Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Georgia State University Research Foundation Inc filed Critical Georgia State University Research Foundation Inc
Priority to US12/935,413 priority Critical patent/US20110097742A1/en
Assigned to GEORGIA STATE UNIVERSITY RESEARCH FOUNDATION reassignment GEORGIA STATE UNIVERSITY RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZHOU, YUBIN, WEI, LIXIA, JIANG, JIE, QIAO, JIN-JUAN, XUE, SHENGHUI, LI, SHUNYI, LIU, ZHIREN, YANG, Jenny Jie
Publication of US20110097742A1 publication Critical patent/US20110097742A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • A61K49/14Peptides, e.g. proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • A61K49/12Macromolecular compounds
    • A61K49/126Linear polymers, e.g. dextran, inulin, PEG
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • A61K49/14Peptides, e.g. proteins
    • A61K49/143Peptides, e.g. proteins the protein being an albumin, e.g. HSA, BSA, ovalbumin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Magnetic resonance imaging is a non-invasive technique providing high resolution, three-dimensional images of morphological features as well as functional and physiological information about tissues in vivo. It is capable of detecting abnormalities in deep tissues and allows for whole body imaging. It has emerged as a primary diagnostic imaging technique for human diseases.
  • Exogenous MRI contrast agents are often used to enhance the contrast between pathological and normal tissues by altering the longitudinal and transverse (i.e., T 1 and T 2 ) relaxation times of water protons.
  • the relaxivity (unit capability of the agent to change the relaxation time) of a contrast agent is dependent on several factors including the number of water molecules in the coordination shell, the exchange rate of the coordinated water with the bulk water, and the rotational correlation time ⁇ R of the contrast agent.
  • the MRI contrast agent can have: 1) high relaxivity for high contrast-to-noise ratio (CNR) and dose efficiency, 2) thermodynamic stability, especially metal selectivity for the target ions over excess physiological metal ions, to minimize the release of toxic paramagnetic metal ions, 3) adequate vascular, tissue retention time to allow imaging, and 4) proper excretion from the body.
  • CNR contrast-to-noise ratio
  • thermodynamic stability especially metal selectivity for the target ions over excess physiological metal ions, to minimize the release of toxic paramagnetic metal ions
  • 3) adequate vascular, tissue retention time to allow imaging and 4) proper excretion from the body.
  • One exemplary contrast agent includes: a) a scaffold protein, and b) at least one metal ion chelating site, wherein the scaffold protein includes at least one metal ion chelating site that is already present or is integrated into the scaffold protein, wherein the scaffold protein includes a metal ion bound to a metal ion chelating site, wherein the contrast agent is stable in a physiological environment.
  • One exemplary method of imaging a sample includes: administering at least one of the contrast agent described herein to the sample; introducing the sample to an imaging system; and imaging the sample.
  • One exemplary method for preparing a contrast agent includes: a) selecting a scaffold protein; b) constructing at least one metal ion chelating site; c) operatively embedding the metal ion binding site into the protein, wherein the metal ion has contrast agent properties, and d) attaching at least one polyethylene glycol (PEG) to the scaffold protein.
  • PEG polyethylene glycol
  • FIG. 1.1 Schematic descriptions of different classes of MRI contrast agents and simulation of T 1 relaxivity.
  • FIG. 1.1( a ) Different constructs of MRI contrast agents.
  • ⁇ Rs slow ⁇ R due to its internal mobility;
  • Schematic description of the design of reported MRI contrast agents by directly coordinating Gd 3+ ions to ligand residues on a rigid protein frame to eliminate the high internal mobility.
  • FIG. 1.1( b ) Simulated T 1 relaxivity at the given rotational correlation time ⁇ R (100 ps, below, or 10 ns, above), water dwelling time ⁇ m , correlation time of splitting ⁇ v (1 and 10 ps, solid and dashed lines, respectively), and mean square zero field splitting energy ⁇ 2 (10 18 s ⁇ 2 ).
  • the ⁇ m valves are 10 ⁇ 10 ( ⁇ ), 10 ⁇ 9 ( ⁇ ), and 10 ⁇ 8 s ( ⁇ ) for 100 ps ⁇ R and 10 ⁇ 9 ( ⁇ ), 10 ⁇ 8 ( ⁇ ), and 10 ⁇ 7 s ( ⁇ ) for 10 ns ⁇ R according to the theory developed by Blombergen, Solomon (refs 6 and 7 in Example 1).
  • the water coordination number, q is assumed to be 1 and the agent concentration is 0.001 M. See on-line supporting materials for r1 and r2 simulations.
  • FIG. 1.1( c ) Modeled structure of designed Gd 3+ -CA1.CD2 based on the designed NMR structure 1T6W (ref 31 in Example 1). Ligand residues E15, E56, D58, D62 and D64 at the B, E, and D ⁇ -strands of the host protein CD2 are shown in red.
  • FIG. 1.2 Comparison of in vitro relaxivity between DTPA and designed contrast agents.
  • FIG. 1.2( a ) MR images produced using Spin-echo sequence, TR 6000 ms, TI 960 ms, TE 7.6 ms at 3T.
  • Samples are 1) dH 2 O, 2) 10 mM Tris-HCl pH 7.4, 3) 0.10 mM Gd-DTPA in H 2 O, 4) 0.10 mM Gd-DTPA in 10 mM Tris-HCl pH 7.4, 5) 0.10 mM Gd 3+ and CD2, 6) 0.077 mM Gd-CA4.CD2, 7) 0.050 mM Gd-CA2.CD2, 8) 0.10 mM Gd-CA9.CD2, 9) 0.020 mM Gd-CA1.CD2, and 10) 0.050 mM Gd-CA1.CD2.
  • FIG. 1.2( b ) Proton relaxivity values of Gd-CA1.CD2 (r 1 , solid black; r 2 , cross) and Gd-DPTA (r 1 , shield; r 2 , open) at indicated field strength were measured.
  • FIG. 1.2( c ) In vitro relaxivity of contrast agents Gd-DPTA (DTPA), Gd-CA1.CD2 (CA1) and Gd-CA2.CD2 (CA2) in the absence of Ca 2+ (black and grey), presence of 1 mM Ca 2+ (left strip and open) and 10 mM Ca 2+ (right strip and cross) at 3T.
  • T 1 (black, left & right strips) and T 2 (grey, open and cross) were determined using a Siemens whole-body MR system.
  • FIG. 1.3 Dynamic properties and hydration water number of designed contrast agents.
  • FIG. 1.3( a ) S 2 order values of the engineered metal binding protein. The positions of ligand residues are shown in vertical bars. Order factors of CA2-CD2 with discontinuous ligand residues have the same dynamic properties as the scaffold protein. Arrows indicate the position of ligand residues.
  • FIG. 1.3( b ) Measurement of coordination water number by monitoring Tb 3+ life time. Luminescence decay lifetime was obtained by fitting the acquired data in both H 2 O and D 2 O with a mono-exponential decay function.
  • FIG. 1.4 In vivo MR images and biodistribution of designed contrast agents.
  • FIG. 1.4( b ) The MRI signal intensity changes at kidney ( ⁇ ), liver ( ⁇ ), and muscle ( ⁇ ) as a function of time. The 0 refers to the pre-injection.
  • FIG. 1.4( a ) MR images of mouse (26 g) pre (left) and 40 minutes post (right) the injection of 50 ⁇ L, of 1.2 mM Gd-CA1.CD2 through the tail vein. The MRI was performed using a spin echo sequence with
  • FIG. 1.5 The ESI-TOF MS spectrum of Gd-CA1.CD2.
  • the ESI-TOF MS spectra of the complex were recorded by Q-TOF Micro Mass Spectrometer (Micromass).
  • FIG. 1.6 Measurement of metal binding constants. Gd 3+ binding affinity ( FIG. 1.6 a ) and Zn 2+ binding affinity ( FIG. 1.6 b ) of CA1.CD2 measured by dye competition assays. CA1.CD2 stock solution was gradually added into the 1:1 dye-metal complex to compete for the dye-bound metal ions. The insets show the titration curve for the dye indicators fluo 5N ( FIG. 1.6 a ) and FluoZin ( FIG. 1.6 b ), respectively.
  • FIG. 1.6 c La 3+ binding affinity obtained by Aromatic residue sensitized Tb 3+ luminescence energy transfer. The Tb 3+ fluorescence of a protein-Tb 3+ mixture decreases with the addition of La 3+ .
  • the La 3+ concentrations are 0, 0.30, 0.76, 5.95, 11.03, and 28.95 ⁇ M from top to bottom.
  • the Tb 3+ fluorescence decrease competition was fitted (line) by a normal competition plus a nonspecific quenching effect (inset).
  • FIG. 1.7 MRI imaging of CA1.CD2 at 9.4 T.
  • FIG. 1.7( a ) MR images of CD-1 mouse (26 g) (four mice were imaged) at 9.4T field using Gd-CA1.CD2 as a T 2 -weighted contrast agent. The images were recorded pre-(i) and 2 hours post-(ii & iii) injection of 50 ⁇ l of 1.2 mM Gd-CA1.CD2 agent through the tail vein. MR images were recorded using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence. The in-plane resolution is 0.2 ⁇ 0.3 ⁇ 1.0 mm.
  • CPMG Carr-Purcell-Meiboom-Gill
  • the MRI intensity in muscle at 10 minutes post contrast administration was defined as 1.
  • MRI intensities at other tissue sites were normalized to the muscle intensity.
  • FIG. 1.8 Detection of CA1.CD2 in serum and cytotoxicity of CA1.CD2.
  • FIG. 1.8( a ) Sandwich ELISA detection of CA1.CD2 in mouse blood using OX45 and PabCD2. PabCD2 was used as the capture antibody and OX45 was used as the detection antibody in the sandwich-ELISA experiments. Serum samples were obtained from test mice at 0, 0.5, 1.0, 2, and 3 hours post injection (tail vein) of Gd-CA1.CD2 ( ⁇ 1.0 ⁇ mole/kg). The amounts of CA1.CD2 are expressed as percentages of the injected dose. Calculations are based on the assumption that the total blood volume is 8% of each individual mouse body weight. FIG.
  • 1.8( b ) The cytotoxicity of designed protein Gd-CA1.CD2.
  • the SW480 cells were grown under standard conditions (1 ⁇ 10 4 cells in 100 ⁇ l medium). The cells were treated by addition of wild type CD2 (stripe bars), CA1.CD2 (grey bars), Gd-CA1.CD2 (black bars), and Gd-DTPA (cross bars) with concentrations of 30 ⁇ M (left panel) or 50 ⁇ M (right panel).
  • the open bars are controls where cells were treated with PBS buffer. The cells were incubated with the treatments for 48 hours. The cells were then subjected for MTT assay. The results were presented as percentages of viable cells using the cells that were treated with buffer alone (filled bars) as a reference (100%).
  • the cell lines SW620 and HEK293 were similarly examined.
  • FIG. 2.1( a ) Illustrates the model structure of the designed contrast agent CA1.CD2 with eight Lys residues highlighted. The solvent accessibility calculated by Getarea is also listed.
  • FIG. 2.1 ( b ) Examples of PEGylation reagents used with different chain lengths and molecular weights and related chemical reactions.
  • FIG. 2.2 (top) illustrates a FPLC profile for the separation of pegylated protein CA1.CD2 with P12K and reaction mixture by gel filtration column. (bottom) The SDS gel of FPLC fractions (peaks 123) of CA1.CD2 pegylated with P40 stained by idiol (left) and commassie blue (right).
  • FIG. 2.3 The SDS gel stained by commassie blue (middle) for protein and iodine (top) for PEG moiety with 5:1 PEG:protein using the preactivated PEG reagents.
  • CA1.CD2 was Pegylated mainly with 3, 4, 5 PEG. (bottom) MALD-Mass analyses of mixture after PEGylation with PEG40.
  • FIG. 2.4 Illustrates the conformational analysis of PEGylaed CA1.CD2.
  • Trp emission fluorescence spectrum of CA1.CD2 is similar to that PEGylated CA1.CD2-PEG12 and CA1.CD2-PEG40 excited at 280 nm.
  • the Terbium-sensitized energy transfer was used to monitor the binding of metal ions in the designed binding pocket. The Tb3+ emission is gradually increased upon addition of terbium excited at 280 nm.
  • FIG. 2.5 Pegylated CA1.CD2-P40 remains intact after incubate with human serum for 24 hours at 37° C. monitored by SDS Page.
  • FIG. 2.6 R1 (left) and R2 (right) relaxivity values of CA1.CD2 alone, pegylated with P12, and P40 compared with DTPA at different field strengths (0.47, 3.0, 9.4, and 11.4T).
  • FIG. 2.7 Illustrates ELISA (left) or western blot (right) analyses of antibody produced in rabbit serum after i.p. injection of 3 ng/kg of protein CA1.CD2 or PEGylated CA1.CD2 (PEGCA1.CD2).
  • Pre is the serum from pre-bleeding before antigen injection.
  • CA1.CD2 was mixed with adjuvant (CA1.CD2+Ad) or with buffer saline (CA1.CD2+Sal) before injection.
  • PabCD2 is the anti-serum from rabbits produced by a commercial source use CD2 as antigen.
  • the open bars are the first bleed after first injection.
  • the gray bars are the second bleed after second injection.
  • the rabbit blood was taken 3 weeks after each injection.
  • the error bars are standard deviations of four measurements.
  • Western blots were performed with anti-serum (1st bleed) from rabbits that were injected; CA1.CD2 mixed with buffered saline (left panel, CA1.CD2+Sal), CA1.CD2 mixed with adjuvant (middle panel, CA1.CD2+Ad), and the PEGylated CA1.CD2 (right panel, PEG-CA1.CD2).
  • the Western blots experiments were carried out with 0.5 mg of PEGylated CA1.CD2 (PEG-CA1.CD2) or unmodified CA1.CD2 (CA1.CD2). Arrow indicates the position of the detected protein bands 19 hours post injection.
  • FIG. 2.8 Table 2.1 is a summary of water number in CA1.CD2 and its variants.
  • FIG. 3.1 Illustrates the development of MRI contrast agents by modifying natural calcium binding proteins such as calmodulin with four metal binding sites.
  • FIG. 3.2 Illustrates the determination of Gd 3+ stability constant of CaM variants.
  • CaM titration FIG. 3.2A
  • FIG. 3.2B curve fitting with Fura-2 fluorescence spectra. The measurement was performed at 20 mM Gd 3+ and 20 mM Fura-2 with 10 mM Tris and pH 7.4.
  • FIG. 3.2( c ) The meal selectivity of CBPP (left) and CBPP56 (right).
  • FIG. 3.3 Illustrates the SDS gel of PEGylation of CaM variants with P12 (lanes 1-6), P40 (lanes 7-10), P5K (Lanes 11-14), and P40 (lanes 15-17) at different reaction time with PEG:protein 5:1 ratio stained by Idiol (top A) and commossie blue (bottom B).
  • FIG. 3.4 Illustrates the separation of PEGylation of CaM variants-P12 with mono-Q column (left) and UV absorption spectrum of purified protein (right).
  • FIG. 3.5 Illustrates Tyr emission spectra of CAM variant without and with PEGYlation by P12K excited at 280 nm ( FIG. 3.5A ) Emission spectra of Tb fluorescence of CAM variant ( FIG. 3.5B ) and its PEGylated one in the presence of 0 (bottom), 5 uM (middle), and 20 uM of protein (top) excited at 280 nm ( FIG. 3.5C ).
  • FIG. 3.6 Shows the SDS PAGE results of serum stability for new designed protein based MRI Contrast Agents at different time points incubating with serum at 37° C. ( FIG. 3.6A ). CBP1; (FIG. 3 . 6 B).CBPP.
  • FIG. 3.7 Illustrates MRI images of Mice at 4.7T with tail vein injection of 6 mM CBP1 at 0, 10, and 30 mins post injection (top). Relative MRI intensity at different organs (bottom).
  • FIG. 3.8 Illustrates relaxivity of CBP1 at 0.47 T. Both R1 and R2 values are 5-8 fold higher than DTPA.
  • FIG. 3.9 Illustrates MRI images of Mice at 4.7T with tail vein injection of 6 mM CBP1-P40 at 0, and 13 mins post injection at slice 3 (top) and slice 4 (bottom). (right) These graphs illustrate the relative MRI intensity at different organs.
  • FIG. 3.10 Illustrates Table 3.1, which shows the dissociation constant of C 2+ , Tb 3+ and Gd 3+ to CBPP were measured by Fluorescence spectroscopy.
  • the intrinsic tryptophan fluorescence change were used to monitor the binding process between CBP1/CBPP and metal.
  • the aromatic residue-sensitized Tb 3+ fluorescence at 545 nm were applied to monitor the process of Tb 3+ binding to CBP1/CBPP and the variants.
  • FIG. 4.1 The affibody that can specifically bind to Her2 biomarker on the cancer cells was fused to the C-terminal of the protein contrast agent CA1.CD2 with a designed Gd 3+ binding site (denoted as CA1-Affi). This contrast agent was surface modified with PEG40 to reduce immunogenesity and increase solubility and serum stability (PEG-CA1-Affi). The affibody was further conjugated with near infra-red dye Cy5.5 via a Cys at the C-terminal to generate a dual labeled contrast agent PEG-CA1-Affi-Cy5.5.
  • FIG. 4.2 Contrast agent with PeGylation (PEG-CA1-affi) and without PeGylation (CA1-Aff) is able to bind to the positive cell line AU565 with membrane staining at 4 C (top right) and both membrane and cytosol staining at 37 C (top left). This developed contrast agent does not bind to negative cell line EMT-6 at either 4 or 37 C. (Bottom right) Near IR labeled contrast agent PEG-CA1-affi-Cy5.5 is able to bind to positive cell lines AU565 and AKOV-3 with NIR fluorescence signal. These data suggest that our contrast agent fused with the affibody can target to HER2 positive tumor cell lines specifically. PEGylation does not change the target capability to the cancer cell. At 37° C., the contrast agent is endocytosized.
  • FIG. 4.3 Specific targeting to the positive cancer cell line (AU565) and negative cell line (EMT6) monitored by 153 Gd ( FIG. 4.3A ) and ELISA ( FIG. 4.3B ). Similar radioactivity (CPM) of the contrast agents without PEGylation (CA1-Affi) and with PEGylation (PEG-CA1-Affi) at 75, 150, 375 nM were observed for AU565 cell line. Under identical conditions, negative cell line EMT has a small radioactivity after incubating with 153 Gd labeled contrast agent.
  • CPM radioactivity
  • FIG. 4.4 Breast cancer biomarker HER2 positive tumor and negative tumor were implanted on the left and right back in nude mice.
  • 5 mM contrast agent CA1.Affi-P40 (100 fold lower than clinic used DTPA) was injected via tail vein.
  • MRI images at 4.7 T using fast spin echo were acquired before injection, and at 5 min, 30 min, 3 hr, 24 hr and 52 hr post injection.
  • Positive tumor shows a strong contrast after 30 mins and peaked at 24 hour with about 35% enhancement. Contrast capability was decreased after 52 hours, suggesting that the contrast agent was secreted out of the animal. This mouse was alive and looks normal after 52 hours MRI scanning.
  • FIG. 4.5 Nude mice were inoculated with negative cell line MDA-MB-231 and positive cell line SKOV-3 (top). The cell number for each spot was about 5 ⁇ 10 6 .
  • the specific binding of positive tumor on the right upon injection of the dual labeled contrast PeG-CA1-Affi-Cy5.5 can be visualized using Kodak NIR in vivo FX-pro animal imaging system 21 hours poster injection.
  • (middle) Traverse MR images of tumor mice at 4.7T with fast spin echo obtained at 3 min., 35 min., and 21 hours following administration of the contrast agent.
  • Bottom right The intensity enhancement at the positive tumor by our contrast agent analyzed by Image J.
  • FIG. 4.6 Western Blot with PAbPEGCA1 (top) and NIR imaging (bottom) of different tissues of the tumor nice after MRI imaging.
  • FIG. 4.7 Immunohistochemistry (IHC) staining using the antibody PAbPGCA1 with tissue slides made from the tissue samples from the imaged mice, including HER2 tumors. Strongest staining was observed with liver and HER2 positive tumor tissue slides. The kidney slides also gave strong immunostaining. Interestingly, the areas near proximal tubes showed the strongest staining in the slides made from the kidney, indicating that the protein contrast agent was ready to be filtered through the kidney.
  • IHC Immunohistochemistry
  • FIG. 4.8 The binding of CAM-Affi with HER2 positive cells was measured by western blot.
  • FIG. 4.9 Immune staining of CaM-Affi treated breast cancer cells for skov-3 (Left, Her 2 positive) and MDA-MB-231 (right, Her2 negative).
  • FIG. 4.10 Immunostaining CA-Bom, CA-52I-Bom, CA at different time points for PC-3 and DU145 (GRPR high expression), and H441 (GRPR low expression).
  • FIG. 4.11 Near IR imaging of nude mice xenografted with DU-145 tumor (high expression of GRPR, left) and H441 tumor (low expression of GRPR, control, right) post injection of CA1.CD2-52Ibom-cy5.5-P40 26 hours via tail vein.
  • FIG. 4.12 NIR imaging (top) and NIR intensity (bottom) of CA1.CD2-52I-Bom-Cy5.5-P40 at different organs of the mice.
  • Embodiments of the present disclosure will employ, unless otherwise indicated, techniques of imaging, synthetic organic chemistry, biochemistry, biology, molecular biology, recombinant DNA techniques, pharmacology, and the like, which are within the skill of the art. Such techniques are explained fully in the literature.
  • a “contrast agent” is intended to include any agent that is physiologically tolerable and capable of providing enhanced contrast for magnetic resonance imaging.
  • a suitable contrast agent is preferably biocompatible, e.g., non-toxic, chemically stable, not absorbed by the body or reactive with a tissue, and eliminated from the body within a short time.
  • polymer means any compound that is made up of two or more monomeric units covalently bonded to each other, where the monomeric units may be the same or different, such that the polymer may be a homopolymer or a heteropolymer.
  • Representative polymers include peptides, polysaccharides, nucleic acids and the like, where the polymers may be naturally occurring or synthetic.
  • polypeptides includes proteins and fragments thereof. Polypeptides are disclosed herein as amino acid residue sequences. Those sequences are written left to right in the direction from the amino to the carboxy terminus. In accordance with standard nomenclature, amino acid residue sequences are denominated by either a three letter or a single letter code as indicated as follows: Alanine (Ala, A), Arginine (Arg, R), Asparagine (Asn, N), Aspartic Acid (Asp, D), Cysteine (Cys, C), Glutamine (Gln, Q), Glutamic Acid (Glu, E), Glycine (Gly, G), Histidine (His, H), Isoleucine (Ile, I), Leucine (Leu, L), Lysine (Lys, K), Methionine (Met, M), Phenylalanine (Phe, F), Proline (Pro, P), Serine (Ser, S), Threonine (Thr, T), Tryptophan
  • the protein can include non-standard and/or non-naturally occurring amino acids, as well as other amino acids that may be found in phosphorylated proteins in organisms such as, but not limited to, animals, plants, insects, protists, fungi, bacteria, algae, single-cell organisms, and the like.
  • the non-standard amino acids include, but are not limited to, selenocysteine, pyrrolysine, gamma-aminobutyric acid, carnitine, ornithine, citrulline, homocysteine, hydroxyproline, hydroxylysine, sarcosine, and the like.
  • the non-naturally occurring amino acids include, but are not limited to, trans-3-methylproline, 2,4-methanoproline, cis-4-hydroxyproline, trans-4-hydroxyproline, N-methyl-glycine, allo-threonine, methylthreonine, hydroxy-ethylcysteine, hydroxyethylhomocysteine, nitro-glutamine, homoglutamine, pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, 3,3-dimethylproline, tert-leucine, norvaline, 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, and 4-fluorophenylalanine.
  • Variant refers to a polypeptide or polynucleotide or polymer that differs from a reference polypeptide or polynucleotide or polymer, but retains essential properties.
  • a typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical.
  • a variant and reference polypeptide may differ in amino acid sequence by one or more modifications (e.g., substitutions, additions, and/or deletions).
  • a variant of a polypeptide includes conservatively modified variants.
  • a substituted or inserted amino acid residue may or may not be one encoded by the genetic code.
  • a variant of a polypeptide may be naturally occurring, such as an allelic variant, or it may be a variant that is not known to occur naturally.
  • a variant of a polypeptide may contain different modifications such as with PEGylation groups or the same type of groups with different sizes or lengths of the modifications.
  • Variant generated such as by modifying metal binding sites may have different metal binding properties and relaxivities and vivo peroperties.
  • the hydropathic index of amino acids can be considered.
  • the importance of the hydropathic amino acid index in conferring interactive biologic function on a polypeptide is generally understood in the art. It is known that certain amino acids can be substituted for other amino acids having a similar hydropathic index or score and still result in a polypeptide with similar biological activity. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics.
  • Those indices are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cysteine (+2.5); methionine (+1.9); alanine (+1.8); glycine ( ⁇ 0.4); threonine ( ⁇ 0.7); serine ( ⁇ 0.8); tryptophan ( ⁇ 0.9); tyrosine ( ⁇ 1.3); proline ( ⁇ 1.6); histidine ( ⁇ 3.2); glutamate ( ⁇ 3.5); glutamine ( ⁇ 3.5); aspartate ( ⁇ 3.5); asparagine ( ⁇ 3.5); lysine ( ⁇ 3.9); and arginine ( ⁇ 4.5).
  • the relative hydropathic character of the amino acid determines the secondary structure of the resultant polypeptide, which in turn defines the interaction of the polypeptide with other molecules, such as enzymes, substrates, receptors, antibodies, antigens, and the like. It is known in the art that an amino acid can be substituted by another amino acid having a similar hydropathic index and still obtain a functionally equivalent polypeptide. In such changes, the substitution of amino acids whose hydropathic indices are within ⁇ 2 is preferred, those within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • hydrophilicity can also be made on the basis of hydrophilicity, particularly, where the biological functional equivalent polypeptide or peptide thereby created is intended for use in immunological embodiments.
  • the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ⁇ 1); glutamate (+3.0 ⁇ 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); proline ( ⁇ 0.5 ⁇ 1); threonine ( ⁇ 0.4); alanine ( ⁇ 0.5); histidine ( ⁇ 0.5); cysteine ( ⁇ 1.0); methionine ( ⁇ 1.3); valine ( ⁇ 1.5); leucine ( ⁇ 1.8); isoleucine ( ⁇ 1.8); tyrosine ( ⁇ 2.3); phenylalanine ( ⁇ 2.5); tryptophan ( ⁇ 3.4).
  • an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent polypeptide.
  • substitution of amino acids whose hydrophilicity values are within ⁇ 2 is preferred, those within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
  • Exemplary substitutions that take various of the foregoing characteristics into consideration are well known to those of skill in the art and include (original residue: exemplary substitution): (Ala: Gly, Ser), (Arg: Lys), (Asn: Gln, His), (Asp: Glu, Cys, Ser), (Gln: Asn), (Glu: Asp), (Gly: Ala), (His: Asn, Gln), (Ile: Leu, Val), (Leu: Ile, Val), (Lys: Arg), (Met: Leu, Tyr), (Ser: Thr), (Thr: Ser), (Tip: Tyr), (Tyr: Trp, Phe), and (Val: Ile, Leu).
  • Embodiments of this disclosure thus contemplate functional or biological equivalents of a polypeptide as set forth above.
  • embodiments of the polypeptides can include variants having about 50%, 60%, 70%, 80%, 90%, and 95% sequence identity to the polypeptide of interest.
  • Identity is a relationship between two or more polypeptide sequences, as determined by comparing the sequences. In the art, “identity” also means the degree of sequence relatedness between polypeptides as determined by the match between strings of such sequences. “Identity” and “similarity” can be readily calculated by known methods, including, but not limited to, those described in (Computational Molecular Biology, Lesk, A. M., Ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., Ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H.
  • Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. The percent identity between two sequences can be determined by using analysis software (e.g., Sequence Analysis Software Package of the Genetics Computer Group, Madison Wis.) that incorporates the Needelman and Wunsch, (J. Mol. Biol., 48: 443-453, 1970) algorithm (e.g., NBLAST and XBLAST). The default parameters are used to determine the identity of the polypeptides of the present disclosure.
  • a polypeptide sequence may be identical to the reference sequence, that is 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the % identity is less than 100%.
  • Such alterations are selected from: at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence.
  • the number of amino acid alterations for a given % identity is determined by multiplying the total number of amino acids in the reference polypeptide by the numerical percent of the respective percent identity (divided by 100) and then subtracting that product from said total number of amino acids in the reference polypeptide.
  • Non-naturally occurring amino acids include, without limitation, trans-3-methylproline, 2,4-methanoproline, cis-4-hydroxyproline, trans-4-hydroxyproline, N-methyl-glycine, allo-threonine, methylthreonine, hydroxy-ethylcysteine, hydroxyethylhomocysteine, nitro-glutamine, homoglutamine, pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, 3,3-dimethylproline, tert-leucine, norvaline, 2-azaphenyl-alanine, 3-azaphenylalanine, 4-azaphenylalanine, and 4-fluorophenylalanine.
  • coli cells are cultured in the absence of a natural amino acid that is to be replaced (e.g., phenylalanine) and in the presence of the desired non-naturally occurring amino acid(s) (e.g., 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, or 4-fluorophenylalanine).
  • the non-naturally occurring amino acid is incorporated into the protein in place of its natural counterpart.
  • Naturally occurring amino acid residues can be converted to non-naturally occurring species by in vitro chemical modification. Chemical modification can be combined with site-directed mutagenesis to further expand the range of substitutions (Wynn, et al., Protein Sci., 2: 395-403, 1993).
  • polynucleotide generally refers to any polyribonucleotide or polydeoxyribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides as used herein refers to, among others, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
  • Polynucleotide encompasses the terms “nucleic acid,” “nucleic acid sequence,” or “oligonucleotide” as defined above.
  • polynucleotide as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the strands in such regions may be from the same molecule or from different molecules.
  • the regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules.
  • One of the molecules of a triple-helical region often is an oligonucleotide.
  • polynucleotide includes DNAs or RNAs as described above that contain one or more modified bases.
  • DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein.
  • DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples are polynucleotides as the term is used herein.
  • polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells, inter alias.
  • a polynucleotide sequence of the present disclosure may be identical to the reference sequence, that is be 100% identical, or it may include up to a certain integer number of nucleotide alterations as compared to the reference sequence.
  • Such alterations are selected from the group including at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5′ or 3′ terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.
  • the number of nucleotide alterations is determined by multiplying the total number of nucleotides in the reference nucleotide by the numerical percent of the respective percent identity (divided by 100) and subtracting that product from said total number of nucleotides in the reference nucleotide. Alterations of a polynucleotide sequence encoding the polypeptide may alter the polypeptide encoded by the polynucleotide following such alterations.
  • Codon means a specific triplet of mononucleotides in the DNA chain. Codons correspond to specific amino acids (as defined by the transfer RNAs) or to start and stop of translation by the ribosome.
  • degenerate nucleotide sequence denotes a sequence of nucleotides that includes one or more degenerate codons (as compared to a reference polynucleotide molecule that encodes a polypeptide).
  • Degenerate codons contain different triplets of nucleotides, but encode the same amino acid residue (e.g., GAU and GAC triplets each encode Asp).
  • antibody is used to refer both to a homogenous molecular entity, or a mixture such as a serum product made up of a plurality of different molecular entities.
  • Monoclonal or polyclonal antibodies, which specifically react with the virosomes of the present disclosure, may be made by methods known in the art. (e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual , Cold Spring Harbor Laboratories; Goding (1986) Monoclonal Antibodies: Principles and Practice, 2d ed., Academic Press, New York; and Ausubel et al. (1987)).
  • recombinant immunoglobulin may be produced by methods known in the art, including but not limited to, the methods described in U.S. Pat. No. 4,816,567, which is hereby incorporated by reference herein.
  • Affibody® ligands (U.S. Pat. No. 5,831,012, which is incorporated herein by reference) are highly specific affinity proteins that may be designed and used like aptamers.
  • Affibodies may be produced or purchased from commercial sources (Affibody AB, Bromma, Sweden). Aptamers and affibodies may be used in combination with antibodies to increase the functional avidity of translucent or non-translucent solid matrices for probe molecule binding. Increased binding in turn results in an increased signal strength, greater signal-to-noise ratio, more reproducible target molecule detection and greater sensitivity of detection.
  • Aptamers must also be differentiated from the naturally occurring nucleic acid sequences that bind to certain proteins. These latter sequences generally are naturally occurring sequences embedded within the genome of the organism that bind to a specialized sub-group of proteins or polypeptides, or their derivatives, that are involved in the transcription, translation, and transportation of naturally occurring nucleic acids, i.e., protein-binding nucleic acids. Aptamers on the other hand are short, isolated, non-naturally occurring nucleic acid molecules. While aptamers can be identified that bind nucleic acid-binding proteins, in most cases such aptamers have little or no sequence identity to the sequences recognized by the nucleic acid-binding proteins in nature.
  • aptamers can be selected to bind virtually any protein (not just nucleic acid-binding proteins) as well as almost any target of interest including small molecules, carbohydrates, peptides, etc.
  • a naturally occurring nucleic acid sequence to which it binds does not exist.
  • nucleic acid-binding proteins such sequences will differ from aptamers as a result of the relatively low binding affinity used in nature as compared to tightly binding aptamers.
  • Aptamers are capable of specifically binding to selected targets and modulating the target's activity or binding interactions, e.g., through binding, aptamers may block their target's ability to function.
  • the functional property of specific binding to a target is an inherent property of an aptamer.
  • a typical aptamer is 6-35 kDa in size (20-100 nucleotides), binds its target with micromolar to sub-nanomolar affinity, and may discriminate against closely related targets (e.g., aptamers may selectively bind related proteins from the same gene family).
  • Aptamers are capable of using intermolecular interactions such as hydrogen bonding, electrostatic complementarities, hydrophobic contacts, and steric exclusion to bind with a specific target.
  • aptamers also employ boronic acid-Lewis base/nucleophile (such as hydroxyl groups, diols, and amino groups) interactions for binding. Aptamers have a number of desirable characteristics for use as therapeutics and diagnostics including high specificity and affinity, low immunogenicity, biological efficacy, and excellent pharmacokinetic properties.
  • PEGylation means and refers to modifying a polymer (e.g., a protein) by covalently attaching polyethylene glycol (PEG) to the polymer, with “PEGylated” referring to a polymer having a PEG attached.
  • PEGylation methods see, for example, the Nektar Advanced PEGylation Catalogs 2004 and 2005-2006, as well as the references cited therein.
  • PEGYlation can be achieved by non-specific interaction with functional group of polypeptide chain such as via amino group or specific interaction at certain location of the macromolecules such as amino terminal or at the Cys residues.
  • biomarker or “biomarker probe” are used to refer to a substance used as an indicator of a biologic state. It is a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, and/or pharmacologic responses to a therapeutic intervention.
  • disease marker is used to refer to substances, such as proteins, bio-chemicals, nucleic acids, carbohydrates, or enzymes, produced by disease cells or by the body in response to disease cells during disease development and progression. These substances are indicative of a particular disease process.
  • treatment As used herein, the terms “treatment”, “treating”, and “treat” are defined as acting upon a disease, disorder, or condition with an agent to reduce or ameliorate the pharmacologic and/or physiologic effects of the disease, disorder, or condition and/or its symptoms.
  • Treatment covers any treatment of a disease in a host (e.g., a mammal, typically a human or non-human animal of veterinary interest), and includes: (a) reducing the risk of occurrence of the disease in a subject determined to be predisposed to the disease but not yet diagnosed as infected with the disease (b) impeding the development of the disease, and (c) relieving the disease, i.e., causing regression of the disease and/or relieving one or more disease symptoms. “Treatment” is also meant to encompass delivery of a contrast agent including a compound to provide a pharmacologic effect, even in the absence of a disease or condition.
  • treatment encompasses delivery of a disease or pathogen compound via the contrast agent that provides for enhanced or desirable effects in the subject (e.g., reduction of pathogen load, reduction of disease symptoms, etc.).
  • prophylactically treat or “prophylactically treating” refers to completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and/or animal subjects, each unit containing a predetermined quantity of a contrast agent calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for unit dosage forms depend on the particular compound employed, the route and frequency of administration, the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • a “pharmaceutically acceptable excipient,” “pharmaceutically acceptable diluent,” “pharmaceutically acceptable carrier,” or “pharmaceutically acceptable adjuvant” means an excipient, diluent, carrier, and/or adjuvant that are useful in preparing a pharmaceutical composition that are generally safe, non-toxic and neither biologically nor otherwise undesirable, and include an excipient, diluent, carrier, and adjuvant that are acceptable for veterinary use and/or human pharmaceutical use.
  • “A pharmaceutically acceptable excipient, diluent, carrier and/or adjuvant” as used in the specification and claims includes one or more such excipients, diluents, carriers, and adjuvants.
  • a “pharmaceutical composition” is meant to encompass a contrast agent suitable for administration to a subject, such as a mammal, especially a human.
  • a “pharmaceutical composition” is sterile, and preferably free of contaminants that are capable of eliciting an undesirable response within the subject (e.g., the compound(s) in the pharmaceutical composition is pharmaceutical grade).
  • Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, intravenous, buccal, rectal, parenteral, intraperitoneal, intradermal, intracheal, intramuscular, subcutaneous, inhalational and the like.
  • the terms “therapeutically effective amount” and “an effective amount” are used interchangeably herein and refer to that amount of a contrast agent being administered that is sufficient to effect the intended application.
  • the effective amount of the contrast agent includes enough so that the disease, for example, in the host can be imaged, studied, diagnosed, or the like.
  • an effective amount of a contrast agent including a compound will relieve to some extent one or more of the symptoms of the disease being treated, and/or that amount that will prevent, to some extent, one or more of the symptoms of the disease that the host being treated has or is at risk of developing.
  • the therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • the term “host,” “subject,” “patient,” or “organism” includes humans and mammals (e.g., mice, rats, pigs, cats, dogs, and horses). Typical hosts to which compounds of the present disclosure may be administered will be mammals, particularly primates, especially humans. For veterinary applications, a wide variety of subjects will be suitable, e.g., livestock such as cattle, sheep, goats, cows, swine, and the like; poultry such as chickens, ducks, geese, turkeys, and the like; and domesticated animals particularly pets such as dogs and cats.
  • livestock such as cattle, sheep, goats, cows, swine, and the like
  • poultry such as chickens, ducks, geese, turkeys, and the like
  • domesticated animals particularly pets such as dogs and cats.
  • living host refers to a host noted above or another organism that is alive.
  • living host refers to the entire host or organism and not just a part excised (e.g., a liver or other organ) from the living host.
  • Cancer as used herein, shall be given its ordinary meaning, as a general term for diseases in which abnormal cells divide without control.
  • cancer refers to angiogenesis related cancer. Cancer cells can invade nearby tissues and can spread through the bloodstream and lymphatic system to other parts of the body.
  • carcinoma is cancer that begins in the skin or in tissues that line or cover internal organs.
  • Sarcoma is cancer that begins in bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue.
  • Leukemia is cancer that starts in blood-forming tissue such as the bone marrow, and causes large numbers of abnormal blood cells to be produced and enter the bloodstream.
  • Lymphoma is cancer that begins in the cells of the immune system.
  • a tumor When normal cells lose their ability to behave as a specified, controlled and coordinated unit, a tumor is formed.
  • a solid tumor is an abnormal mass of tissue that usually does not contain cysts or liquid areas (some brain tumors do have cysts and central necrotic areas filled with liquid). A single tumor may even have different populations of cells within it, with differing processes that have gone awry.
  • Solid tumors may be benign (not cancerous), or malignant (cancerous). Different types of solid tumors are named for the type of cells that form them. Examples of solid tumors are sarcomas, carcinomas, and lymphomas. Leukemias (cancers of the blood) generally do not form solid tumors.
  • Representative cancers include, but are not limited to, bladder cancer, breast cancer, colorectal cancer, endometrial cancer, head and neck cancer, lung cancer, lymphoma, melanoma, non-small-cell lung cancer, ovarian cancer, prostate cancer, testicular cancer, uterine cancer, cervical cancer, thyroid cancer, gastric cancer, brain stem glioma, cerebellar astrocytoma, cerebral astrocytoma, glioblastoma, ependymoma, Ewing's sarcoma family of tumors, germ cell tumor, extracranial cancer, Hodgkin's disease, leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, liver cancer, medulloblastoma, neuroblastoma, brain tumors generally, non-Hodgkin's lymphoma, osteosarcoma, malignant fibrous histiocytoma of bone, retinoblastoma, rhabdomyosarcom
  • a tumor can be classified as malignant or benign. In both cases, there is an abnormal aggregation and proliferation of cells. In the case of a malignant tumor, these cells behave more aggressively, acquiring properties of increased invasiveness. Ultimately, the tumor cells may even gain the ability to break away from the microscopic environment in which they originated, spread to another area of the body (with a very different environment, not normally conducive to their growth), and continue their rapid growth and division in this new location. This is called metastasis. Once malignant cells have metastasized, achieving a cure is more difficult.
  • Benign tumors have less of a tendency to invade and are less likely to metastasize. Brain tumors spread extensively within the brain but do not usually metastasize outside the brain. Gliomas are very invasive inside the brain, even crossing hemispheres. They do divide in an uncontrolled manner, though. Depending on their location, they can be just as life threatening as malignant lesions. An example of this would be a benign tumor in the brain, which can grow and occupy space within the skull, leading to increased pressure on the brain.
  • precancerous cells cancer cells, cancer, and tumors may be used interchangeably in the disclosure.
  • embodiments of the present disclosure in one aspect, relate to contrast agents, compositions including contrast agents, methods of making contrast agents, methods of imaging, methods of diagnosing, methods of studying, and the like. More particularly, embodiments of the contrast agents include magnetic resonance imaging contrast agents that accumulate in tissue and can be used to determine the presence and/or location of a target.
  • contrast agents of the present disclosure can include targeting agents to target cells or tissue (e.g., cancer).
  • Embodiments of the present disclosure can be tuned to have properties for diagnostic imaging.
  • the contrast agent includes a metal ion interacting (e.g., bonding with or chelating with) with the metal ion binding site.
  • a contrast agent can include two metal ions interacting (e.g., bonding with or chelating with) with two metal ion binding sites.
  • the metal ion binding site may be developed by a design approach or by a grafting approach. After the site has been developed, the site or sites are operatively integrated into the select areas of the scaffold polymer.
  • the contrast agent is stable in a physiological environment.
  • physiological environment can be described as cell, cellular conditions, tissues, organs, and vertebrate/invertebrates, animal/human or buffer conditions (e.g., pH of about 6-8 and a temperature of about 5-45° C.) mimic closely to the cellular, or in vivo conditions.
  • stable in reference to “physiological environment” means that the contrast agent is able to provide contrast capability and remain intact.
  • the phrase “stable in a physiological environment” refers to the contrast agent including at least one metal ion and the binding of the metal ion causes no changes or substantially no changes (e.g., less than 50%) to the protein (scaffold protein) conformation or to the binding affinity of the tailored metal ion binding site under clinical conditions (physiological environment) that would cause premature release of the metal ion, and that the contrast agent functions as a contrast agent as described herein.
  • the scaffold polymer of the contrast agent includes polyethylene glycerol compounds (PEG) attached to the polymer.
  • PEG polyethylene glycerol compounds
  • the PEGs can be attached (e.g., bonded) to the polymer via an amino acid residue such as lysine (Lys), glutamic acid (Glu), aspartic acid (Asp), cysteine (Cys), and/or carboxyl/amino terminals.
  • the position of the amino acids on the polymer can be selected to position the PEGs so that the PEGs do not substantially interfere (e.g., decrease metal binding affinity more than 20%) with or interfere with the metal ion binding sites ability to interact with the metal ion of interest or the conformation of the polymer.
  • the PEGs are attached to one or more Lys residues since the position of the Lys residues on the polymer is such that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
  • reference to “contrast agent” refers to a contrast agent that includes PEGs.
  • embodiments of the present disclosure include contrast agents that include PEGs and contrast agents that do not include PEGs. Additional details regarding PEGs are described herein.
  • Embodiments of the present disclosure provide for PEGylated contrast agents, where the PEGylation increases the blood circulation time of the contrast agent in CD-1 mice.
  • PEGylation of the contrast agent increased the solubility of the contrast agent by more than two-fold, three-fold, four-fold, five-fold, six-fold, seven-fold, eight-fold, nine-fold, ten-fold, or more relative to the un-PEGylated contrast agent.
  • PEGylation of the contrast agent further increased the in vitro of one or both R1 and R2 relaxivities of the contrast agents by about 10%, 25%, 50%, 75%, 100%, or 2-3 fold or more, relative to the un-PEGylated contrast agent.
  • the increase in molecular size due to the PEGylation and the addition of a hydration layer due to water retention by Poly-PEG chain on protein surface may be the reasons for the increases in the relaxivities.
  • Embodiments of this disclosure include contrast agents capable of enhancing image contrast by affecting water molecule proton relaxation rates. Such contrast agents are effective for magnetic resonance imaging, in part, because the water proton relaxation rate in the target tissue is affected differently from the relaxation rate of the water protons in the surrounding tissue.
  • the contrast agents are paramagnetic species, which form complexes with metal ions, so to alter the relaxation rates of adjacent nuclei.
  • the scaffold polymer (referred to as a “protein” or “peptide” hereinafter) for MRI applications are a protein that will host the tailored metal ion binding sites and has the following characteristics:
  • a rotational correlation time optimized for the magnetic field e.g., around 100 milliseconds in a magnetic field of 1.3 to 3T
  • higher magnetic field application can demand a host protein with a larger molecular weight
  • the contrast agent for use in MRI applications can include a scaffold protein (referred to as a “protein”, “polymer”, or “peptide”) that includes a natural metal binding protein or a fragment/domain of natural metal binding proteins either with metal binding sites modified by at least one amino acid or protein modification.
  • a scaffold protein referred to as a “protein”, “polymer”, or “peptide” that includes a natural metal binding protein or a fragment/domain of natural metal binding proteins either with metal binding sites modified by at least one amino acid or protein modification.
  • Properties of the scaffold protein also may include water solubility, low interaction with the other cellular metal ions and low toxicity. While all these properties are not required, the optimal properties of the scaffold protein can depend on the specific parameters of the imaging application.
  • the scaffold protein has a three-dimensional structure or an amino sequence with some homology to the proteins whose structures have been solved, at least in part. Specifically, the scaffold protein is screened to determine whether it can tolerate the integration of various binding sites without excessive denaturation. For example, the integration of metal ion binding sites into the scaffold protein should not denature or unfold the protein. Thus, the metal ion binding site should not be placed by mutating a hydrophobic core or in a position that results in substantial structural perturbation. This can be examined by sequence alignment of proteins in the same family. In an embodiment, the amino acids that have an essential role in folding of the structure or the function will be conserved among different species of this same type of the protein.
  • metal ion binding sites are placed into a scaffold protein such that the metal can be tumbled together with the protein. It is better to find a location that is not so flexible or the same flexibility as the protein body so as to match the correction time. In an embodiment, it is preferred to design or create the binding pocket in the protein. Although insertion could work, it is preferable to do so in a relatively not so flexible region.
  • the protein can be checked by looking at the B factor (temperature factor for X-ray) or S2 factor (dynamic flexibility factor for NMR) of the pdb (protein data bank) file of the structure.
  • more than one metal binding site may be integrated into a scaffold protein.
  • the inclusion of more than one binding site improves the sensitivity of the contrast agent.
  • the site could have different affinities, but should still have strong enough affinity for the selected metal so to avoid competition with physiological metal ions. Both metal ions should be embedded into the host protein with preferred rotational correlation times and water exchange rates to provide MRI contrast with an increased sensitivity.
  • the contrast agent can have a high affinity to and can preferentially select a particular metal ion (e.g., Gd 3+ , Mn 2+ , or Fe 3+ ).
  • exemplary contrast agents showed a dissociation constant K d less than 10 12 [M] for Gd 3+ in an environment having physiological metal ions and prevented those metal ions from precipitation under physiological conditions.
  • the present disclosure may be used to create contrast agents having optimal selectivity for a specific metal ion.
  • Embodiments of the present disclosure can provide a new mechanism to increase the relaxivity of contrast agents. This is accomplished by designing the metal ion binding sites, e.g., Gd 3+ , in proteins, which can eliminate the mobility and flexibility of the chelating moiety associated with currently available contrast agents. High proton relaxivity by contrast agents can further enhance images.
  • the metal ion binding sites e.g., Gd 3+
  • An advantage of the present disclosure is that it provides contrast agents that can preferentially chelate a specific metal ion.
  • a preferred contrast agent having Gd 3+ binding site(s) will preferentially chelate Gd 3+ over other metal ions, such as Mg 2+ or Ca 2+ .
  • the ability to preferentially chelate a specific metal ion can improve the specificity of a contrast agent and reduces the cytotoxicity of the contrast agent.
  • the contrast agents include PEGs attached to the protein. Inclusion of the PEGs in the contrast agent increases blood circulation time, increases the solubility of the contrast agent in a physiological system, and/or increases R1 and R2 relaxivities, relative to un-PEGylated contrast agents.
  • the PEGs are bonded to the protein via an amino acid residue such as lysine (Lys), glutamic acid (Glu), aspartic acid (Asp), cysteine (Cys), carboxyl/amino terminals, or combinations thereof.
  • the position of the amino acids on the polymer should position the PEGs so that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
  • a fusion protein/peptide/polymer or a non-degradable particle moiety can be added to the protein contrast agent with a linker to tune the correlation time for optimal contrast sensitivity, targeting (e.g., subcellular, cellular, tissue and organ selectivity), biodistribution (e.g., affiibody against to Her-2 was fused to CA1 as a targeted contrast agent to breast cancer), and/or bioelimination (e.g., using proteins with molecular weight less than 60 KDa).
  • targeting e.g., subcellular, cellular, tissue and organ selectivity
  • biodistribution e.g., affiibody against to Her-2 was fused to CA1 as a targeted contrast agent to breast cancer
  • bioelimination e.g., using proteins with molecular weight less than 60 KDa
  • Scaffold proteins suitable with the present disclosure include proteins or organic polymers containing amino acids.
  • the scaffold proteins can be modified.
  • the scaffold proteins are inclusive of both natural amino acids and unnatural amino acids (e.g., beta-alanine, phenylglycine, and homoarginine, Gamma-carboxyglutamate (Gla)).
  • the amino acids are alpha-amino acids, which can be either of the L-optical isomer or the D-optical isomer.
  • the amino acids are D-optical isomers, as such isomers are less subject to proteolytic degradation.
  • Such amino acids can be commonly encountered amino acids that are not gene-encoded, although preferred amino acids are those that are encodable.
  • a Near-IR functional group e.g., Cy5.5, Cy7, Alexflour, and indocyanine green
  • a targeting agent for Near-IR detection is covalently bound to the scaffold protein, a PEG, and/or a targeting agent.
  • the scaffold proteins should include one or more amino acid residues (e.g., Lys, Glu, Asp, Cys, or combinations thereof) able to bond with the PEGs or otherwise modified.
  • the position of the amino acids on the protein should position the PEGs so that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
  • scaffold proteins may be used according to the disclosure, but in general they will be proteins, and organic polymers. More specifically, suitable scaffold proteins can be selected properties suitable for diagnostic applications.
  • the scaffold protein for use with this disclosure may be of unitary construction (a particulate, a polychelant or a dendrimeric polymer). Scaffold proteins suitable with this disclosure may be selected without undue experimentation.
  • Embodiments of the present disclosure can include proteins such as CD2 proteins (a cell adhesion protein) that exhibit high stability against proteolysis, thermal conditions (Tm 67° C.), pH (2-10), and salt (0-4 M NaCl) denaturation.
  • CD2 proteins can be suitable with this disclosure because such proteins are stable in physiological environments, have a topology suitable for the integration of at least one or multiple metal ion chelating sites, and typically have a relaxivity greater than 10 mM ⁇ 1 s ⁇ 1 (some of them up to about 50 mM ⁇ 1 s ⁇ 1 ).
  • CD2 proteins can tolerate multiple surface mutations without unfolding the protein.
  • the CD2 proteins can be used as a host protein to design calcium binding sites. Examples using CD2 are described herein.
  • Fluorescent proteins are another class of preferred scaffold protein for this disclosure, as these proteins are stable in a physiological environment against proteolytic degradation and pH denaturation (pH 5-10).
  • Such fluorescent proteins include an array of fluorescent proteins including those related to Aequorea . Suitable fluorescent proteins should have a useful excitation and emission spectra and may have been engineered from naturally occurring Aequorea victoria green fluorescent proteins (GFPs).
  • GFPs green fluorescent proteins
  • modified GFPs may have modified nucleic acid and protein sequences and may include elements from other proteins.
  • the cDNA of GFPs may be concatenated with those encoding many other proteins—the resulting chimerics are often fluorescent and retain the biochemical features of the partner proteins.
  • Such proteins also are included in the disclosure.
  • contrast agents constructed from such proteins can be multi-functional probes.
  • the contrast agent constructed from fluorescent proteins can be screened using both fluorescence and MR imaging. This can be advantageous as such properties equip the contrast agent with both the fluorescence needed for fluorescence detection methods and sensitivity needed for the deep tissue detection from MRI.
  • Such contrast agents are multifunctional contrast agents.
  • scaffold proteins are able to tolerate the addition of the metal ion binding site without substantial disruption to its structure.
  • metal binding site such as calcium binding sites as scaffold protein proteins.
  • These natural metal binding proteins such as calmodulin, calbindin D9K, troponin C, and parvalbumin, can be engineered to bind paramagnetic metal ions with very strong metal binding affinity thus are capable of enhancing image contrast by affecting water molecule proton relaxation rates.
  • Embodiments of scaffold protein sequences that can be included in the contrast agent are provided that include the unmodified scaffold proteins and modified scaffold proteins (insertions and/or deletions) for a variety of illustrative scaffold proteins that include metal ion binding sites.
  • the scaffold protein sequences include one or more possible locations for attachment of PEGs, mutation sites, C-terminal sites for pegylation or conjugation of moieties (e.g., fluorescent dyes), and the like.
  • embodiments of the present disclosure include contrast agents where PEGs are attached to the protein via one or more amino acid residues such as Lys, Glu, Asp, Cys, carboxyl/amino terminals, or combinations thereof.
  • the PEGs are attached to amino acid residues so that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
  • the PEGs can be attached to the amino acid residues through PEGylation processes known in the art. The PEGylation may, for example, be performed at a pH of about 7.5 to 9 or about 8 to 8.5.
  • the PEGs can be linear PEGs, multi-arm PEGs, branched PEGs, and combinations thereof.
  • the molecular weight of the PEGs can be about 1 kDa to 100 kDa, about 1 kDa to 50 kDa, about 1 kDa to 40 kDa, about 1 kDa to 30 kDa, about 1 kDa to 20 kDa, about 1 kDa to 12 kDa, about 1 kDa to 10 kDa, or about 1 kDa to 8 kDa. It should be noted that the molecular weight can be any integer within any of the values mentioned above.
  • the word “about” indicates an approximate average molecular weight and reflects the fact that there will normally be a certain molecular weight distribution in a given polymer preparation.
  • 1 to 10 PEGs can be attached to the scaffold protein.
  • 2 to 6 PEGs can be attached to the scaffold protein.
  • 2 to 4 PEGs can be attached to the scaffold protein.
  • the PEGs can have additional functional groups to allow us to further modify the contrast agent by adding other moieties such as signal peptides (such as GRP signal peptide for targeting to prostate cancer).
  • signal peptides such as GRP signal peptide for targeting to prostate cancer.
  • a targeting agent can be attached (e.g., directly or indirectly) to the scaffold polymer or the PEG, where the targeting agent has an affinity for a target (e.g., a cell, a tissue, a protein, an antibody, an antigen, and the like).
  • the targeting agent can include, but is not limited to, polypeptides (e.g., proteins such as, but not limited to, antibodies (monoclonal or polyclonal)), antigens, nucleic acids (both monomeric and oligomeric), polysaccharides, sugars, fatty acids, steroids, purines, pyrimidines, ligands, or combinations thereof, where the targeting agent binds or otherwise interacts with the target.
  • the targeting agent specifically interacts with a specific type of target or specific target and substantially (e.g., 90%, 95%, 99% or more specificity to the target or type of target) or completely excludes other targets.
  • the targeting agent has an affinity for one or more targets.
  • the targeting agent can include: a biomarker probe, a precancerous targeting agent, a cancer targeting agent, a tumor targeting agent, and a probe or agent that targets at least two of a biomarker, a precancerous cell, a cancer cell, and a tumor.
  • the targeting agent can be linked, directly or indirectly, using a stable physical, biological, biochemical, and/or chemical association.
  • the targeting agent can be independently linked to the scaffold polymer or the PEG using, but not limited to, a covalent link, a non-covalent link, an ionic link, a chelated link, as well as being linked through interactions such as, but not limited to, hydrophobic interactions, hydrophilic interactions, charge-charge interactions, ⁇ -stacking interactions, combinations thereof, and like interactions.
  • the targeting agent can include, but is not limited to, (gastrin release peptide (GRP) that can bind to specific types of cancer receptors, i.e. GRP receptors, and, RGD peptides (Arg-Gly-Asp) (corresponding to integrin ⁇ v ⁇ 3 target).
  • GRP gastrin release peptide
  • RGD Arg-Gly-Asp
  • molecules that can be targets include, but are not limited to, vascular receptors (e.g., Vascular endothelial growth factor receptor (VEGF-R)), extracellular matrix proteins (e.g., proteases, MMP, thrombin), cell membrane receptors (e.g., epidermal growth factor receptor (EGFR) (e.g., HER2)), intracellular proteins, enzymes (e.g., caspases and PSA), serum proteins (e.g., albumin), and the like.
  • vascular receptors e.g., Vascular endothelial growth factor receptor (VEGF-R)
  • extracellular matrix proteins e.g., proteases, MMP, thrombin
  • cell membrane receptors e.g., epidermal growth factor receptor (EGFR) (e.g., HER2)
  • intracellular proteins e.g., enzymes (e.g., caspases and PSA), serum proteins (e.g., albumin), and the like.
  • An engineered metal binding site can be created by a combination of more than one above mentioned methods.
  • the computational design approach focuses on designing a metal ion binding site de novo. This design approach focuses on using an algorithm to construct and engineer an optimal binding site. Preferably, the computation design approach is used to create optimal binding sites by, e.g., varying the coordination geometry of the site, the water number in the coordination shells, the ligand types, and the charges.
  • the computational design approach comprises the following steps:
  • the metal ion binding site may be incorporated into a scaffold protein, e.g., a fluorescent or CD2 protein. Further, such a method may be used to alter metal ion binding properties of proteins and generate new materials with various ion binding affinities.
  • the method involves searching and accessing public and or private databases for preferred components of a metal ion binding site.
  • databases that may be searched for the criteria or components may include public domain banks (e.g., NBCI or PubMed) or knowledge banks such as protein modeling structure data banks (e.g., Cambridge or RCSB Protein Data Bank Data Bank and BioMagResBank database) or data bank.
  • the database could include structural data from metal ion binding proteins whose structures have been characterized previously.
  • One of ordinary skill in the art can identify databases and sources of material for databases suitable with this disclosure. Use of a computer obviously would greatly speed up the searching and is preferred.
  • These databases may be used to provide structural analysis of one to several thousand different small molecules or metal ions that bind to a protein.
  • Such analysis may include local coordination properties, types of residues or atoms commonly used to bind a desired metal ion, chemical features (e.g., pKa or changes), the number of charged residues on a site, and the range or deviation of the known binding sites.
  • Further, such analysis may include the environment, such as types of atoms, residues, hydrophobicity, solvent accessibility, shapes of the metal binding sites, electrostatic potentials, and the dynamic properties (e.g., B-factors or the order factors of the proteins) of the binding sites.
  • Such analysis also may include whether a binding site for a particular metal ion is a continuous or discontinuous binding site.
  • one or more suitable metal ion binding sites may be generated based on rational factors.
  • different search algorithms may be used to generate potential metal ion binding sites based on other key features in addition to, for example, the geometric descriptors. These key features include the properties of the original residues in the scaffold protein, ligand positions that are essential to protein folding, the number of the charged residues and their arrangement and number of water molecules in the coordination shell. The hydrogen bond network and the electrostatic interactions with the designed ligand residues also can be evaluated.
  • the protein environments of metal ion binding sites can be analyzed according to solvent accessibility, charge distribution, backbone flexibility, and properties of scaffold proteins. Thus, one of ordinary skill in the art may rationally select a binding site based on desired parameters.
  • a site may be tailored using two complementary approaches of computational design and grafting (see below).
  • the computational design approach includes modifying the metal ion binding site by modifying residues in the scaffold of the metal ion binding site.
  • a geometric description of the ligands around a metal ion, a three-dimensional structure of the backbone of proteins, and a library of side-chain rotamers of amino acids (or atoms from the main chain) can identify a set of potential metal-binding sites using a computer.
  • key ligand residues are carefully placed in the amino acid sequence to form the metal (metal ion) binding pocket. This binding pocket can be created automatically by the computer algorithm according to the coordination description and the user's preferred affinity.
  • the created potential metal ion binding sites can be optimized and tuned to specification.
  • a backbone structure of the metal ion binding site with different degrees of flexibility may be used according to the need or the flexibility of the metal ion binding site.
  • the designed metal ion binding sites are further filtered and scored based on the local factors, which may include the shape of the metal ion binding sites, locations, charge numbers, dynamic properties, the number of mutation needed, solvent accessibility, and side chain clashes.
  • one to two oxygen atoms from the solvent water molecules in the coordination shell may provide additional coordination without reducing the required binding affinity and selectivity.
  • Stronger metal ion binding affinities of the designed sites may be developed based on several modeled factors that contribute to metal ion affinity.
  • the number of ligand residues is a factor to directly chelate a specific metal ion.
  • the number of ligand residues is a factor to directly chelate a specific metal ion.
  • the number of charged residues is able to change metal ion affinity.
  • the ligand type is a factor as the binding preferences of a chelate may depend on the particular ligand type.
  • An illustrative version of this computational approach is the computerized (or otherwise automated) querying of one or more databases that comprise structural data on metal ion binding sites using selected criteria relevant to the metal ion binding site, generating at least one preliminary metal ion binding site from the database information based on compatibility with the selected criteria, and selecting one or more suitable metal ion binding sites from the preliminary metal ion binding sites based on optimal compatibility with the selected criteria.
  • the nucleic acid sequence of the selected metal ion binding site is obtained, tailored, and operatively linked with a scaffold protein sequence, whereby the nucleic acid sequence of the selected metal ion binding site is tailored so to achieve the metal ion binding site having a desired specificity for the metal ion.
  • a nucleic acid sequence encoding the preliminary binding sites can be generated from the structural or model data. The computational approach also can be used to produce the metal ion binding site.
  • the computational approach can be performed on or by a system comprising at least one database that comprises the structural data on metal ion binding sites, an algorithm for generating the preliminary metal ion binding sites from portions of the structural or model data using selected criteria relevant to the metal ion binding site and rating the preliminary metal ion binding sites based on specificity for a selected metal ion, and a computer for executing the algorithm so as to query the databases to generate the preliminary metal ion binding sites.
  • the algorithm generally is a relatively simple searching algorithm that will query the databases based on inputted criteria.
  • the grafting method focuses on engineering and constructing a metal ion binding site by modifying the primary, secondary, tertiary, and/or quaternary structure of an identified binding site. By selectively manipulating the structure of the binding site, it is possible to obtain a metal ion binding site that can be engineered into a scaffold protein, e.g., CD2 or fluorescent protein, without significantly denaturing the protein. Using the grafting method, it is possible to achieve a binding site that has a stronger preference for one metal ion over another metal ion. Such modifications may allow for improved contrast abilities.
  • a scaffold protein e.g., CD2 or fluorescent protein
  • an identified binding site for use with the grafting method may be any continuous sequence site that has some affinity for a metal ion.
  • binding sites may derive from either known binding peptides such as an individual EF-hand site or from short fragments that have demonstrated the ability to bind specific metal ions such as alpha-lactalbumin.
  • Such peptides may be highly conserved in nature and prevalent throughout nature or may be unnatural but known to have an affinity for a particular metal ion.
  • One of ordinary skill in the art is able to identify binding sites with affinity for a metal ion without undue experimentation.
  • the primary structure of the metal ion binding site may be altered and tuned to achieve a metal ion binding site with improved binding characteristics.
  • more charged ligand residues such aspartate and glutamate may be engineered by inserting codon(s) into the metal ion binding site so as to tune the responsiveness of the site or the scaffold protein.
  • the inclusion of additional charged ligands can allow the contrast agent to achieve an affinity for selected paramagnetic metal ions and to have a desired selectivity.
  • one or two water molecules can also be introduced into the coordination shell by removing or modifying ligand residues and their environments.
  • Further other mutations to the primary structure include removing or adding amino acids to change properties such as flexibility or rigidity of the site. Adding or removing amino acids from the binding site alters the primary structure of the binding site.
  • the secondary structure of the metal ion binding site may be modified to tune the sensitivity and responsiveness of the metal ion binding site.
  • the residues on the site itself, the flanking or the neighboring structures such as helices, beta strands, or turns may be modified by changing properties such as hydrophobicity, salt bridges, secondary structure propensity (e.g., helicity and ⁇ -sheets), and charge interactions with different amino acids, which all may inherently change the secondary structure.
  • the tertiary structure of the metal ion binding site may be modified to further tune the sensitivity and responsiveness of the metal ion binding site.
  • the affinity of the metal ion binding site for the metal ion may be varied by selectively manipulating and adding helices, loops, bridges and/or linkers and chemical properties such as hydrogen bonding, electrostatic interactions and hydrophobic interactions.
  • variations in tertiary structure may add stability and affinity by increasing secondary structure propensity, adding charge interaction of the side chains, and by stabilizing the metal ion binding coordination chemistry.
  • the dynamic properties can be modified by increasing the packing of the protein and replacing residues with amino acids or other moieties with more rigid (e.g., Pro) or flexible (e.g., Gly) properties,
  • One method of directly altering the primary, secondary, and/or tertiary structure of the metal ion binding site is by altering the charges in the site.
  • the charges in any binding site have a significant role in the structure of the site, changing the charges or charge ratio may have significant impact on the structure of the site.
  • the charged side chains exhibit a strong influence on the metal ion binding affinity even though they are not directly involved as ligands, the variation of these chains results in variations in metal ion binding affinities and selectivity.
  • a metal ion binding site may have stronger affinities to and better selectivity for a desired metal ion over a competitive metal ion by designing or modifying the site, e.g., changing the number of charged ligand residues to form metal ion binding pockets.
  • the metal ion binding affinity of the metal ion binding site may be varied by changing the charged side chains that are present on the metal ion binding site and/or the neighboring environment. The replacement of charged residues such as aspartate or glutamate with a residue such as alanine may dramatically reduce the binding affinity for the metal ion by up to 100 times.
  • the contrast agent in the case of multifunctional contrast agents, e.g., where the contrast agent is a fluorescent protein, it can be a factor to induce the metal binding site without altering significantly the chromophore environment to reduce the fluorescent signal.
  • These metal binding sites can be added at remote locations away from the chromophore or simply fusion to the fluorescent moieties. Such locations can be evident from the sequence and protein folding.
  • the grafting approach may be used with the design approach to create an optimal metal binding site.
  • metal binding sites can be created by using part of continuous site and part of ligand residues created by computer design.
  • the loops or any sequences of the proteins can be removed or modified to achieve optimal required binding affinity, metal selectivity, relaxivity and stability.
  • Natural metal binding proteins' their metal binding affinity can be altered by directly modifying the proteins such as the addition of metal ligand residues in the calcium binding proteins to increase metal binding affinity to lanthanides.
  • fragments and/or domains of the natural metal binding proteins encompassing metal binding sites can also serve as scaffold protein of the contrast agents if they exhibit strong metal binding affinity for Ln 3+ or other paramagnetic metal ions, serum stability, and desired relaxation properties.
  • the affinity to natural metal ions such as physiological metal ions, e.g., calcium, zinc, and magnesium, will be significantly reduced by deleting metal binding ligand residues or reducing the cooperativity between coupled metal binding sites.
  • the calcium binding sites in the natural calcium binding protein such as calmodulin and parvalbumin were modified so that the modified proteins have a strong metal binding affinity to lanthanides.
  • the metal selectivity for lanthanides over calcium, magnesium and zinc are very high. If it is necessary, the molecular recognition sites of these natural calcium binding proteins can be altered by deletion at the active sites or PEGylation.
  • sequences for N- and C-terminal domains of calmodulin and its variants are listed that can be serve as a protein contrast agents (See sequences included herein). Additional modifications can performed to reduce their intrinsic biological function, avoid immunogenicity, increase serum stability, and targeting capability.
  • the metal ion binding sites may be selectively introduced into numerous sites of a scaffold protein without substantially impairing its secondary structure.
  • a number of methods for identifying integration sites in proteins such CD2 proteins, fluorescent proteins (e.g., GFP, YFP, CFP, and RFP) are known in the art, including, for example, site directed mutagenesis, insertional mutagenesis, and deletional mutagenesis.
  • Other methods including the one exemplified below and in the Examples, are known or easily ascertained by one skilled in art.
  • the sites of the fluorescent protein that can tolerate the insertion of a metal ion binding site also may be determined and identified by gene manipulation and screening. By generating mutant proteins and by manipulating the DNA sequence, it is possible to obtain a variety of different insertions, which then may be screened to determine whether the protein maintains its intrinsic activities. Preferably, sites that remove or interfere with the intrinsic fluorescence of the fluorescent protein are not optimal and may be screened out. Variants identified in this fashion reveal sites that can tolerate insertions while retaining fluorescence.
  • the metal ion binding sites for use with scaffold proteins may be selected by considering five criteria so to as optimize the local properties of the metal binding site, the fluorescent protein, and the protein environment.
  • Third, the water coordination shell of the metal ion chelating sites should be able to coordinate at least 1-2 water molecules.
  • the residues from the loops between the secondary structures with good solvent accessibility are desired for both the folding of the protein and the fast kinetics required for the contrast agent.
  • the mutation or the introduction of the metal ion binding site should not substantially interfere with the synthesis and folding of the protein. More particularly, the introduction of the metal ion binding site does not interfere with either post-translational chromophore formation or intermolecular interactions required for stabilizing the chromophores and folding of the protein frame. Furthermore, the introduced side chain should not be overpacked and should not clash with the protein frame of the scaffold protein (e.g., the fluorescent protein).
  • the direct use of chromophore residues as chelating sites is not preferred but is within the scope of this disclosure.
  • the metal binding sites in the natural metal binding proteins can be directly modified to have proper metal binding affinity to the desired metal ions.
  • One or more metal ions are atoms and ions, including the respective isotopes and radioisotopes, that can bind to proteins or peptides.
  • a metal ion may bind reversibly or irreversibly and such a bond may be covalent or non-covalent.
  • Gd 3+ is used in some embodiments of this disclosure as an exemplary metal ion, it is understood that metal ions suitable with this disclosure include, but are not limited to metal ions including Group IIA metal ions, transition metal ions, and Lanthanide Series ions.
  • Exemplary metal ions include, but are not limited to, the ion, isotope, and/or radioisotope forms of magnesium, calcium, scandium, titanium, manganese, iron, boron, chromium, cobalt, nickel, cooper, zinc, gallium, strontium, yttrium, strontium, technetium, ruthenium, indium, hafnium, tungsten, rhenium, osmium, and bismuth.
  • radioisotopes include, but are not limited to, 64 Cu, 67 Cu, 67 Ga, 68 Ga, 88 Y, 89 Sr, 90 Y, 97 Ru, 99m Tc, 103 Ru, 111 In, 153 Sm, 186 Re, 188 Re, 203 Pb, 211 Bi, 212 Bi, 213 Bi, and 214 Bi.
  • the metal ions chosen to be chelated by the contrast agents depend in part on the diagnostic role of the metal ion.
  • Metals that can be incorporated, e.g., through chelation include lanthanides and other metal ions, including isotopes and radioisotopes thereof.
  • the preferred metal ion is gadolinium (III).
  • One of ordinary skill in the art can select a metal ion for chelation, based on the intended diagnostic application, without undue experimentation.
  • the choice of metal ions to be held in chelate complexes by the contrast agents of the disclosure depends upon the diagnostic technique for which the agent is to be used.
  • the metal ions should be paramagnetic, and preferably non-radioactive.
  • heavy metal ions e.g., with atomic numbers of at least 37, and in an embodiment, at least 50, should be used, again preferably non-radioactive species.
  • the metal ions should be ions of radioactive isotopes.
  • chelating groups For MR, X-ray, EIT or magnetometric imaging, one may use chelating groups to bind to heavy metal clusters (e.g., polyoxoanions and full or partial sulfur analogues) or to iron oxides or other superparamagnetic polyatomic species.
  • heavy metal clusters e.g., polyoxoanions and full or partial sulfur analogues
  • iron oxides or other superparamagnetic polyatomic species e.g., iron oxides or other superparamagnetic polyatomic species.
  • Metal may be incorporated into the contrast agent, i.e., the tailored binding sites, by direct incorporation, template synthesis, and transmetallation.
  • the metal ion is chelated into the contrast by direct incorporation, which involves titration with solution of sub-stoichiometric levels up to full incorporation.
  • one or two or more metal ions can bind to the contrast agent.
  • the contrast agent includes one or two or more metal ion binding sites.
  • each of the metal ion binding sites binds to the same metal ion.
  • each of the metal ion binding sites binds to a different metal.
  • Embodiments of the contrast agents can be used in any one of a number of methods.
  • Embodiments of this disclosure include, but are not limited to: methods of detecting, studying, monitoring, evaluating, and/or screening, diseases, conditions, and other biological events in vivo or in vitro.
  • the conditions can include, but are not limited to, altered growth rate of tissues, cancerous transformation of tissues, inflammation or infection of a tissue, altered volume of a tissue, altered density of a tissue, altered blood flow in a tissue, altered physiological function, altered metabolism of a tissue, loss of tissue viability, presence of edema or fibrosis in a tissue, altered perfusion in tissue, and combinations thereof.
  • embodiments of the present disclosure include: methods of imaging tissue; methods of diagnosing the presence of a disease, precancerous cells or tissue, cancer cells or tissue cancer, and tumors, as well as related biological events; methods of monitoring the progress of a disease, precancerous cells or tissue, cancer cells or tissue cancer, and tumors, as well as related biological events; and the like.
  • Embodiments of the present disclosure include, but are not limited to, imaging, detecting, studying, monitoring, evaluating, and/or screening biological materials (e.g., organs, tissues, tumors, cells, and the like), in vivo or in vitro.
  • biological materials e.g., organs, tissues, tumors, cells, and the like
  • the tissue types that can be studied using the methods of the present disclosure include, but are not limited to, myocardial tissues, nervous tissue, lymphoid tissue, skeletal and smooth muscle tissue, bones and cartilages, tissues of various organs (e.g., the kidney, the liver, the spleen, the prostate, the uterus, the testicles, and the ovaries), and select portions of each.
  • Embodiments of the methods can use one or more types of detecting or imaging systems such as, but not limited to, magnetic resonance imaging (MRI), SPECT, PET, ultrasound, X-ray, CAT, optical imaging, and combinations thereof.
  • the contrast agent is a multimodality contrast agent that includes a polymer having optical properties (GFP).
  • GFP polymer having optical properties
  • the polymer having optical properties can be detected using optical imaging, while the metal can be detected using another technique such as a MRI system.
  • embodiments of the contrast agent are administered to a host using one or more techniques or routes (e.g., oral, mucosal, parenteral, and the like).
  • the host can be introduced to an appropriate detection or imaging system.
  • the detection or imaging system can detect the contrast agent.
  • the detection or imaging system can detect the location(s) of the contrast agents, the concentration of the contrast agent, and the like.
  • the information obtained from the detection or imaging system can be used to create or form an image of the host or a portion thereof. The image would include the position and/or concentration of the contrast agent in the host.
  • the contrast agents can be used to study, image, diagnose the presence of, and/or treat cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors.
  • the presence and location of the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors can provide insight into the appropriate diagnosis and/or treatment.
  • contrast agents could include targeting agents specific for other diseases or conditions so that other diseases or conditions can be imaged, diagnosed, and/or treated using embodiments of the present disclosure.
  • other diseases and/or conditions can be studied, imaged, diagnosed, and/or treated in a manner consistent with the discussion below as it relates to cancerous cells, precancerous cells, cancer, and/or tumors.
  • the contrast agent can be used to study, image, diagnose the presence of, and/or treat cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors.
  • studying, imaging, diagnosing, and/or treating cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors in a host the contrast agent is administered to the host in an amount effective to result in uptake of the contrast agent into the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors.
  • the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors that takes up the contrast agent is detected using an appropriate imaging system.
  • Embodiments of the present disclosure can non-invasively image the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors throughout a host.
  • the contrast agent includes a targeting agent having an affinity for a specific cancer. Detecting the presence of the contrast agent, in particular, the presence of the contrast agent at the typical location of the specific cancer can be used in the diagnosis of the presence of the cancer (or vice versa). Imaging the host over a time period (e.g., days, weeks, months, or years) can provide information about the progression of the cancer or other disease or condition.
  • the contrast agent or compositions including the contrast agent may be administered to a subject in an amount effective to achieve the desired result at the appropriate dosages and for the desired periods of time.
  • An effective amount of the contrast agent or compositions may vary according to factors such as the age, body weight, general health, sex, and diet of the host; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; the existence of other drugs used in combination or coincidental with the specific compositions employed; the ability of the composition to elicit a desired response in the subject; and like factors well known in the medical arts.
  • An effective amount is also one in which any toxic or detrimental effects (e.g., side effects) of the contrast agent or compositions are outweighed by the therapeutically or diagnostically beneficial effects.
  • the contrast agent or compositions of the disclosure may be administered at a concentration of, for example, about 1 to 3.0 ⁇ mole/kg or about 6-20 mM.
  • Unit dosage forms of the contrast agents of this disclosure may be suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., intramuscular, subcutaneous, intravenous, intra-arterial, or bolus injection), topical, or transdermal administration to a patient.
  • mucosal e.g., nasal, sublingual, vaginal, buccal, or rectal
  • parenteral e.g., intramuscular, subcutaneous, intravenous, intra-arterial, or bolus injection
  • topical e.g., topical, or transdermal administration to a patient.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as hard gelatin capsules and soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • suspensions e.g.,
  • composition, shape, and type of dosage forms of the contrast agents of the disclosure typically vary depending on their use.
  • a parenteral dosage form may contain smaller amounts of the active ingredient than an oral dosage form used to treat the same condition or disorder.
  • compositions including the contrast agent and dosage forms of the compositions of the disclosure can include one or more excipients.
  • Suitable excipients are well known to those skilled in the art of pharmacy or pharmaceutics, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient.
  • oral dosage forms such as tablets or capsules, may contain excipients not suited for use in parenteral dosage forms.
  • the suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients can be accelerated by some excipients, such as lactose, or by exposure to water. Active ingredients that include primary or secondary amines are particularly susceptible to such accelerated decomposition.
  • compositions including the contrast agent and dosage forms of the compositions of the disclosure that can include one or more compounds that reduce the rate by which an active ingredient will decompose.
  • Such compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • pharmaceutical compositions or dosage forms of the disclosure may contain one or more solubility modulators, such as sodium chloride, sodium sulfate, sodium or potassium phosphate, or organic acids.
  • An exemplary solubility modulator is tartaric acid.
  • the amounts and specific type of active ingredient in a dosage form may differ depending on various factors. It will be understood, however, that the total daily usage of the compositions of the present disclosure will be decided by the attending physician or other attending professional within the scope of sound medical judgment.
  • the specific effective dose level for any particular host will depend upon a variety of factors, including for example, the activity of the specific composition employed; the specific composition employed; the age, body weight, general health, sex, and diet of the host; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; the existence of other drugs used in combination or coincidental with the specific composition employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of the composition at levels lower than those required to achieve the desired effect and to gradually increase the dosage until the desired effect is achieved.
  • kits which may include, but are not limited to, a contrast agent and directions (instructions for their use (written or electronic)).
  • the components listed above can be tailored to the particular disease or condition to be monitored.
  • the kit can further include appropriate reagents known in the art for administering various combinations of the components listed above to the host organism or patient.
  • This Example describes the rational design of a novel class of magnetic resonance imaging contrast agents with an engineered protein chelated with gadolinium.
  • the design of protein based contrast agents involves creating high coordination Gd 3+ binding sites in a stable host protein using amino acid residues and water molecules as metal coordinating ligands. Designed proteins show strong selectivity for Gd 3+ over physiological metal ions such as Ca 2+ , Zn 2+ , and Mg 2+ . These agents exhibit a 20-fold increase in longitudinal and transverse relaxivity values over the current clinically used contrast agent, Gd-DTPA. They provide strong contrast enhancement in vivo with much longer vascular retention time. These protein contrast agents have good biocompatibility and potential functionalities may extend MRI applications in targeting disease markers.
  • Magnetic resonance imaging is a non-invasive technique providing high resolution, three-dimensional images of morphological features as well as functional and physiological information about tissues in vivo. It is capable of detecting abnormalities in deep tissues and allows for whole body imaging. It has emerged as a primary diagnostic imaging technique for human diseases.
  • 1,2 Exogenous MRI contrast agents are often used to enhance the contrast between pathological and normal tissues by altering the longitudinal and transverse (i.e., T 1 and T 2 ) relaxation times of water protons.
  • 3-5 Gadolinium (Gd 3+ ) is the most frequently used MRI contrast agent due to its high magnetic moment, asymmetric electronic ground state and potential for increased MRI intensity.
  • the relaxivity (unit capability of the agent to change the relaxation time) of a contrast agent is dependent on several factors including the number of water molecules in the coordination shell, the exchange rate of the coordinated water with the bulk water, and the rotational correlation time ⁇ R of the contrast agent. 8-10 .
  • the MRI contrast agent can have: 1) high relaxivity for high contrast-to-noise ratio (CNR) and dose efficiency, 2) thermodynamic stability, especially metal selectivity for the target ions over excess physiological metal ions, to minimize the release of toxic paramagnetic metal ions, 3) adequate vascular, tissue retention time to allow imaging, and 4) proper excretion from the body.
  • Gd-DTPA the most commonly used MRI contrast agent in diagnostic imaging
  • Gd-DTPA-BMA the most commonly used MRI contrast agent in diagnostic imaging
  • ⁇ R the intrinsic rotational correlation time
  • these small molecular gadolinium contrast agents have longitudinal and transverse proton relaxivities, r 1 and r 2 , less than 10 mM ⁇ 1 s ⁇ 1 , much lower than the theoretically maximal value (>100 mM ⁇ 1 s ⁇ 1 ).
  • these small molecule contrast agents exhibit very short blood circulation (within several minutes) and tissue retention time, limiting some MRI applications that require longer data collection time.
  • This Example describes the development of a new class of MRI contrast agents with significantly improved relaxivity using rational design of Gd 3+ -binding proteins.
  • This class of contrast agents was created by designing the metal binding sites into a stable host protein with desired dynamic properties and metal selectivity to increase relaxivity by optimizing local ⁇ R . This approach provides a new platform for developing MRI contrast agents with high relaxivity and functionality.
  • Relaxation times, T 1 and T 2 were determined at 1.5, 3, 9.4 Tesla using a Siemens whole-body MR system (1.5, 3T) or a Bruker MRI scanner (9.4T). T 1 was determined using inversion recovery and T 2 using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence.
  • the contrast agent samples (200 ⁇ l) with different concentrations were placed in eppendorf tubes. The tubes were placed on a tube rack, which was placed in MRI scanners for the measurement of relaxation times.
  • the number of water ligands coordinated to Gd 3+ -CA1.CD2 complex was determined by measuring Tb 3+ luminescence decay in H 2 O or D 2 O.
  • Tb 3+ excited state lifetime was measured using a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Following excitation at 265 nm with a XenoFlash (Photon Technology International, Inc.), Tb 3+ emission was monitored at 545 nm in a time series experiment in both H 2 O and D 2 O systems.
  • Luminescence decay lifetime was obtained by fitting the acquired data with a mono-exponential decay function.
  • Gd 3+ -binding affinity of CA1.CD2 was determined by a competition titration with Fluo-5N applied as a Gd 3+ indicator.
  • the fluorescence spectra of Fluo-5N were obtained with a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Fluo-5N emission spectra were acquired at 500 nm to 650 nm with an excitation at 488 nm.
  • Gd 3+ -binding affinity of Fluo-5N, K d1 was first determined by a Gd 3+ titration with Gd 3+ buffer system of 1 mM nitrilotriacetic acid (NTA). Free Gd 3+ concentration was calculated with a NTA Gd 3+ -binding affinity of 2.6 ⁇ 10 ⁇ 12 M. 28 Fluo-5N was mixed with Gd 3+ in 1:1 ratio for a competition titration. The experiment was performed with a gradual addition of CA1.CD2. An apparent constant, K app , was estimated by fitting the fluorescence emission intensity of Fluo-5N at 520 nm with different CA1.CD2 concentrations as a 1 to 1 binding model. Gd 3+ -binding affinity of CA1.CD2, K d2 , was calculated with the following equation:
  • K d ⁇ ⁇ 2 K app ⁇ K d ⁇ ⁇ 1 K d ⁇ ⁇ 1 + [ Fluo - 5 ⁇ N ] T ( 1 )
  • mice 25-30 g, four mice were imaged
  • the anesthetized animal was positioned and stabilized with soft-supporting material (e.g., foam) in the scanner in the coil cradle and was kept warm during the MRI scan.
  • the mice were scanned prior to the administration of any contrast agent (pre-contrast).
  • any contrast agent pre-contrast.
  • Gd-CA1.CD2 ⁇ 1.2 mM
  • Gd-DTPA ⁇ 300 mM
  • T 2 weighted imaging at 9.4T MR images were recorded using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence. The data were collected and processed by Dicomworks software. The MR signal intensity in several organs was ascertained by the average intensity in ROIs or points within the organs. Signal intensity for each organ was normalized to that of the leg muscle.
  • CPMG Carr-Purcell-Meiboom-Gill
  • the organ/tissue samples were collected. Tissue extracts were freshly made from collected samples using commercially available tissue extracting kits (Qiagen).
  • CA1.CD2 was detected and quantified by immunoblotting and Sandwich-ELISA using a monoclonal antibody (OX45, detecting antibody) and a home made polyclonal antibody (PabCD2, capture antibody).
  • OX45 monoclonal antibody
  • PabCD2, capture antibody a home made polyclonal antibody
  • a series of known amounts of CA1.CD2 samples mixed with blank mouse serum or tissue extracts were used as standard in Sandwich-ELISA.
  • ELISA signal from HRP was monitored using a Fluorstar fluorescence microplate reader.
  • mice that received the i.v. administered Gd-CA1.CD2 (at a dose of ⁇ 2.4 ⁇ mol/kg) were returned to their cages (one mouse per cage). The mice were observed for five days and were euthanized at the end of the fifth day. Tissue samples from kidney, liver, spleen, and lung were collected. Gd 3+ ion contents in the tissue samples were analyzed by ICP-MS (see above paragraph).
  • mice Two groups of mice were used to examine potential renal and/or liver damage by Gd-CA1.CD2.
  • ALT Alanine transaminase
  • ALP Alkaline phosphatase
  • AST Aspartate transaminase
  • GTT Gamma glutamyl transpeptidase
  • bilirubin and urea nitrogen were analyzed by a commercially available source (MU Research Animal Diagnostic Laboratory). All clinical chemistry parameters were measured on an Olympus AU 400 analyzer.
  • Cytotoxicity was analyzed by MTT assay of the cells that were treated with Gd-CA1.CD2 at appropriate doses (indicated in figure).
  • the cells were grown under normal growth medium in 96 well plates.
  • Gd-CA1.CD2 or saline-phosphate buffer was added to the cell culture medium. The cells were incubated for appropriate times.
  • a standard MTT assay was employed to assess the cell growth status of the treated cells.
  • CA1.CD2 (40 ⁇ M) in complex with Gd 3+ was incubated with 75% human serum over 3 or 6 hours at 37° C.
  • the degradation of the protein was analyzed by SDS-PAGE and visualized by coomassie blue staining.
  • the degradation of the protein was also analyzed by immunoblot using antibodies OX54 or PabCD2.
  • the identities of the 12 kDa bands as CA1.CD2 were always verified by immunobloting using antibody PabCD2.
  • FIG. 1.1 shows the simulation of the dependence of r 1 and r 2 on the rotational correlation time, ⁇ R , of a contrast agent at different magnetic field strengths according to the theory developed by Blombergen, Solomon 6,7 (for detailed simulation procedures, please see on-line supporting materials).
  • ⁇ R the rotational correlation time
  • the relaxivity is ⁇ 10 mM ⁇ 1 s ⁇ 1 regardless of how the other parameters are adjusted.
  • the simulation clearly suggests that contrast agents with ⁇ R of 10-50 ns have the highest r 1 and r 2 values at clinically relevant magnetic field strengths from 0.47-4.7 Tesla (T).
  • high-relaxivity MRI contrast agents can be developed by directly designing Gd 3+ binding sites in proteins with desired ⁇ R . Coordinating Gd 3+ ions directly to the rigid protein frame eliminates the high internal mobility associated with chelator-macromolecule conjugates ( FIG. 1.1 a ).
  • CD2 rat CD2
  • a cell adhesion protein with a common immunoglobin fold as a scaffold
  • FIG. 1.1 c a cell adhesion protein with a common immunoglobin fold
  • CD2 protein exhibits strong stability against pH changes and excellent tolerance against various mutations, 29 which are essential features of functional protein engineering.
  • ⁇ R rotational correlation time
  • ⁇ R rotational correlation time
  • ⁇ 10 ns corresponding to optimal relaxivity for the current clinically allowed magnetic field strength.
  • its molecular size (12 kDa) is suitable for good tissue penetration and easy renal exclusion. 17
  • Gd 3+ binding sites into CD2 using computational methods.
  • 32,33 The design was based on the established structural parameters obtained from detailed analysis of metal binding sites in over 500 small chelators and metalloproteins.
  • Gd 3+ , Tb 3+ , La 3+ and other Ln 3+ ions have coordination properties similar to those of Ca 2+ with a strong preference for oxygen ligand atoms.
  • DTPA has 5 oxygen ligand atoms and 2 nitrogen ligand atoms.
  • the coordination atoms are almost always oxygen atoms, and the coordination numbers are lower than small chelators with an average of 7.2 for Ln 3+ and 6.0-6.5 for Ca 2+ . These effects are possibly due to steric crowding and sidechain packing. 34 Previously, we successfully designed Ca 2+ and Ln 3+ binding sites in a scaffold protein with strong selectivity over excess physiological metal ions. 35 Structure determination by solution NMR revealed that the actual coordination geometry in a designed variant is the same as our design, verifying the computational methods and the design strategy of metal-binding sites in proteins. 31
  • FIG. 1.1 c shows an example of designed Gd 3+ -binding protein CA1.CD2 with a metal binding site formed by the six potential oxygen ligands from the carboxyl side chains of Glu15, Glu56, Asp58, Asp62 and Asp64.
  • 36 we placed 5 negatively charged residues to provide these six oxygen ligand atoms in the coordination shell of CA1.CD2 to increase the selectivity for Gd 3+ over Ca 2+ .
  • one position of the metal binding geometry was left open in the design to allow fast water exchange between the paramagnetic metal ion and the bulk solvent ( FIG.
  • the Gd 3+ -binding site has minimal internal flexibility as the ligand residues originate from rigid stretches of the protein frame.
  • another Gd 3+ -binding protein, CA9.CD2 was engineered by fusing a continuous cation-binding EF-hand loop from calmodulin with flexible glycine linkers to the host protein. 37,38 This protein mimics previously reported highly flexible chelate-based contrast agents conjugated to macromolecules. 24,25
  • CA1.CD2 exhibited disassociation constants (K d ) of 7.0 ⁇ 10 ⁇ 13 , 1.9 ⁇ 10 ⁇ 7 , 6 ⁇ 10 ⁇ 3 , and >1 ⁇ 10 ⁇ 2 M for Gd 3+ , Zn 2+ , Ca 2+ , and Mg 2+ , respectively.
  • the selectivity K d ML /K d GdL for Gd 3+ over physiological divalent cations Zn 2+ , Ca 2+ , and Mg 2+ are 10 5.34 , >10 9.84 , and >10 10.06 , respectively.
  • the metal selectivity of CA1.CD2 is significantly greater than or comparable to that of FDA approved contrast agents DTPA- and DTPA-BMA 40 (Table 1).
  • Example 1 Metal binding constants (Log K a ) and metal selectivity of DTPA, DTPA-BMA and CA1.
  • CD2 Log Log Log Log Sample Gd 3+ Zn 2+ Ca 2+ Mg 2+ (K Gd /K Zn ) (K Gd /K Ca ) (K Gd /K Mg ) DTPA 28 22.45 18.29 10.75 18.20 4.17 11.70 4.25 DTPA-BMA 41 16.85 12.04 7.17 na* 4.81 9.68 na* CA1.CD2 12.06 6.72 ⁇ 2.22 ⁇ 2.0 5.34 >9.84 >10.06 *na: not available.
  • the designed Gd-binding proteins exhibit high r1 and r2 relaxivity.
  • FIG. 1.2 a shows that, at a concentration of 50 ⁇ M, the designed contrast agents Gd-CA1.CD2 and Gd-CA2.CD2 were able to introduce contrast enhancement in T1 weighted imaging at 3.0 T while 100 ⁇ M Gd-DTPA and protein CA1.CD2 alone did not lead to significant enhancement.
  • the in vitro relaxivity values of the designed Gd-binding proteins were measured (Table 2).
  • Gd-CA1.CD2 exhibits r 1 up to 117 mM ⁇ 1 s ⁇ 1 at 1.5T, about 20-fold higher than that of Gd-DTPA.
  • Gd-CA9.CD2 which carries a flexibly-conjugated Gd 3+ -binding site, had significantly lower relaxivity values (3.4 and 3.6 mM ⁇ 1 s ⁇ 1 , for r1 and r2 respectively, at 3.0 T), that are comparable to those of Gd-DTPA (Table 2).
  • the r 1 and r 2 of Gd-CA1.CD2 exhibited an inverse relationship with the magnetic field strength (Table 2, Example 1).
  • the r 1 and r 2 of Gd-DPTA showed weak dependence on field strengths.
  • the magnetic field strength dependent changes in relaxivity are consistent with our simulation results based on the rotational ⁇ R of the contrast agent ( FIG. 1.1 b ).
  • the results showed that the protein contrast agent offers much higher relaxivities for MRI contrast enhancement at clinical magnetic field strengths (1.5-3.0 T).
  • the transverse relaxivity of designed contrast agent is very high (i.g. >50 mM ⁇ 1 s ⁇ 1 ) at 9.4T compared to Gd-DTPA, making it appropriate as a T 2 contrast agent (Table 2) at high fields.
  • r2 of our protein-based contrast agent is smaller than the currently used r2 agents such as iron oxides. 42 This property allows our contrast agents to fill a gap between small Gd-chelators and iron oxide nanoparticles, extending the range of MRI applications both at clinically relevant field strength and possibly higher field strength.
  • the hydration number of an MRI contrast agent is another determinant for r 1 and r 2 .
  • the hydration number of the designed protein-based contrast agents was determined by measuring the luminescence lifetime of Tb 3+ . 26
  • the free Tb 3+ in H 2 O and D 2 O has a life time value of 410 ⁇ s and 2,796 ⁇ s, respectively.
  • the formation of M-protein complex significantly increases Tb 3+ life time to 859 ⁇ s.
  • the Tb 3+ life time values of CA1.CD2 were 1,679 ⁇ s in D 2 O, suggesting a hydration number of 2.1 ( FIG. 1.3 b ).
  • a well-known Ca 2+ -binding protein troponin C exhibits a hydration number of 1.8 ( FIG.
  • Gd-CA1.CD2 was sustained over 4-7 hours at multiple organs ( FIG. 1.8 b ), indicating much longer tissue retention time of the agent than that of Gd-DTPA.
  • the tissue retention and blood circulation time of Gd-CA1.CD2 in mice were characterized by administering various doses of agents in mice and analyzing the collected blood samples or tissue sections from sacrificed animals using immunoblots and ELISA with monoclonal (OX45) and in-house developed polyclonal (PabCD2) antibodies.
  • OX45 monoclonal
  • PabCD2 polyclonal
  • Gd-CA1.CD2 exhibited a prolonged blood circulation time. No significant decrease in the CA1.CD2 levels in blood was observed until 45 minutes after i.v. administration.
  • the protein remained in blood circulation for more than 3 hours ( FIG. 1.8 a ). This property is considered for imaging of biological events that require prolonged imaging time, or imaging of pathological features that require time for delivery of the agent to the targeted site.
  • CA1.CD2 was first detectable at 15 minutes and peaked at 4-5 hours. There was less than 10% of the injected dose of the contrast agent remaining in the kidney 15 hours after injection (by measurements of both Gd 3+ and CA1.CD2). This result, along with the observation of MRI contrast changes in the bladder, suggests a clearance of the agent by kidney.
  • Gd-CA1.CD2 did not exhibit acute toxicity at the dose ( ⁇ 2.4 ⁇ mole/kg) used for MRI. All mice that received the contrast agents (>10) showed no adverse effects before euthanization five days after agent injection. The effects of Gd-CA1.CD2 on liver enzymes (ALT, ALP, AST), serum urea nitrogen, bilirubin, and total protein from CD-1 mice 48 hours post-contrast injection were negligible compared to those in the control mice (Example 1, Table 3). In addition, no cytotoxicity was observed in tested cell lines, SW620, SW480 and HEK293, that were treated with 50 ⁇ M Gd-CA1.CD2, by MTT assay ( FIG. 1.8 b ). Based on the preliminary characterization of toxicity, we conclude that the protein contrast agent did not possess acute toxicity at current dosages for mice.
  • Example 1 Summary of Animal Clinical Pathology Profiles Test mice a Control mice b Normal rang c Urea Nitrogen (mg/L) 26.0 ⁇ 0.2 d 27.0 ⁇ 0.2 18-31 Total Urine Protein 5.2 ⁇ 0.2 5.3 ⁇ 0.2 5.9-10.3 (g/L) Total Bili (mg/L) 0.4 ⁇ 0.2 0.5 ⁇ 0.2 0.3-0.8 Direct Bili (mg/L) 0.0 0.0 0.0 ALT (U/L) 49.0 ⁇ 0.2 72.0 ⁇ 0.2 44-87 ALP (U/L) 115.0 ⁇ 0.2 302.0 ⁇ 0.2 43-71 AST (U/L) 280.0 ⁇ 0.2 219.0 ⁇ 0.2 101-214 GGT (U/L) 0.0 ⁇ 3.0 a Four mice per group were injected with Gd-CA1.CD2 at dose of 4.0 ⁇ mole/kg.
  • mice All clinical chemistry parameters are measured on an Olympus AU 400 analyzer by MU Research Animal Diagnostic Laboratory (details see Material and Methods).
  • c Normal range values are from Quesenberry, K. E. and J. W. Carpenter; Ferrets, Rabbits, and Rodents Clinical Medicine and Surgery; W. B. Saunders: Philadelphia, 2003.
  • Using protein to chelate Gd 3+ as an MRI contrast agent has several potential advantages over currently used Gd-DTPA in functional and molecular imaging applications: 1) it greatly increases the contrast-to-noise ratio (CNR); 2) it improves dose efficiency with reduced metal toxicity; 3) it prolongs the tissue retention time, which enables imaging of abnormalities that requires prolonged tissue enhancement; and 4) provide a potential functioning protein or a protein carrier that can conjugate target specific ligands to a biomarker for targeted molecular MR imaging.
  • CNR contrast-to-noise ratio
  • R 1 cq 55.5 ⁇ 1 T 1 ⁇ m + ⁇ m ( 1 )
  • R 2 cq 55.5 ⁇ T 2 ⁇ m - 2 + ⁇ m - 1 ⁇ T 2 ⁇ m - 1 + ⁇ ⁇ ⁇ ⁇ m 2 ⁇ m ⁇ ( ( ⁇ m - 1 + T 2 ⁇ m - 1 ) 2 + ⁇ ⁇ ⁇ ⁇ m 2 ) ( 2 )
  • Equation 2 the water coordination number, q, is assumed to be 1 and the agent concentration is 0.001 M; ⁇ m is the dwelling time of the coordination water; and ⁇ m is the chemical shift difference between the bound and free water. Since ⁇ m 2 is much smaller than other components, equation 2 is simplified to supplementary equation 3 and used in this simulation:
  • T im is determined by dipole-dipole (DD) and scalar or contact (SC) mechanisms as shown in supplementary equation 4:
  • the frequency of proton ⁇ 1 equals to the ⁇ 1 multiplied by the magnetic field while the frequency of electron ⁇ s is 658-fold of ⁇ 1 .
  • the ⁇ ci is determined by the rotational correlation time ⁇ R , the water dwelling time ⁇ m , and T ie as shown in supplementary equation 7:
  • T ie is related to the electron frequency ⁇ s as well as the ⁇ v (correlation time of splitting) and ⁇ 2 (mean square zero field splitting energy) of the Gd 3+ as in supplementary equations 8-10:
  • ⁇ R (1 ps, 10 ps, 100 ps, 1 ns, 10 ns, and 100 ns
  • ⁇ m (1 ps, 10 ps, 100 ps, 1 ns, 10 ns, and 100 ns
  • ⁇ v (1 and 10 ps)
  • ⁇ 2 10 17 , 10 18 , 10 19 , and 10 20 s ⁇ 2
  • the relaxivity is ⁇ 10 mM ⁇ 1 s ⁇ 1 no matter how the other parameters are adjusted.
  • the relaxivity can reach a much higher level by adjusting other parameters such as the ⁇ m .
  • the protein contrast agent Gd-CA1.CD2 did not exhibit acute toxicity at the MRI imaging dose ( ⁇ 2.4 mole/kg), as demonstrated by the fact that all MR imaged mice that received the contrast agent (>10) behaved normally and remained healthy (sacrificed five days after agent injection).
  • the effects of Gd-CA1.CD2 on liver enzymes (ALT, ALP, AST), serum urea nitrogen, bilirubin, and total protein from CD-1 mice 48 hours post-contrast injection were negligible compared to the control mice (Table 3).
  • no cytotoxicity was observed in tested cell lines, SW620, SW480 and HEK293 that were treated with 50 ⁇ M Gd-CA1.CD2, by MTT assay ( FIG. 1.7 b ). Based on the preliminary characterization of toxicity, we conclude that the protein contrast agent is relatively safe.
  • PEGylation of protein is a method used to improve the pharmacokinetics and pharmaco-dynamics of various protein and peptide drugs.
  • PEGylation involves modifications of Lys, Glu, Asp, or Cys residues of a protein or peptide with various sizes of polyethylene glycerol chain.
  • the result of PEGylation modifications is the attachment of the different size of polyethylene glycerol chains on the surface of the modified protein or peptide.
  • the protein or peptide experiences several property changes, especially in pharmacokinetics and pharmaco-dynamics. Two changes are obvious: (1) the increase in molecular size, especially in the case of small peptide, and (2) the reduction in surface charges of protein and peptide.
  • Protein was expressed in E. coli as inclusion body and purified using urea refolding and ion-exchange column. N15 labeled protein was expressed in SV medium as GST fusion (Yang et al., Biochem 2006) and purification was by GST-4B affinity column and SP column. Protein was verifed by mass spectrometry. Protein concentration was calculated using extinction coefficient of w.t. CD2 of 11,000 (Ye et al., 2001). Protein solubility was determined by concentrating proteins to reach to precipitation using speed vac.
  • ESI and MALDI spectrometry were used to identify the number of pegylation sites and metal binding stoichiometry.
  • the pegylation sites were identified using trypsin cleavage followed by Mass analysis using TOF/TOF.
  • Gd 3+ -binding affinities of CA1.CD2 and its pegylated variants were determined by a competition titration with Fluo-5N applied as a Gd 3+ indicator (Yang et al., JACS, 2008).
  • the fluorescence spectra of Fluo-5N were obtained with a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Fluo-5N emission spectra were acquired at 500 nm to 650 nm with an excitation at 488 nm.
  • Gd 3+ -binding affinity of Fluo-5N, K d1 was first determined by a Gd 3+ titration with Gd 3+ buffer system of 1 mM nitrilotriacetic acid (NTA). Free Gd 3+ concentration was calculated with a NTA Gd 3+ -binding affinity of 2.6 ⁇ 10 ⁇ 12 M. 28 Fluo-5N was mixed with Gd 3+ in 1:1 ratio for a competition titration. The experiment was performed with a gradual addition of CA1.CD2 or its variants. An apparent constant, K app , was estimated by fitting the fluorescence emission intensity of Fluo-5N at 520 nm with different CA1.CD2 concentrations as a 1 to 1 binding model. Gd 3+ -binding affinity of CA1.CD2, K d2 , was calculated with the following equation:
  • K d ⁇ ⁇ 2 K app ⁇ K d ⁇ ⁇ 1 K d ⁇ ⁇ 1 + [ Fluo - 5 ⁇ N ] T ( 1 )
  • the numbers of water ligands coordinated to Gd 3+ -CA1.CD2 and variants complex were determined by measuring Tb 3+ luminescence decay in H 2 O or D 2 O (Yang et al., 2008).
  • Tb 3+ excited state lifetime was measured using a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Following excitation at 265 nm with a XenoFlash (Photon Technology International, Inc.), Tb 3+ emission was monitored at 545 nm in a time series experiment in both H 2 O and D 2 O systems.
  • Luminescence decay lifetime was obtained by fitting the acquired data with a mono-exponential decay function.
  • Pulse field diffusion NMR was applied to measure the hydrodynamic radii of the protein (Lee et al., BBA 2003) with Protein samples CA1.CD2 and its variants of 0.2 mM in buffer. Lysozme and diaxone were used as an external and internal reference for calibration. The correlation size of the protein was measured using TauC pulse sequence developed by the Prestegard lab at UGA using 15 N labeled protein.
  • CD-1 mice 25-30 g, four mice were imaged
  • the anesthetized animal was positioned and stabilized with soft-supporting material (e.g. foam) in the scanner in the coil cradle and was kept warm during the MRI scan.
  • the mice were scanned prior to the administration of any contrast agent (pre-contrast).
  • any contrast agent pre-contrast.
  • Gd-CA1.CD2 ⁇ 1.2 mM
  • Gd-DTPA ⁇ 300 mM
  • T 2 weighted imaging at 9.4T MR images were recorded using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence. The data were collected and processed by Dicomworks software. The MR signal intensity in several organs was ascertained by the average intensity in ROIs or points within the organs. Signal intensity for each organ was normalized to that of the leg muscle.
  • CPMG Carr-Purcell-Meiboom-Gill
  • CD-1 mice 25-30 g were anesthetized with isoflurane. Appropriate dosages of Gd-CA1.CD2 and its PEgylated variants (with Gd and 153 Gd) or Gd-DTPA were i.v. injected (via tail vein). Blood ( ⁇ 50 ⁇ l) samples were collected via orbital sinus of the mouse at different time points. The mouse was euthanized at the final time point. Tissue samples from kidney, liver, heart, and lung were collected. Serum samples were prepared from the collected blood. For the bio-distribution analyses, the animals were euthanized at single time point (indicated) after i.v. administration of the contrast agent (indicated). The organ/tissue samples were collected.
  • Tissue extracts were freshly made from collected samples using commercially available tissue extracting kits (Qiagen).
  • CA1.CD2 and its variants were detected and quantified by immunoblotting and Sandwich-ELISA using a monoclonal antibody (OX45, detecting antibody) and a home made polyclonal antibody (PabCD2, capture antibody).
  • OX45 monoclonal antibody
  • PabCD2, capture antibody a home made polyclonal antibody
  • a series of known amounts of CA1.CD2 samples mixed with blank mouse serum or tissue extracts were used as standard in Sandwich-ELISA.
  • ELISA signal from HRP was monitored using a Fluorstar fluorescence microplate reader.
  • Rabbits were i.p. injected with Gd-CA1.CD2 and its pegylated variants.
  • the agents (CA1.CD2) were mixed with adjuvant or with buffer saline and injected at a dose of 3.0 nmole/kg according to the standard protocol for the antibody production.
  • the rabbits were subjected to double immunolizations in four weeks interval. Blood samples were taken from the immunolized rabbits 3 weeks after each injection. Production of antibodies against the protein contrast agent in each rabbit was examined by ELISA
  • the MR imaged CD-1 mice that received the i.v. administered Gd-CA1.CD2 and its variants (at a dose of ⁇ 2.4 ⁇ mol/kg) spiked with 157 Gd 3+ were returned to their cages (one mouse per cage). The mice were observed for five days and were euthanized at the end of the fifth day. Tissue samples from kidney, liver, spleen, and lung were collected. Gd 3+ contents in the tissue samples were analyzed by radioactivity counter.
  • mice Two groups of mice were used to examine potential renal and/or liver damage by Gd-CA1.CD2.
  • ALT Alanine transaminase
  • ALP Alkaline phosphatase
  • AST Aspartate transaminase
  • GTT Gamma glutamyl transpeptidase
  • bilirubin and urea nitrogen were analyzed by a commercially available source (MU Research Animal Diagnostic Laboratory). All clinical chemistry parameters were measured on an Olympus AU 400 analyzer.
  • Cytotoxicity was analyzed by MTT assay of the cells that were treated with Gd-CA1.CD2 and its variants at appropriate doses (indicated in figure).
  • the cells were grown under normal growth medium in 96 well plates.
  • Gd-CA1.CD2 or saline-phosphate buffer was added to the cell culture medium. The cells were incubated for appropriate times.
  • a standard MTT assay was employed to assess the cell growth status of the treated cells.
  • CA1.CD2 (40 ⁇ M) in complex with Gd 3+ was incubated with 75% human serum over 3 or 6 hours at 37° C.
  • the degradation of the protein (disappearance of 12 kDa protein band) was analyzed by SDS-PAGE and visualized by coomassie blue staining for protein and idiol staining for PEG moiety.
  • the degradation of the protein was also analyzed by immunoblot using antibodies OX54 or PabCD2.
  • the identities of CA1.CD2 and pegylated variants were verified by immunobloting using antibody PabCD2 and idiol staining.
  • the contrast agents exhibit a 20 fold increase in R1 and R2 relaxivities and provide a strong contrast enhancement in the mouse MR imaging (Yang, et. al. JACS 2008).
  • the developed MRI contrast agent also demonstrated a prolonged blood circulation time, which is considered for the application of the agents in disease targeted molecular imaging.
  • DTPA 300-500 mM
  • the solubility of the current formulation of our protein contrast CA1.CD2 is ⁇ 0.7 mM.
  • the designed contrast agent CA1.CD2 has eight Lys residues with different solvent accessibility and six Lys residues are well exposed. Based on the considerations of solubility, circulation, relaxivity and serum stability, we therefore carried out PEGylation of our protein contrast agents by modifying surface Lys residues and site specific pegylation. First, preactivated PEG units with varied chain length and branches of 4, 12 (Poly-PEG 12 ), 40 (PolyPEG 40k ), 5K, and 12 K (see FIG. 2.1 b ) were used to modify surface lys residues. Site-specific pegylation was performed at both N-terminal Lys and C-terminal Cys.
  • FIGS. 2 . 2 - 2 . 4 shows the SDS gel of the protein contrast agent CA1.CD2 pegylated with PEGylation kits with different reaction moieties stained by both commassie blue for protein and idiol for the PEG moiety.
  • FIG. 2.2 shows that the PEGylated proteins were separated using ion exchange column, size exchange column, and a C 18 reverse phase HPLC chromatography column. The modified proteins were purified by HPLC to relative homogeneity.
  • FIG. 2.2 shows that there were several major peaks of the PEGylated proteins on the FPLC chromatography. MALD-Mass analyses of the separated protein fraction reveal that at pH 7.4 with 5:1 pEG:protein:preactivated pEG reagents, CA1.CD2 was pegylated mainly with 2, 3 with P4-P40 and 1 PEG unit with PEG 12 K to 20K.
  • the CA1.CD2 has total of seven potential PEGylation sites.
  • the PEGylation sites of the purified protein were examined first by trypsin cleavage to generate the peptides/fragments and then sequenced by TOF/TOF MS. Usually 2-4 PEG units were attached to each protein depending on the ratio of PEG:protein under reaction and the reaction conditions.
  • FIG. 2.4 a of Trp fluorescence spectra shows that these PEGylated proteins (CA1.CD2-PEG12 and CA1.CD2-PEG40) maintain the native structure of the protein contrast agents with unchanged Trp emission maximum compared with CA1.CD2.
  • Tb-FRET was first used to monitor the effect of pegylation on the metal binding capabilities.
  • the pegylated A1.CD2 is able to have Tb-sensitized energy transfer similar to that of CA1.CD2.
  • FIG. 2.5 shows that PEGylated CA1.CD2-P40 remains intact after incubate with human serum for 24 hours at 37° C. monitored by SDS Page.
  • Gd 3+ -binding affinity of CA1.CD2 was further determined by a competition titration with fluorescent dye applied as a Gd 3+ indicator in various chelate-metal buffer systems.
  • the PEGylated variants exhibit similar metal binding affinities to unpegylated ones for Ca 2+ , Gd 3+ and Zn 2+ .
  • FIG. 2.6 shows the R1 and R2 relaxivities of the PEGylated protein.
  • the R1 and R2 relaxivities of the PEGylated protein were also affected by the size of the PEG chains.
  • the relaxivities of the PEGylated protein contrast agents demonstrated higher increases when the protein was modified by a longer PEG chain.
  • both R1 and R2 relaxivities of the PEGylated protein contrast agent experienced the most dramatic increases at higher magnetic field. This is contrary to the case of unPEGylated protein, in which dramatic decreases in R1 and R2 relaxivities were observed at high field (Yang et al., JACS, 2008).
  • FIG. 2.6 shows the MRI relaxivity as a function of chain dependencies. Specific modification of the N-terminal amine and Cys residues at the C-terminus have a similar effect on the relaxivity of the protein.
  • Table 2.1 of Example 2 shows that the water numbers in the coordination shell of CA1.CD2 increased from 2 to 3 upon PEGylation with PEG40. This may contribute to the increased relaxivity by PEGylation.
  • PEGylation has been demonstrated to increase blood circulation time and reduce immunogenicity of a number of protein drugs. While longer blood circulation time is a desired property for the applications of MRI contrast agents. Elimination or reduction of immunogenicity is essential for clinical applications of the protein MRI contrast agents.
  • PEGylation of CA1.CD2 changed the bio-distribution and immunogenicity properties of the agent.
  • the PEGylated or unPEGylated CA1.CD2 was introduced to CD-1 mice via i.v. tail vein injection.
  • Immunogenicity is one of the main concerns on the application of our developed protein MRI contrast agent.
  • the agents (CA1.CD2) were mixed with adjuvant or with buffer saline and injected at a dose of 3.0 nmole/kg according to the standard protocol for antibody production.
  • the rabbits were subjected to double immunolizations in four weeks interval. Blood samples were taken from the immunolized rabbits 3 weeks after each injection. Production of antibodies against the protein contrast agent in each rabbit was examined by ELISA ( FIG. 2.7 Left) and Immunobloting ( FIG. 2.7 Right) using our previous polyclonal antibody PabCD2 as positive control and the pre-bleed from each rabbit as negative controls.
  • FIG. 2.7 shows that PEGylation with P40 completely eliminates the binding of CA1.CD2 by antibody OX55 and decreases by 50% binding of OX34 in Western Blot. Since the epitope binding sites of OX55 and OX34 are well known, we have shown that K66, K45, K44, and K47 exhibit a high probability for the specific PEGylation by PEG12. These results confirm that antibody recognition sites can be eliminated by PEGylation, which is essential for the reduction of immunogenicity.
  • Natural calcium binding proteins with continuous calcium binding sites such as calmodulin, calbindin D9K, troponin C, parvalbumin and discontinuous calcium binding sites such as thermintase subtilisin have exhibited high metal binding affinity for calcium as shown in Table 3.1 (shown in FIG. 3.10 ). Most of these calcium binding proteins have multiple calcium binding sites and their protein stabilities against temperature and proteolysis were significantly increased. For example, calcium binding to calmodulin has increased its stability to higher than 100° C. While a great deal of research has shown that the calcium binding affinity of naturally evolved proteins can be reduced by site-directed mutagenesis, methods to increase calcium binding affinity have rarely been reported.
  • CBPP and its variants exhibit strong metal selectivity for Ln3+ over calcium, magnesium, zinc and copper ( FIG. 3.2 c ). Furthermore, the water numbers in the coordination site can be estimated by Tb-sensitized energy transfer in water and in D 2 O. These data suggest that more than one water molecules are in the coordination shells and protein surface, and this likely contributes to their extremely high relaxivity.
  • FIG. 3.8 shows that calmodulin and its variants can be pegylated with peg units of 4, 12, 40, 5K, 12K, 20 K and reaction products can be well separated by size-exclusion and ion exchange columns ( FIG. 3.3 and FIG. 3.4 ).
  • FIG. 3.6 shows that both BCBP1 and CBBP1 and their variants have strong serum stability revealed by SDS page. These proteins remain intact upon incubation with serum greater than 48 hours.
  • Embodiment of the PEGyation method of the present disclosure was applied to enhance the performance of developed contrast agents in imaging cancer biomarker.
  • Example 4 describes a novel protein-based MRI contrast agent for molecular targeting cancer marker HER2 has been developed by modification with pegylation.
  • the MRI contrast agent also carries a near-IR dye Cy5.5 for dual modality imaging of HER2 in cancer (Section 4.1).
  • Example 4 describes the development of a contrast agent that demonstrates specific interaction with HER2 positive cancer cells but not HER2 negative cells (4.2).
  • the in vitro MR imaging experiments with different cells that express different levels of HER2 have shown a close correlation between MRI contrast enhancements and HER2 levels (Section 4.3).
  • Example 4 describes that the MR imaging of xenograft models of human HER2 positive cell line SKOV3 and HER2 negative cell line MDA-MB-231 has indicated a HER2 level-dependent MRI contrast enhancement. This image intensity enhancement is further confirmed by NIR imaging (Section 4.4).
  • Example 4 describes immunohistochemical staining of tissue samples (including tumor tissue) collected after imaging analyses has shown that the designed contrast agent penetrated deep into the tumor mass (i.e., distant from tumor vasculature), demonstrating a good tissue penetration of the MRI contrast agent.
  • Differential targeting of the MRI contrast agent to HER2 positive and to HER2 negative tumors was further verified by immunoblot and ELISA analyses of the tissue samples collected from imaging mice (Section 4.5).
  • PEGylation of the designed Gd-binding protein not only increases protein solubility and blood circulation time, but also decreases immunogenicity of the protein without decrease of MRI relaxivity of the protein contrast agent. Therefore the designed HER2 targeting protein contrast agent was PEGylated using PEG-40, a PEG molecule with triple-branched 12 units PEG.
  • the resulting agent (PEG-CA1-Affi) exhibits very similar Gd 3+ binding properties and its T1 and T2 relaxivity values are similar to its parent protein CA1.CD2. Modifications did not change the overall protein fold as demonstrated by circular dichroism and fluorescence spectra.
  • AU565 is derived from breast carcinoma.
  • EMT6 is a mouse mammary tumor cell line.
  • AU565 cells express very high levels of HER2.
  • the EMT is regarded as a HER2 negative cell line.
  • Binding of the Gd-CA1.HER2/Affi to the cancer cells was first analyzed by immuno-fluorescence staining using a polyclonal antibody against PEGylated parental protein CA1.CD2 (PAbPGCA1). A substantial increase in staining intensities of CA1-Affi bound to AU565 cells was observed at both 37 and 4° C.
  • the protein did not bind to EMT6 cells.
  • CA1.CD2 without HER2 affibody did not bind to either of the tested cultured cells ( FIG. 4.2 ).
  • Proteins binding to cell surface HER2 with a clear membrane staining pattern in AU565 cells was observed at 4° C.
  • the binding of the proteins to the cells triggered receptor-mediated endocytosis at 37° C. as demonstrated by the staining of the protein inside the cells.
  • PEGylation of the targeted contrast agent PEG-CA1-Affi does not change its target capability the positive cell as shown in FIG. 4.2 at both 4 and 37° C.
  • Binding of the Gd-CA1-Affi to the cancer cells was further analyzed by quantification of cell bound Gd 3+ with AU565 and EMT6 cells.
  • the amounts of Gd 3+ were quantified by ⁇ -counting the trace of isotope 153 Gd 3+ in the Gd-protein complexes.
  • the quantification of cell bound Gd 3+ supported our immuno-analyses that Gd-CA1-Affi exhibited targeted binding to AU565 cells but not to EMT6 cells ( FIG. 4.3 ).
  • the protein without the HER2 affibody targeting moiety could not bind to the cells that express HER2.
  • mice were imaged using a Kodak in vivo FX-pro animal imaging system. Consistent with MRI imaging, we observed strong NIR light emission from both positive and negative tumor sites at early time points (50 minutes post-injection).
  • FIG. 4.4 shows the result of another mouse.
  • 5 mM of contrast agent CA1.Affi-P40 (100 fold lower than clinic used DTPA) was injected via tail vein.
  • MRI images at 4.7 T using fast spin echo were acquired before injection, 5 min, 30 min, 3 hr, 24 hr and 52 hr post injection.
  • Positive tumor shows a strong contrast after 30 mins and peaked at 24 hour with about 35% enhancement. Contrast capability was decreased after 52 hours, suggesting that the contrast agent was secreted of out the animal. This mouse was alive and looks normal after 52 hours MRI scanning.
  • Protein contrast agent Targeting of the protein contrast agent to HER2 positive tumor was further analyzed by immunoblot.
  • protein extracts were made from the tissue samples that were collected from the imaged mice. Immunoblot experiments were performed with the protein extracts using the antibody PAbPGCA1. Consistently, we detected very high levels of PEGylated protein contrast agent in extracts made from liver, kidney, and positive tumor. The antibody detected very faint bands in the extracts made from muscle and negative tumor samples ( FIG. 4.6 ). The results strongly suggested that our protein contrast agent led to a HER2 specific MR image enhancement.
  • CBP1-affi calmodulin
  • CBP1-Bom calmodulin
  • These targeted contrast agents were then PEGylated by PEG with different units and purified.
  • Initial binding to HER2 has been studied.
  • the HER2 positive cell line SKOV-3 and HER2 negative cell line MDA-MB-231 was treated with CBP1-affi at different concentration: 10 ⁇ M and 20 ⁇ M in 1 ml medium. Both of the cell numbers is about 1 ⁇ 10 5 .
  • FIG. 4.8 shows that the bands in SKOV-3 cells are much darker than those in MDA-MB-231 cells, which means the binding of CBP1-affi to HER2 is specific.
  • FIG. 4.9 shows the HER2 positive cell line SKOV-3 has been stained. The staining on the negative cell line is rarely.
  • FIG. 4.9 shows the uptake and internalization of CA1.Bom (CA1.CD2-52I-Bom) specifically to GRPR positive cell lines (PC-3 and DU-145 cells) by using confocal fluorescent microscope.
  • DU-145, PC-3 and H441 cells (8 ⁇ 10 4 ) were seeded in 4-well chambers (BD) at 37° C. overnight. In the second day, fresh medium was changed. Contrast agents were incubated with cells at different time point at 37° C. Subsequently, the cells were washed with PBS for three times and fixed with 3% formaldehyde for 15 minutes. After cells were rinsed in PBS for three times, 0.2% Triton X-100 was added to permeablize cells for 10 minutes. The cells were washed and four drops of Image-IT Fx signal enhancer (Invitrogen, CA) was applied in each sample to block unspecific binding. After three time washes, CD2 antibody was used as primary antibody. Goat anti-mouse IgG conjugated with Alexa Fluor 488 was used as detection antibody (Invitrogen, CA). The fluorescence image was acquired using 488 nm and UV lasers.
  • FIG. 4.11 shows NIR-fluorescence imaging of nude mice xenografted with DU-145 tumor (positive control left) and H441 tumor (control, right) post injection of CA1.CD2-52Ibom-cy5.5-P40 26 hours via tail vein. 50 uM Cy5-CA1.CD2-52I was injected into mouse tail vein. After 26 hours, mouse was analyzed by Kodak Imaging System. Tissue organs were taken out and analyzed. Tumor intensity was analyzed by Image J.
  • FIG. 4.12 shows NIR imaging (top) and NIR intensity (bottom) of CA1.CD2-52I-Bom-Cy5.5-P40 at different organs of the mice. The developed contrast agent is able to target to the GRPR expressed tumor monitored by NIR fluorescence.
  • ratios, concentrations, amounts, and other numerical data may be expressed herein in a range format. It is to be understood that such a range format is used for convenience and brevity, and thus, should be interpreted in a flexible manner to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited.
  • a concentration range of “about 0.1% to 5%” should be interpreted to include not only the explicitly recited concentration of about 0.1 wt % to about 5 wt %, but also include individual concentrations (e.g., 1%, 2%, 3%, and 4%) and the sub-ranges (e.g., 0.5%, 1.1%, 2.2%, 3.3%, and 4.4%) within the indicated range.
  • the term “about” can include ⁇ 1%, ⁇ 2%, ⁇ 3%, ⁇ 4%, ⁇ 5%, ⁇ 6%, ⁇ 7%, ⁇ 8%, ⁇ 9%, or ⁇ 10%, or more of the numerical value(s) being modified.
  • the phrase “about ‘x’ to ‘y’” includes “about ‘x’ to about ‘y’”.
  • sequences are representative examples from protein families where sequences of the same protein across different species typically share 50-98% sequence homology.
  • attributes in the cited sequence can be inferred to apply to homologous, related sequences, which would be known to someone familiar with the art.
  • ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN 60 D93A ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN 60 D129A
  • ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEV A ADGN 60 D20A ADQLTEEQIAEFKEAFSLF A KDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN 60 Y99W (6)
  • Rat N-Calmodulin N-Terminal Domain of Calmodulin and Variants
  • N-CaM (11) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN 60 N-N6OD (12) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGD 60 N-CaM GTIDFPEFLTMMARK 75 N-N6OD GTIDFPEFLTMMARK 75
  • Rat C-Calmodulin N-Terminal Domain of Calmodulin and Variants
  • ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADDL 60 Mouse (18) ADQLTEEQIAEFKEAFSLFDKDGDNTITTKELGTVMRSLGQNPTEAELQDMINEVDAD-- 58 Rat (19) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDAD-- 58 Rabbit (20) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDAD-- 58 Paramecium (21) AEQLTEEQIAEFKEAFALFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDAD-- 58 Human PGNGTIDFPEFLTMMARKMKDTDSEEEIREAFRVEDKDGNGYISAAELRHVMTNLGEKLT 120 Mouse -GNGTIDFPEFLTMMARKM
  • CA1-WT (22) GSRDSGTVWGALGHGIELNIPNFQMTDDIDEVRWERGSTLVAEFKRKMKPFLKSGAFEID 60 CA1-Z HER2-4 (23) GSRDSGTVWGALGHGIELNIPNFQMTDDIDEVRWERGSTLVAEFKRKMKPFLKSGAFEID 60 CA1-Z HER342 (24) GSRDSGTVWGALGHGIELNIPNFQMTDDIDEVRWERGSTLVAEFKRKMKPFLKSGAFEID 60 CA1-WT ANGDLDIKNLTRDDSGTYNVTVYSTNGTRILNKALDLRILEGGSGGVDNKENKEQQNAFY 120 CA1-Z HER2-4 ANGDLDIKNLTRDDSGTYNVTVYSTNGTRILNKALDLRILEGGSGGVDNKFNKE LRQAYW 120 CA1-Z HER342 ANGDLDIKNLTRDDSGTYNVTVYSTNGTRILNKALDLRILEGGSGGVDNKFNKE MRNA

Abstract

Embodiments of the present disclosure provide for contrast agents, methods of making contrast agents, and methods of using contrast agents, and the like.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims priority to copending U.S. provisional application entitled, “CONTRAST AGENTS, METHODS FOR PREPARING CONTRAST AGENTS, AND METHODS OF IMAGING,” having Ser. No. 61/041,693, filed Apr. 2, 2008, which is entirely incorporated herein by reference.
  • BACKGROUND
  • Magnetic resonance imaging (MRI) is a non-invasive technique providing high resolution, three-dimensional images of morphological features as well as functional and physiological information about tissues in vivo. It is capable of detecting abnormalities in deep tissues and allows for whole body imaging. It has emerged as a primary diagnostic imaging technique for human diseases.
  • Exogenous MRI contrast agents are often used to enhance the contrast between pathological and normal tissues by altering the longitudinal and transverse (i.e., T1 and T2) relaxation times of water protons. The relaxivity (unit capability of the agent to change the relaxation time) of a contrast agent is dependent on several factors including the number of water molecules in the coordination shell, the exchange rate of the coordinated water with the bulk water, and the rotational correlation time τR of the contrast agent. The MRI contrast agent can have: 1) high relaxivity for high contrast-to-noise ratio (CNR) and dose efficiency, 2) thermodynamic stability, especially metal selectivity for the target ions over excess physiological metal ions, to minimize the release of toxic paramagnetic metal ions, 3) adequate vascular, tissue retention time to allow imaging, and 4) proper excretion from the body. There is a need in the art to meet some or all of these properties.
  • SUMMARY
  • Embodiments of the present disclosure provide for contrast agents, methods of making contrast agents, and methods of using contrast agents, and the like. One exemplary contrast agent, among others, includes: a) a scaffold protein, and b) at least one metal ion chelating site, wherein the scaffold protein includes at least one metal ion chelating site that is already present or is integrated into the scaffold protein, wherein the scaffold protein includes a metal ion bound to a metal ion chelating site, wherein the contrast agent is stable in a physiological environment.
  • One exemplary method of imaging a sample, among others, includes: administering at least one of the contrast agent described herein to the sample; introducing the sample to an imaging system; and imaging the sample.
  • One exemplary method for preparing a contrast agent, among others, includes: a) selecting a scaffold protein; b) constructing at least one metal ion chelating site; c) operatively embedding the metal ion binding site into the protein, wherein the metal ion has contrast agent properties, and d) attaching at least one polyethylene glycol (PEG) to the scaffold protein.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • Many aspects of the disclosure can be better understood with reference to the following drawings. The components in the drawings are not necessarily to scale, emphasis instead being placed upon clearly illustrating the principles of the present disclosure. Moreover, in the drawings, like reference numerals designate corresponding parts throughout the several views.
  • FIG. 1.1: Schematic descriptions of different classes of MRI contrast agents and simulation of T1 relaxivity. FIG. 1.1( a): Different constructs of MRI contrast agents. (i) Small chelator DPTA with a fast τR Rf) at ˜100 ps level; (ii) Small contrast agents after being covalently conjugated to macromolecules with a slow τR Rs) still possess a fast τR due to its internal mobility; (iii) Schematic description of the design of reported MRI contrast agents by directly coordinating Gd3+ ions to ligand residues on a rigid protein frame to eliminate the high internal mobility. The rotational correlation time of the Gd3+ binding site is the same as that of the whole protein (τRs). FIG. 1.1( b): Simulated T1 relaxivity at the given rotational correlation time τR (100 ps, below, or 10 ns, above), water dwelling time τm, correlation time of splitting τv (1 and 10 ps, solid and dashed lines, respectively), and mean square zero field splitting energy Δ2 (1018 s−2). The τm valves are 10−10 (◯), 10−9 (□), and 10−8 s (Δ) for 100 ps τR and 10−9 (), 10−8 (▪), and 10−7 s (▴) for 10 ns τR according to the theory developed by Blombergen, Solomon ( refs 6 and 7 in Example 1). The water coordination number, q, is assumed to be 1 and the agent concentration is 0.001 M. See on-line supporting materials for r1 and r2 simulations. FIG. 1.1( c): Modeled structure of designed Gd3+-CA1.CD2 based on the designed NMR structure 1T6W (ref 31 in Example 1). Ligand residues E15, E56, D58, D62 and D64 at the B, E, and D β-strands of the host protein CD2 are shown in red.
  • FIG. 1.2: Comparison of in vitro relaxivity between DTPA and designed contrast agents. FIG. 1.2( a): MR images produced using Spin-echo sequence, TR 6000 ms, TI 960 ms, TE 7.6 ms at 3T. Samples are 1) dH2O, 2) 10 mM Tris-HCl pH 7.4, 3) 0.10 mM Gd-DTPA in H2O, 4) 0.10 mM Gd-DTPA in 10 mM Tris-HCl pH 7.4, 5) 0.10 mM Gd3+ and CD2, 6) 0.077 mM Gd-CA4.CD2, 7) 0.050 mM Gd-CA2.CD2, 8) 0.10 mM Gd-CA9.CD2, 9) 0.020 mM Gd-CA1.CD2, and 10) 0.050 mM Gd-CA1.CD2. FIG. 1.2( b): Proton relaxivity values of Gd-CA1.CD2 (r1, solid black; r2, cross) and Gd-DPTA (r1, shield; r2, open) at indicated field strength were measured. FIG. 1.2( c): In vitro relaxivity of contrast agents Gd-DPTA (DTPA), Gd-CA1.CD2 (CA1) and Gd-CA2.CD2 (CA2) in the absence of Ca2+ (black and grey), presence of 1 mM Ca2+ (left strip and open) and 10 mM Ca2+ (right strip and cross) at 3T. T1 (black, left & right strips) and T2 (grey, open and cross) were determined using a Siemens whole-body MR system.
  • FIG. 1.3: Dynamic properties and hydration water number of designed contrast agents. FIG. 1.3( a): S2 order values of the engineered metal binding protein. The positions of ligand residues are shown in vertical bars. Order factors of CA2-CD2 with discontinuous ligand residues have the same dynamic properties as the scaffold protein. Arrows indicate the position of ligand residues. FIG. 1.3( b): Measurement of coordination water number by monitoring Tb3+ life time. Luminescence decay lifetime was obtained by fitting the acquired data in both H2O and D2O with a mono-exponential decay function. A standard curve correlating the Δkobs with water number was established by using well-characterized chelators, such as EDTA (q=3), DTPA (q=1), NTA (q=5), and Aquo Tb3+ (q=9) solution with R2=0.997.26,27 Water numbers coordinated to Tb3+-protein complexes were then obtained by fitting the acquired Δkobs value to the standard curve.
  • FIG. 1.4: In vivo MR images and biodistribution of designed contrast agents. FIG. 1.4( a): MR images of mouse (26 g) pre (left) and 40 minutes post (right) the injection of 50 μL, of 1.2 mM Gd-CA1.CD2 through the tail vein. The MRI was performed using a spin echo sequence with TE/TR/Angle=15 ms/500 ms/90° using a 3T scanner. The arrows indicate the contrast enhancements at different organ sites. FIG. 1.4( b): The MRI signal intensity changes at kidney (), liver (▴), and muscle (⋄) as a function of time. The 0 refers to the pre-injection. FIG. 1.4( c): Tissue distributions 1 hour post intravenous injection of Gd-CA1.CD2 (3.0 μmole/kg, solid black), Gd-DTPA (150 μmole/kg, grey), and GdCl3 (100 μmole/kg, open) The Gd3+ in tissues was measured by ICP-MS and was calculated and expressed as percent of the injected dose.45 The Gd3+ in carcass is the average of randomly picked 10 different sites from rest of whole body after removing indicated organs. Error bars in FIGS. 1.4( a), (b), and (c) are standard deviations of four measurements or four animals (n=4).
  • FIG. 1.5: The ESI-TOF MS spectrum of Gd-CA1.CD2. The CA1.CD2 (10 μM) in 1 mM ammonium acetate (pH 7.0) was mixed with 20 μM of gadolinium chloride (GdCl3). The ESI-TOF MS spectra of the complex were recorded by Q-TOF Micro Mass Spectrometer (Micromass).
  • FIG. 1.6: Measurement of metal binding constants. Gd3+ binding affinity (FIG. 1.6 a) and Zn2+ binding affinity (FIG. 1.6 b) of CA1.CD2 measured by dye competition assays. CA1.CD2 stock solution was gradually added into the 1:1 dye-metal complex to compete for the dye-bound metal ions. The insets show the titration curve for the dye indicators fluo 5N (FIG. 1.6 a) and FluoZin (FIG. 1.6 b), respectively. (FIG. 1.6 c) La3+ binding affinity obtained by Aromatic residue sensitized Tb3+ luminescence energy transfer. The Tb3+ fluorescence of a protein-Tb3+ mixture decreases with the addition of La3+. The La3+ concentrations are 0, 0.30, 0.76, 5.95, 11.03, and 28.95 μM from top to bottom. The Tb3+ fluorescence decrease competition was fitted (line) by a normal competition plus a nonspecific quenching effect (inset).
  • FIG. 1.7: MRI imaging of CA1.CD2 at 9.4 T. FIG. 1.7( a): MR images of CD-1 mouse (26 g) (four mice were imaged) at 9.4T field using Gd-CA1.CD2 as a T2-weighted contrast agent. The images were recorded pre-(i) and 2 hours post-(ii & iii) injection of 50 μl of 1.2 mM Gd-CA1.CD2 agent through the tail vein. MR images were recorded using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence. The in-plane resolution is 0.2×0.3×1.0 mm. FIG. 1.7( b): The T2 MRI relative intensity changes at kidney cortex (cross bars), kidney medulla (bars), kidney center (bars), liver (bars), and muscle (bars) as a function of time (indicated). The MRI intensity in muscle at 10 minutes post contrast administration was defined as 1. MRI intensities at other tissue sites were normalized to the muscle intensity.
  • FIG. 1.8: Detection of CA1.CD2 in serum and cytotoxicity of CA1.CD2. FIG. 1.8( a): Sandwich ELISA detection of CA1.CD2 in mouse blood using OX45 and PabCD2. PabCD2 was used as the capture antibody and OX45 was used as the detection antibody in the sandwich-ELISA experiments. Serum samples were obtained from test mice at 0, 0.5, 1.0, 2, and 3 hours post injection (tail vein) of Gd-CA1.CD2 (˜1.0 μmole/kg). The amounts of CA1.CD2 are expressed as percentages of the injected dose. Calculations are based on the assumption that the total blood volume is 8% of each individual mouse body weight. FIG. 1.8( b): The cytotoxicity of designed protein Gd-CA1.CD2. The SW480 cells were grown under standard conditions (1×104 cells in 100 μl medium). The cells were treated by addition of wild type CD2 (stripe bars), CA1.CD2 (grey bars), Gd-CA1.CD2 (black bars), and Gd-DTPA (cross bars) with concentrations of 30 μM (left panel) or 50 μM (right panel). The open bars are controls where cells were treated with PBS buffer. The cells were incubated with the treatments for 48 hours. The cells were then subjected for MTT assay. The results were presented as percentages of viable cells using the cells that were treated with buffer alone (filled bars) as a reference (100%). The cell lines SW620 and HEK293 were similarly examined. The error bars in FIGS. 1.8( a) and (b) are standard deviations of four measurements or four animals (n=4).
  • FIG. 2.1( a): Illustrates the model structure of the designed contrast agent CA1.CD2 with eight Lys residues highlighted. The solvent accessibility calculated by Getarea is also listed. FIG. 2.1 (b): Examples of PEGylation reagents used with different chain lengths and molecular weights and related chemical reactions.
  • FIG. 2.2: (top) illustrates a FPLC profile for the separation of pegylated protein CA1.CD2 with P12K and reaction mixture by gel filtration column. (bottom) The SDS gel of FPLC fractions (peaks 123) of CA1.CD2 pegylated with P40 stained by idiol (left) and commassie blue (right).
  • FIG. 2.3: The SDS gel stained by commassie blue (middle) for protein and iodine (top) for PEG moiety with 5:1 PEG:protein using the preactivated PEG reagents. 1. Marker; 2. CA1.CD2; 3. PEG4; 4. PEG12; 5. PEG40; 6. PEG5K; 7. PEG12K; 8. PEG20K. CA1.CD2 was Pegylated mainly with 3, 4, 5 PEG. (bottom) MALD-Mass analyses of mixture after PEGylation with PEG40.
  • FIG. 2.4: Illustrates the conformational analysis of PEGylaed CA1.CD2. (left) Trp emission fluorescence spectrum of CA1.CD2 is similar to that PEGylated CA1.CD2-PEG12 and CA1.CD2-PEG40 excited at 280 nm. (right) The Terbium-sensitized energy transfer was used to monitor the binding of metal ions in the designed binding pocket. The Tb3+ emission is gradually increased upon addition of terbium excited at 280 nm.
  • FIG. 2.5: Pegylated CA1.CD2-P40 remains intact after incubate with human serum for 24 hours at 37° C. monitored by SDS Page.
  • FIG. 2.6: R1 (left) and R2 (right) relaxivity values of CA1.CD2 alone, pegylated with P12, and P40 compared with DTPA at different field strengths (0.47, 3.0, 9.4, and 11.4T).
  • FIG. 2.7: Illustrates ELISA (left) or western blot (right) analyses of antibody produced in rabbit serum after i.p. injection of 3 ng/kg of protein CA1.CD2 or PEGylated CA1.CD2 (PEGCA1.CD2). In (left), Pre is the serum from pre-bleeding before antigen injection. CA1.CD2 was mixed with adjuvant (CA1.CD2+Ad) or with buffer saline (CA1.CD2+Sal) before injection. PabCD2 is the anti-serum from rabbits produced by a commercial source use CD2 as antigen. The open bars are the first bleed after first injection. The gray bars are the second bleed after second injection. The rabbit blood was taken 3 weeks after each injection. The error bars are standard deviations of four measurements. In (right), Western blots were performed with anti-serum (1st bleed) from rabbits that were injected; CA1.CD2 mixed with buffered saline (left panel, CA1.CD2+Sal), CA1.CD2 mixed with adjuvant (middle panel, CA1.CD2+Ad), and the PEGylated CA1.CD2 (right panel, PEG-CA1.CD2). The Western blots experiments were carried out with 0.5 mg of PEGylated CA1.CD2 (PEG-CA1.CD2) or unmodified CA1.CD2 (CA1.CD2). Arrow indicates the position of the detected protein bands 19 hours post injection.
  • FIG. 2.8: Table 2.1 is a summary of water number in CA1.CD2 and its variants.
  • FIG. 3.1: Illustrates the development of MRI contrast agents by modifying natural calcium binding proteins such as calmodulin with four metal binding sites.
  • FIG. 3.2: Illustrates the determination of Gd3+ stability constant of CaM variants. CaM titration (FIG. 3.2A) and its curve fitting (FIG. 3.2B) with Fura-2 fluorescence spectra. The measurement was performed at 20 mM Gd3+ and 20 mM Fura-2 with 10 mM Tris and pH 7.4. The arrows show fluorescence intensity changes at 340 nm and 380 nm excitation wavelengths with the increase of CaM concentration, respectively. lem=510 nm. FIG. 3.2( c): The meal selectivity of CBPP (left) and CBPP56 (right). The addition of 1.5 μM protein to free Tb3+ (40 μM) solution resulted in an increase of fluorescence intensity at 545 nm by over 20-fold due to the binding of Tb3+ to the protein and the resultant FRET. 10 mM Mg2+, 2 mM Ca2+, 1 μM Ca2+, 0.1 mM Zn2+, and 10 μM La3+ and 10 μM Gd3+ were subsequently added to individually prepared solutions containing 40 μM Tb3+, 100 mM Kcl and 1.5 μM CBPP or CBPP56.
  • FIG. 3.3: Illustrates the SDS gel of PEGylation of CaM variants with P12 (lanes 1-6), P40 (lanes 7-10), P5K (Lanes 11-14), and P40 (lanes 15-17) at different reaction time with PEG:protein 5:1 ratio stained by Idiol (top A) and commossie blue (bottom B).
  • FIG. 3.4: Illustrates the separation of PEGylation of CaM variants-P12 with mono-Q column (left) and UV absorption spectrum of purified protein (right).
  • FIG. 3.5: Illustrates Tyr emission spectra of CAM variant without and with PEGYlation by P12K excited at 280 nm (FIG. 3.5A) Emission spectra of Tb fluorescence of CAM variant (FIG. 3.5B) and its PEGylated one in the presence of 0 (bottom), 5 uM (middle), and 20 uM of protein (top) excited at 280 nm (FIG. 3.5C).
  • FIG. 3.6: Shows the SDS PAGE results of serum stability for new designed protein based MRI Contrast Agents at different time points incubating with serum at 37° C. (FIG. 3.6A). CBP1; (FIG. 3.6B).CBPP.
  • FIG. 3.7: Illustrates MRI images of Mice at 4.7T with tail vein injection of 6 mM CBP1 at 0, 10, and 30 mins post injection (top). Relative MRI intensity at different organs (bottom).
  • FIG. 3.8: Illustrates relaxivity of CBP1 at 0.47 T. Both R1 and R2 values are 5-8 fold higher than DTPA.
  • FIG. 3.9: Illustrates MRI images of Mice at 4.7T with tail vein injection of 6 mM CBP1-P40 at 0, and 13 mins post injection at slice 3 (top) and slice 4 (bottom). (right) These graphs illustrate the relative MRI intensity at different organs.
  • FIG. 3.10: Illustrates Table 3.1, which shows the dissociation constant of C2+, Tb3+ and Gd3+ to CBPP were measured by Fluorescence spectroscopy. To determine the Ca2+ and Gd3+ dissociation constant, the intrinsic tryptophan fluorescence change were used to monitor the binding process between CBP1/CBPP and metal. The free metal concentration were controlled by the metal-EGTA buffer system and calculated by [Metal]free=Kd*[EGTA-metal]/[EGTA]free. The aromatic residue-sensitized Tb3+ fluorescence at 545 nm were applied to monitor the process of Tb3+ binding to CBP1/CBPP and the variants.
  • FIG. 4.1: The affibody that can specifically bind to Her2 biomarker on the cancer cells was fused to the C-terminal of the protein contrast agent CA1.CD2 with a designed Gd3+ binding site (denoted as CA1-Affi). This contrast agent was surface modified with PEG40 to reduce immunogenesity and increase solubility and serum stability (PEG-CA1-Affi). The affibody was further conjugated with near infra-red dye Cy5.5 via a Cys at the C-terminal to generate a dual labeled contrast agent PEG-CA1-Affi-Cy5.5.
  • FIG. 4.2: Contrast agent with PeGylation (PEG-CA1-affi) and without PeGylation (CA1-Aff) is able to bind to the positive cell line AU565 with membrane staining at 4 C (top right) and both membrane and cytosol staining at 37 C (top left). This developed contrast agent does not bind to negative cell line EMT-6 at either 4 or 37 C. (Bottom right) Near IR labeled contrast agent PEG-CA1-affi-Cy5.5 is able to bind to positive cell lines AU565 and AKOV-3 with NIR fluorescence signal. These data suggest that our contrast agent fused with the affibody can target to HER2 positive tumor cell lines specifically. PEGylation does not change the target capability to the cancer cell. At 37° C., the contrast agent is endocytosized.
  • FIG. 4.3: Specific targeting to the positive cancer cell line (AU565) and negative cell line (EMT6) monitored by 153Gd (FIG. 4.3A) and ELISA (FIG. 4.3B). Similar radioactivity (CPM) of the contrast agents without PEGylation (CA1-Affi) and with PEGylation (PEG-CA1-Affi) at 75, 150, 375 nM were observed for AU565 cell line. Under identical conditions, negative cell line EMT has a small radioactivity after incubating with 153Gd labeled contrast agent.
  • FIG. 4.4: Breast cancer biomarker HER2 positive tumor and negative tumor were implanted on the left and right back in nude mice. 5 mM contrast agent CA1.Affi-P40 (100 fold lower than clinic used DTPA) was injected via tail vein. MRI images at 4.7 T using fast spin echo were acquired before injection, and at 5 min, 30 min, 3 hr, 24 hr and 52 hr post injection. Positive tumor shows a strong contrast after 30 mins and peaked at 24 hour with about 35% enhancement. Contrast capability was decreased after 52 hours, suggesting that the contrast agent was secreted out of the animal. This mouse was alive and looks normal after 52 hours MRI scanning.
  • FIG. 4.5: Nude mice were inoculated with negative cell line MDA-MB-231 and positive cell line SKOV-3 (top). The cell number for each spot was about 5×106. The specific binding of positive tumor on the right upon injection of the dual labeled contrast PeG-CA1-Affi-Cy5.5 can be visualized using Kodak NIR in vivo FX-pro animal imaging system 21 hours poster injection. (middle) Traverse MR images of tumor mice at 4.7T with fast spin echo obtained at 3 min., 35 min., and 21 hours following administration of the contrast agent. (bottom right) The intensity enhancement at the positive tumor by our contrast agent analyzed by Image J.
  • FIG. 4.6: Western Blot with PAbPEGCA1 (top) and NIR imaging (bottom) of different tissues of the tumor nice after MRI imaging.
  • FIG. 4.7: Immunohistochemistry (IHC) staining using the antibody PAbPGCA1 with tissue slides made from the tissue samples from the imaged mice, including HER2 tumors. Strongest staining was observed with liver and HER2 positive tumor tissue slides. The kidney slides also gave strong immunostaining. Interestingly, the areas near proximal tubes showed the strongest staining in the slides made from the kidney, indicating that the protein contrast agent was ready to be filtered through the kidney.
  • FIG. 4.8: The binding of CAM-Affi with HER2 positive cells was measured by western blot.
  • FIG. 4.9: Immune staining of CaM-Affi treated breast cancer cells for skov-3 (Left, Her 2 positive) and MDA-MB-231 (right, Her2 negative).
  • FIG. 4.10: Immunostaining CA-Bom, CA-52I-Bom, CA at different time points for PC-3 and DU145 (GRPR high expression), and H441 (GRPR low expression).
  • FIG. 4.11: Near IR imaging of nude mice xenografted with DU-145 tumor (high expression of GRPR, left) and H441 tumor (low expression of GRPR, control, right) post injection of CA1.CD2-52Ibom-cy5.5-P40 26 hours via tail vein.
  • FIG. 4.12: NIR imaging (top) and NIR intensity (bottom) of CA1.CD2-52I-Bom-Cy5.5-P40 at different organs of the mice.
  • DETAILED DESCRIPTION
  • Before the present disclosure is described in greater detail, it is to be understood that this disclosure is not limited to particular embodiments described, and the embodiment of the invention as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present disclosure will be limited only by the appended claims.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.
  • All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference and are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
  • As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present disclosure. Any recited method can be carried out in the order of events recited or in any other order that is logically possible.
  • Embodiments of the present disclosure will employ, unless otherwise indicated, techniques of imaging, synthetic organic chemistry, biochemistry, biology, molecular biology, recombinant DNA techniques, pharmacology, and the like, which are within the skill of the art. Such techniques are explained fully in the literature.
  • The examples herein are put forth so as to provide those of ordinary skill in the art with an illustrative disclosure and description of how to perform the methods and use the compounds disclosed and claimed herein. Unless indicated otherwise, parts are parts by weight, temperature is in ° C., and pressure is at or near atmospheric. Standard temperature and pressure are defined as 20° C. and 1 atmosphere.
  • Before the embodiments of the present disclosure are described in detail, it is to be understood that, unless otherwise indicated, the present disclosure is not limited to particular materials, reagents, reaction materials, manufacturing processes, or the like, as such can vary. It is also to be understood that the terminology used herein is for purposes of describing particular embodiments only, and is not intended to be limiting. It is also possible in the present disclosure that steps can be executed in different sequence where this is logically possible.
  • It must be noted that, as used in the specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a compound” includes a plurality of compounds. In this specification and in the claims that follow, reference will be made to a number of terms that shall be defined to have the following meanings unless a contrary intention is apparent.
  • DEFINITIONS
  • In describing and claiming the disclosed subject matter, the following terminology will be used in accordance with the definitions set forth below.
  • As used herein, a “contrast agent” is intended to include any agent that is physiologically tolerable and capable of providing enhanced contrast for magnetic resonance imaging. A suitable contrast agent is preferably biocompatible, e.g., non-toxic, chemically stable, not absorbed by the body or reactive with a tissue, and eliminated from the body within a short time.
  • The term “polymer” means any compound that is made up of two or more monomeric units covalently bonded to each other, where the monomeric units may be the same or different, such that the polymer may be a homopolymer or a heteropolymer. Representative polymers include peptides, polysaccharides, nucleic acids and the like, where the polymers may be naturally occurring or synthetic.
  • The term “polypeptides” includes proteins and fragments thereof. Polypeptides are disclosed herein as amino acid residue sequences. Those sequences are written left to right in the direction from the amino to the carboxy terminus. In accordance with standard nomenclature, amino acid residue sequences are denominated by either a three letter or a single letter code as indicated as follows: Alanine (Ala, A), Arginine (Arg, R), Asparagine (Asn, N), Aspartic Acid (Asp, D), Cysteine (Cys, C), Glutamine (Gln, Q), Glutamic Acid (Glu, E), Glycine (Gly, G), Histidine (His, H), Isoleucine (Ile, I), Leucine (Leu, L), Lysine (Lys, K), Methionine (Met, M), Phenylalanine (Phe, F), Proline (Pro, P), Serine (Ser, S), Threonine (Thr, T), Tryptophan (Trp, W), Tyrosine (Tyr, Y), and Valine (Val, V). In addition, the protein can include non-standard and/or non-naturally occurring amino acids, as well as other amino acids that may be found in phosphorylated proteins in organisms such as, but not limited to, animals, plants, insects, protists, fungi, bacteria, algae, single-cell organisms, and the like. The non-standard amino acids include, but are not limited to, selenocysteine, pyrrolysine, gamma-aminobutyric acid, carnitine, ornithine, citrulline, homocysteine, hydroxyproline, hydroxylysine, sarcosine, and the like. The non-naturally occurring amino acids include, but are not limited to, trans-3-methylproline, 2,4-methanoproline, cis-4-hydroxyproline, trans-4-hydroxyproline, N-methyl-glycine, allo-threonine, methylthreonine, hydroxy-ethylcysteine, hydroxyethylhomocysteine, nitro-glutamine, homoglutamine, pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, 3,3-dimethylproline, tert-leucine, norvaline, 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, and 4-fluorophenylalanine.
  • “Variant” refers to a polypeptide or polynucleotide or polymer that differs from a reference polypeptide or polynucleotide or polymer, but retains essential properties. A typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical. A variant and reference polypeptide may differ in amino acid sequence by one or more modifications (e.g., substitutions, additions, and/or deletions). A variant of a polypeptide includes conservatively modified variants. A substituted or inserted amino acid residue may or may not be one encoded by the genetic code. A variant of a polypeptide may be naturally occurring, such as an allelic variant, or it may be a variant that is not known to occur naturally.
  • A variant of a polypeptide may contain different modifications such as with PEGylation groups or the same type of groups with different sizes or lengths of the modifications.
  • “Variant” generated such as by modifying metal binding sites may have different metal binding properties and relaxivities and vivo peroperties.
  • Modifications and changes can be made in the structure of the polypeptides of this disclosure and still obtain a molecule having similar characteristics as the polypeptide (e.g., a conservative amino acid substitution). For example, certain amino acids can be substituted for other amino acids in a sequence without appreciable loss of activity. Because it is the interactive capacity and nature of a polypeptide that defines that polypeptide's biological functional activity, certain amino acid sequence substitutions can be made in a polypeptide sequence and nevertheless obtain a polypeptide with like properties.
  • In making such changes, the hydropathic index of amino acids can be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a polypeptide is generally understood in the art. It is known that certain amino acids can be substituted for other amino acids having a similar hydropathic index or score and still result in a polypeptide with similar biological activity. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics. Those indices are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cysteine (+2.5); methionine (+1.9); alanine (+1.8); glycine (−0.4); threonine (−0.7); serine (−0.8); tryptophan (−0.9); tyrosine (−1.3); proline (−1.6); histidine (−3.2); glutamate (−3.5); glutamine (−3.5); aspartate (−3.5); asparagine (−3.5); lysine (−3.9); and arginine (−4.5).
  • It is believed that the relative hydropathic character of the amino acid determines the secondary structure of the resultant polypeptide, which in turn defines the interaction of the polypeptide with other molecules, such as enzymes, substrates, receptors, antibodies, antigens, and the like. It is known in the art that an amino acid can be substituted by another amino acid having a similar hydropathic index and still obtain a functionally equivalent polypeptide. In such changes, the substitution of amino acids whose hydropathic indices are within ±2 is preferred, those within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred.
  • Substitution of like amino acids can also be made on the basis of hydrophilicity, particularly, where the biological functional equivalent polypeptide or peptide thereby created is intended for use in immunological embodiments. The following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0±1); glutamate (+3.0±1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); proline (−0.5±1); threonine (−0.4); alanine (−0.5); histidine (−0.5); cysteine (−1.0); methionine (−1.3); valine (−1.5); leucine (−1.8); isoleucine (−1.8); tyrosine (−2.3); phenylalanine (−2.5); tryptophan (−3.4). It is understood that an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent polypeptide. In such changes, the substitution of amino acids whose hydrophilicity values are within ±2 is preferred, those within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred.
  • As outlined above, amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like. Exemplary substitutions that take various of the foregoing characteristics into consideration are well known to those of skill in the art and include (original residue: exemplary substitution): (Ala: Gly, Ser), (Arg: Lys), (Asn: Gln, His), (Asp: Glu, Cys, Ser), (Gln: Asn), (Glu: Asp), (Gly: Ala), (His: Asn, Gln), (Ile: Leu, Val), (Leu: Ile, Val), (Lys: Arg), (Met: Leu, Tyr), (Ser: Thr), (Thr: Ser), (Tip: Tyr), (Tyr: Trp, Phe), and (Val: Ile, Leu). Embodiments of this disclosure thus contemplate functional or biological equivalents of a polypeptide as set forth above. In particular, embodiments of the polypeptides can include variants having about 50%, 60%, 70%, 80%, 90%, and 95% sequence identity to the polypeptide of interest.
  • “Identity,” as known in the art, is a relationship between two or more polypeptide sequences, as determined by comparing the sequences. In the art, “identity” also means the degree of sequence relatedness between polypeptides as determined by the match between strings of such sequences. “Identity” and “similarity” can be readily calculated by known methods, including, but not limited to, those described in (Computational Molecular Biology, Lesk, A. M., Ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., Ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., Eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., Eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM J Applied Math., 48: 1073 (1988).
  • Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. The percent identity between two sequences can be determined by using analysis software (e.g., Sequence Analysis Software Package of the Genetics Computer Group, Madison Wis.) that incorporates the Needelman and Wunsch, (J. Mol. Biol., 48: 443-453, 1970) algorithm (e.g., NBLAST and XBLAST). The default parameters are used to determine the identity of the polypeptides of the present disclosure.
  • By way of example, a polypeptide sequence may be identical to the reference sequence, that is 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the % identity is less than 100%. Such alterations are selected from: at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence. The number of amino acid alterations for a given % identity is determined by multiplying the total number of amino acids in the reference polypeptide by the numerical percent of the respective percent identity (divided by 100) and then subtracting that product from said total number of amino acids in the reference polypeptide.
  • Conservative amino acid variants can also comprise non-naturally occurring amino acid residues. Non-naturally occurring amino acids include, without limitation, trans-3-methylproline, 2,4-methanoproline, cis-4-hydroxyproline, trans-4-hydroxyproline, N-methyl-glycine, allo-threonine, methylthreonine, hydroxy-ethylcysteine, hydroxyethylhomocysteine, nitro-glutamine, homoglutamine, pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, 3,3-dimethylproline, tert-leucine, norvaline, 2-azaphenyl-alanine, 3-azaphenylalanine, 4-azaphenylalanine, and 4-fluorophenylalanine. Several methods are known in the art for incorporating non-naturally occurring amino acid residues into proteins. For example, an in vitro system can be employed wherein nonsense mutations are suppressed using chemically aminoacylated suppressor tRNAs. Methods for synthesizing amino acids and aminoacylating tRNA are known in the art. Transcription and translation of plasmids containing nonsense mutations is carried out in a cell-free system comprising an E. coli S30 extract and commercially available enzymes and other reagents. Proteins are purified by chromatography. (Robertson, et al., J. Am. Chem. Soc., 113: 2722, 1991; Ellman, et al., Methods Enzymol., 202: 301, 1991; Chung, et al., Science, 259: 806-9, 1993; and Chung, et al., Proc. Natl. Acad. Sci. USA, 90: 10145-9, 1993). In a second method, translation is carried out in Xenopus oocytes by microinjection of mutated mRNA and chemically aminoacylated suppressor tRNAs (Turcatti, et al., J. Biol. Chem., 271: 19991-8, 1996). Within a third method, E. coli cells are cultured in the absence of a natural amino acid that is to be replaced (e.g., phenylalanine) and in the presence of the desired non-naturally occurring amino acid(s) (e.g., 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, or 4-fluorophenylalanine). The non-naturally occurring amino acid is incorporated into the protein in place of its natural counterpart. (Koide, et al., Biochem., 33: 7470-6, 1994). Naturally occurring amino acid residues can be converted to non-naturally occurring species by in vitro chemical modification. Chemical modification can be combined with site-directed mutagenesis to further expand the range of substitutions (Wynn, et al., Protein Sci., 2: 395-403, 1993).
  • As used herein, the term “polynucleotide” generally refers to any polyribonucleotide or polydeoxyribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. Thus, for instance, polynucleotides as used herein refers to, among others, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. Polynucleotide encompasses the terms “nucleic acid,” “nucleic acid sequence,” or “oligonucleotide” as defined above.
  • In addition, polynucleotide as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonucleotide.
  • As used herein, the term polynucleotide includes DNAs or RNAs as described above that contain one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein.
  • It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art. The term polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells, inter alias.
  • By way of example, a polynucleotide sequence of the present disclosure may be identical to the reference sequence, that is be 100% identical, or it may include up to a certain integer number of nucleotide alterations as compared to the reference sequence. Such alterations are selected from the group including at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5′ or 3′ terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence. The number of nucleotide alterations is determined by multiplying the total number of nucleotides in the reference nucleotide by the numerical percent of the respective percent identity (divided by 100) and subtracting that product from said total number of nucleotides in the reference nucleotide. Alterations of a polynucleotide sequence encoding the polypeptide may alter the polypeptide encoded by the polynucleotide following such alterations.
  • The term “codon” means a specific triplet of mononucleotides in the DNA chain. Codons correspond to specific amino acids (as defined by the transfer RNAs) or to start and stop of translation by the ribosome.
  • The term “degenerate nucleotide sequence” denotes a sequence of nucleotides that includes one or more degenerate codons (as compared to a reference polynucleotide molecule that encodes a polypeptide). Degenerate codons contain different triplets of nucleotides, but encode the same amino acid residue (e.g., GAU and GAC triplets each encode Asp).
  • The term “antibody” is used to refer both to a homogenous molecular entity, or a mixture such as a serum product made up of a plurality of different molecular entities. Monoclonal or polyclonal antibodies, which specifically react with the virosomes of the present disclosure, may be made by methods known in the art. (e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratories; Goding (1986) Monoclonal Antibodies: Principles and Practice, 2d ed., Academic Press, New York; and Ausubel et al. (1987)). Also, recombinant immunoglobulin may be produced by methods known in the art, including but not limited to, the methods described in U.S. Pat. No. 4,816,567, which is hereby incorporated by reference herein.
  • Affibody® ligands (U.S. Pat. No. 5,831,012, which is incorporated herein by reference) are highly specific affinity proteins that may be designed and used like aptamers. Affibodies may be produced or purchased from commercial sources (Affibody AB, Bromma, Sweden). Aptamers and affibodies may be used in combination with antibodies to increase the functional avidity of translucent or non-translucent solid matrices for probe molecule binding. Increased binding in turn results in an increased signal strength, greater signal-to-noise ratio, more reproducible target molecule detection and greater sensitivity of detection.
  • Aptamers must also be differentiated from the naturally occurring nucleic acid sequences that bind to certain proteins. These latter sequences generally are naturally occurring sequences embedded within the genome of the organism that bind to a specialized sub-group of proteins or polypeptides, or their derivatives, that are involved in the transcription, translation, and transportation of naturally occurring nucleic acids, i.e., protein-binding nucleic acids. Aptamers on the other hand are short, isolated, non-naturally occurring nucleic acid molecules. While aptamers can be identified that bind nucleic acid-binding proteins, in most cases such aptamers have little or no sequence identity to the sequences recognized by the nucleic acid-binding proteins in nature. More importantly, aptamers can be selected to bind virtually any protein (not just nucleic acid-binding proteins) as well as almost any target of interest including small molecules, carbohydrates, peptides, etc. For most targets, even proteins, a naturally occurring nucleic acid sequence to which it binds does not exist. For those targets that do have such a sequence, i.e., nucleic acid-binding proteins, such sequences will differ from aptamers as a result of the relatively low binding affinity used in nature as compared to tightly binding aptamers. Aptamers are capable of specifically binding to selected targets and modulating the target's activity or binding interactions, e.g., through binding, aptamers may block their target's ability to function. The functional property of specific binding to a target is an inherent property of an aptamer.
  • A typical aptamer is 6-35 kDa in size (20-100 nucleotides), binds its target with micromolar to sub-nanomolar affinity, and may discriminate against closely related targets (e.g., aptamers may selectively bind related proteins from the same gene family). Aptamers are capable of using intermolecular interactions such as hydrogen bonding, electrostatic complementarities, hydrophobic contacts, and steric exclusion to bind with a specific target. In the present disclosure, aptamers also employ boronic acid-Lewis base/nucleophile (such as hydroxyl groups, diols, and amino groups) interactions for binding. Aptamers have a number of desirable characteristics for use as therapeutics and diagnostics including high specificity and affinity, low immunogenicity, biological efficacy, and excellent pharmacokinetic properties.
  • As used herein, the term “PEGylation” means and refers to modifying a polymer (e.g., a protein) by covalently attaching polyethylene glycol (PEG) to the polymer, with “PEGylated” referring to a polymer having a PEG attached. For further general information on PEGylation methods see, for example, the Nektar Advanced PEGylation Catalogs 2004 and 2005-2006, as well as the references cited therein. PEGYlation can be achieved by non-specific interaction with functional group of polypeptide chain such as via amino group or specific interaction at certain location of the macromolecules such as amino terminal or at the Cys residues.
  • The terms “biomarker” or “biomarker probe” are used to refer to a substance used as an indicator of a biologic state. It is a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, and/or pharmacologic responses to a therapeutic intervention.
  • The term “disease marker” is used to refer to substances, such as proteins, bio-chemicals, nucleic acids, carbohydrates, or enzymes, produced by disease cells or by the body in response to disease cells during disease development and progression. These substances are indicative of a particular disease process.
  • By “administration” is meant introducing a compound into a subject. The preferred route of administration of the compounds is intravenous. However, any route of administration, such as oral, topical, subcutaneous, peritoneal, intraarterial, inhalation, vaginal, rectal, nasal, introduction into the cerebrospinal fluid, or instillation into body compartments can be used.
  • As used herein, the terms “treatment”, “treating”, and “treat” are defined as acting upon a disease, disorder, or condition with an agent to reduce or ameliorate the pharmacologic and/or physiologic effects of the disease, disorder, or condition and/or its symptoms. “Treatment,” as used herein, covers any treatment of a disease in a host (e.g., a mammal, typically a human or non-human animal of veterinary interest), and includes: (a) reducing the risk of occurrence of the disease in a subject determined to be predisposed to the disease but not yet diagnosed as infected with the disease (b) impeding the development of the disease, and (c) relieving the disease, i.e., causing regression of the disease and/or relieving one or more disease symptoms. “Treatment” is also meant to encompass delivery of a contrast agent including a compound to provide a pharmacologic effect, even in the absence of a disease or condition. For example, “treatment” encompasses delivery of a disease or pathogen compound via the contrast agent that provides for enhanced or desirable effects in the subject (e.g., reduction of pathogen load, reduction of disease symptoms, etc.).
  • As used herein, the terms “prophylactically treat” or “prophylactically treating” refers to completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • The term “unit dosage form,” as used herein, refers to physically discrete units suitable as unitary dosages for human and/or animal subjects, each unit containing a predetermined quantity of a contrast agent calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for unit dosage forms depend on the particular compound employed, the route and frequency of administration, the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • A “pharmaceutically acceptable excipient,” “pharmaceutically acceptable diluent,” “pharmaceutically acceptable carrier,” or “pharmaceutically acceptable adjuvant” means an excipient, diluent, carrier, and/or adjuvant that are useful in preparing a pharmaceutical composition that are generally safe, non-toxic and neither biologically nor otherwise undesirable, and include an excipient, diluent, carrier, and adjuvant that are acceptable for veterinary use and/or human pharmaceutical use. “A pharmaceutically acceptable excipient, diluent, carrier and/or adjuvant” as used in the specification and claims includes one or more such excipients, diluents, carriers, and adjuvants.
  • As used herein, a “pharmaceutical composition” is meant to encompass a contrast agent suitable for administration to a subject, such as a mammal, especially a human. In general a “pharmaceutical composition” is sterile, and preferably free of contaminants that are capable of eliciting an undesirable response within the subject (e.g., the compound(s) in the pharmaceutical composition is pharmaceutical grade). Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, intravenous, buccal, rectal, parenteral, intraperitoneal, intradermal, intracheal, intramuscular, subcutaneous, inhalational and the like.
  • The terms “therapeutically effective amount” and “an effective amount” are used interchangeably herein and refer to that amount of a contrast agent being administered that is sufficient to effect the intended application. In an embodiment, the effective amount of the contrast agent includes enough so that the disease, for example, in the host can be imaged, studied, diagnosed, or the like. For example, an effective amount of a contrast agent including a compound will relieve to some extent one or more of the symptoms of the disease being treated, and/or that amount that will prevent, to some extent, one or more of the symptoms of the disease that the host being treated has or is at risk of developing. The therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • As used herein, the term “host,” “subject,” “patient,” or “organism” includes humans and mammals (e.g., mice, rats, pigs, cats, dogs, and horses). Typical hosts to which compounds of the present disclosure may be administered will be mammals, particularly primates, especially humans. For veterinary applications, a wide variety of subjects will be suitable, e.g., livestock such as cattle, sheep, goats, cows, swine, and the like; poultry such as chickens, ducks, geese, turkeys, and the like; and domesticated animals particularly pets such as dogs and cats. For diagnostic or research applications, a wide variety of mammals will be suitable subjects, including rodents (e.g., mice, rats, hamsters), rabbits, primates, and swine such as inbred pigs and the like. The term “living host” refers to a host noted above or another organism that is alive. The term “living host” refers to the entire host or organism and not just a part excised (e.g., a liver or other organ) from the living host.
  • “Cancer”, as used herein, shall be given its ordinary meaning, as a general term for diseases in which abnormal cells divide without control. In particular, cancer refers to angiogenesis related cancer. Cancer cells can invade nearby tissues and can spread through the bloodstream and lymphatic system to other parts of the body.
  • There are several main types of cancer, for example, carcinoma is cancer that begins in the skin or in tissues that line or cover internal organs. Sarcoma is cancer that begins in bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue. Leukemia is cancer that starts in blood-forming tissue such as the bone marrow, and causes large numbers of abnormal blood cells to be produced and enter the bloodstream. Lymphoma is cancer that begins in the cells of the immune system.
  • When normal cells lose their ability to behave as a specified, controlled and coordinated unit, a tumor is formed. Generally, a solid tumor is an abnormal mass of tissue that usually does not contain cysts or liquid areas (some brain tumors do have cysts and central necrotic areas filled with liquid). A single tumor may even have different populations of cells within it, with differing processes that have gone awry. Solid tumors may be benign (not cancerous), or malignant (cancerous). Different types of solid tumors are named for the type of cells that form them. Examples of solid tumors are sarcomas, carcinomas, and lymphomas. Leukemias (cancers of the blood) generally do not form solid tumors.
  • Representative cancers include, but are not limited to, bladder cancer, breast cancer, colorectal cancer, endometrial cancer, head and neck cancer, lung cancer, lymphoma, melanoma, non-small-cell lung cancer, ovarian cancer, prostate cancer, testicular cancer, uterine cancer, cervical cancer, thyroid cancer, gastric cancer, brain stem glioma, cerebellar astrocytoma, cerebral astrocytoma, glioblastoma, ependymoma, Ewing's sarcoma family of tumors, germ cell tumor, extracranial cancer, Hodgkin's disease, leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, liver cancer, medulloblastoma, neuroblastoma, brain tumors generally, non-Hodgkin's lymphoma, osteosarcoma, malignant fibrous histiocytoma of bone, retinoblastoma, rhabdomyosarcoma, soft tissue sarcomas generally, supratentorial primitive neuroectodermal and pineal tumors, visual pathway and hypothalamic glioma, Wilms' tumor, acute lymphocytic leukemia, adult acute myeloid leukemia, adult non-Hodgkin's lymphoma, chronic lymphocytic leukemia, chronic myeloid leukemia, esophageal cancer, hairy cell leukemia, kidney cancer, multiple myeloma, oral cancer, pancreatic cancer, primary central nervous system lymphoma, skin cancer, small-cell lung cancer, among others.
  • A tumor can be classified as malignant or benign. In both cases, there is an abnormal aggregation and proliferation of cells. In the case of a malignant tumor, these cells behave more aggressively, acquiring properties of increased invasiveness. Ultimately, the tumor cells may even gain the ability to break away from the microscopic environment in which they originated, spread to another area of the body (with a very different environment, not normally conducive to their growth), and continue their rapid growth and division in this new location. This is called metastasis. Once malignant cells have metastasized, achieving a cure is more difficult.
  • Benign tumors have less of a tendency to invade and are less likely to metastasize. Brain tumors spread extensively within the brain but do not usually metastasize outside the brain. Gliomas are very invasive inside the brain, even crossing hemispheres. They do divide in an uncontrolled manner, though. Depending on their location, they can be just as life threatening as malignant lesions. An example of this would be a benign tumor in the brain, which can grow and occupy space within the skull, leading to increased pressure on the brain.
  • It should be noted that precancerous cells, cancer cells, cancer, and tumors may be used interchangeably in the disclosure.
  • The terms “including”, “such as”, “for example”, and the like are intended to refer to exemplary embodiments and not to limit the scope of the present disclosure.
  • Discussion
  • In accordance with the purpose(s) of the present disclosure, as embodied and broadly described herein, embodiments of the present disclosure, in one aspect, relate to contrast agents, compositions including contrast agents, methods of making contrast agents, methods of imaging, methods of diagnosing, methods of studying, and the like. More particularly, embodiments of the contrast agents include magnetic resonance imaging contrast agents that accumulate in tissue and can be used to determine the presence and/or location of a target. In addition, contrast agents of the present disclosure can include targeting agents to target cells or tissue (e.g., cancer). Embodiments of the present disclosure can be tuned to have properties for diagnostic imaging.
  • In general, embodiments of the present disclosure include contrasts agents that include a scaffold polymer (e.g., protein or peptide) that includes (e.g., integrated into the scaffold protein) at least one tailored metal ion binding site (e.g., 1, 2, 3, 4, 5, or more biding sites) (also referred to as “metal ion binding site”) capable of chelating paramagnetic and heavy metal ions. In an embodiment, the contrast agent can include a targeting agent. In an embodiment, the contrast agent can include a Near-IR moiety (e.g., functional group). In an embodiment, the modification of the metal binding site, either by residue mutation or insertion, is intended to alter the metal selectivity of the binding site. In an embodiment, the contrast agent includes a metal ion interacting (e.g., bonding with or chelating with) with the metal ion binding site. In an embodiment, a contrast agent can include two metal ions interacting (e.g., bonding with or chelating with) with two metal ion binding sites. In an embodiment, the metal ion binding site may be developed by a design approach or by a grafting approach. After the site has been developed, the site or sites are operatively integrated into the select areas of the scaffold polymer.
  • In an embodiment, the contrast agent is stable in a physiological environment. The phrase “physiological environment” can be described as cell, cellular conditions, tissues, organs, and vertebrate/invertebrates, animal/human or buffer conditions (e.g., pH of about 6-8 and a temperature of about 5-45° C.) mimic closely to the cellular, or in vivo conditions. The term “stable” in reference to “physiological environment” means that the contrast agent is able to provide contrast capability and remain intact. In an embodiment, the phrase “stable in a physiological environment” refers to the contrast agent including at least one metal ion and the binding of the metal ion causes no changes or substantially no changes (e.g., less than 50%) to the protein (scaffold protein) conformation or to the binding affinity of the tailored metal ion binding site under clinical conditions (physiological environment) that would cause premature release of the metal ion, and that the contrast agent functions as a contrast agent as described herein.
  • In an embodiment, the scaffold polymer of the contrast agent includes polyethylene glycerol compounds (PEG) attached to the polymer. The PEGs can be attached (e.g., bonded) to the polymer via an amino acid residue such as lysine (Lys), glutamic acid (Glu), aspartic acid (Asp), cysteine (Cys), and/or carboxyl/amino terminals. The position of the amino acids on the polymer can be selected to position the PEGs so that the PEGs do not substantially interfere (e.g., decrease metal binding affinity more than 20%) with or interfere with the metal ion binding sites ability to interact with the metal ion of interest or the conformation of the polymer. In an embodiment, the PEGs are attached to one or more Lys residues since the position of the Lys residues on the polymer is such that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer. Unless otherwise indicated or understood from the context of the sentence or the embodiments being described, reference to “contrast agent” refers to a contrast agent that includes PEGs. As noted herein, embodiments of the present disclosure include contrast agents that include PEGs and contrast agents that do not include PEGs. Additional details regarding PEGs are described herein.
  • Embodiments of the present disclosure provide for PEGylated contrast agents, where the PEGylation increases the blood circulation time of the contrast agent in CD-1 mice. In addition, PEGylation of the contrast agent increased the solubility of the contrast agent by more than two-fold, three-fold, four-fold, five-fold, six-fold, seven-fold, eight-fold, nine-fold, ten-fold, or more relative to the un-PEGylated contrast agent. It should also be noted that PEGylation of the contrast agent further increased the in vitro of one or both R1 and R2 relaxivities of the contrast agents by about 10%, 25%, 50%, 75%, 100%, or 2-3 fold or more, relative to the un-PEGylated contrast agent. Although not intending to be bound by theory, the increase in molecular size due to the PEGylation and the addition of a hydration layer due to water retention by Poly-PEG chain on protein surface may be the reasons for the increases in the relaxivities.
  • Embodiments of this disclosure include contrast agents capable of enhancing image contrast by affecting water molecule proton relaxation rates. Such contrast agents are effective for magnetic resonance imaging, in part, because the water proton relaxation rate in the target tissue is affected differently from the relaxation rate of the water protons in the surrounding tissue. In an embodiment of the present disclosure, the contrast agents are paramagnetic species, which form complexes with metal ions, so to alter the relaxation rates of adjacent nuclei.
  • In an embodiment, the scaffold polymer (referred to as a “protein” or “peptide” hereinafter) for MRI applications are a protein that will host the tailored metal ion binding sites and has the following characteristics:
  • (a) stability in a physiological environment,
  • (b) a topology suitable for the integration of metal ion sites,
  • (c) a rotational correlation time optimized for the magnetic field (e.g., around 100 milliseconds in a magnetic field of 1.3 to 3T), e.g., higher magnetic field application can demand a host protein with a larger molecular weight, and
  • (d) a water exchange rate such that the relaxivity of the protein is not limited by the water exchange rate.
  • In another embodiment, the contrast agent for use in MRI applications can include a scaffold protein (referred to as a “protein”, “polymer”, or “peptide”) that includes a natural metal binding protein or a fragment/domain of natural metal binding proteins either with metal binding sites modified by at least one amino acid or protein modification.
  • Properties of the scaffold protein also may include water solubility, low interaction with the other cellular metal ions and low toxicity. While all these properties are not required, the optimal properties of the scaffold protein can depend on the specific parameters of the imaging application.
  • Another property of the scaffold protein is its ability to accept the introduction of metal ion binding sites therein. In an embodiment, the scaffold protein has a three-dimensional structure or an amino sequence with some homology to the proteins whose structures have been solved, at least in part. Specifically, the scaffold protein is screened to determine whether it can tolerate the integration of various binding sites without excessive denaturation. For example, the integration of metal ion binding sites into the scaffold protein should not denature or unfold the protein. Thus, the metal ion binding site should not be placed by mutating a hydrophobic core or in a position that results in substantial structural perturbation. This can be examined by sequence alignment of proteins in the same family. In an embodiment, the amino acids that have an essential role in folding of the structure or the function will be conserved among different species of this same type of the protein.
  • In an embodiment, metal ion binding sites are placed into a scaffold protein such that the metal can be tumbled together with the protein. It is better to find a location that is not so flexible or the same flexibility as the protein body so as to match the correction time. In an embodiment, it is preferred to design or create the binding pocket in the protein. Although insertion could work, it is preferable to do so in a relatively not so flexible region. Usually the protein can be checked by looking at the B factor (temperature factor for X-ray) or S2 factor (dynamic flexibility factor for NMR) of the pdb (protein data bank) file of the structure.
  • In an embodiment, more than one metal binding site may be integrated into a scaffold protein. The inclusion of more than one binding site improves the sensitivity of the contrast agent. In embodiments where more than one binding site is integrated into the protein, the site could have different affinities, but should still have strong enough affinity for the selected metal so to avoid competition with physiological metal ions. Both metal ions should be embedded into the host protein with preferred rotational correlation times and water exchange rates to provide MRI contrast with an increased sensitivity.
  • In an embodiment, the contrast agent can have a high affinity to and can preferentially select a particular metal ion (e.g., Gd3+, Mn2+, or Fe3+). In one example, exemplary contrast agents showed a dissociation constant Kd less than 1012 [M] for Gd3+ in an environment having physiological metal ions and prevented those metal ions from precipitation under physiological conditions. Thus, the present disclosure may be used to create contrast agents having optimal selectivity for a specific metal ion.
  • Embodiments of the present disclosure can provide a new mechanism to increase the relaxivity of contrast agents. This is accomplished by designing the metal ion binding sites, e.g., Gd3+, in proteins, which can eliminate the mobility and flexibility of the chelating moiety associated with currently available contrast agents. High proton relaxivity by contrast agents can further enhance images.
  • An advantage of the present disclosure is that it provides contrast agents that can preferentially chelate a specific metal ion. For example, a preferred contrast agent having Gd3+ binding site(s) will preferentially chelate Gd3+ over other metal ions, such as Mg2+ or Ca2+. The ability to preferentially chelate a specific metal ion can improve the specificity of a contrast agent and reduces the cytotoxicity of the contrast agent.
  • As mentioned above, some embodiments of the contrast agents include PEGs attached to the protein. Inclusion of the PEGs in the contrast agent increases blood circulation time, increases the solubility of the contrast agent in a physiological system, and/or increases R1 and R2 relaxivities, relative to un-PEGylated contrast agents. In an embodiment, the PEGs are bonded to the protein via an amino acid residue such as lysine (Lys), glutamic acid (Glu), aspartic acid (Asp), cysteine (Cys), carboxyl/amino terminals, or combinations thereof. As mentioned above, the position of the amino acids on the polymer should position the PEGs so that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
  • In another embodiment, a fusion protein/peptide/polymer or a non-degradable particle moiety can be added to the protein contrast agent with a linker to tune the correlation time for optimal contrast sensitivity, targeting (e.g., subcellular, cellular, tissue and organ selectivity), biodistribution (e.g., affiibody against to Her-2 was fused to CA1 as a targeted contrast agent to breast cancer), and/or bioelimination (e.g., using proteins with molecular weight less than 60 KDa). One of ordinary skill in the art may determine such linkers without undue experimentation.
  • An additional advantage of the contrast agent of the present disclosure is that targets of interest (e.g., specific tissues, specific organs, and biomarkers for molecular imaging of tissues and tissue growths such as cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors) can be imaged. The active targeting of contrast agents to specific organs or tissues can be achieved by attaching (directly or indirectly via linking) a compound (e.g., peptide, antibody, antigen, and the like) having an affinity for the target of interest. Thus, the contrast agent can be administered to the host, and the contrast agent will interact with the target of interest. Subsequently, the host can be imaged to determine the presence or absence, as well as the location of the target of interest. Additional details are provided herein.
  • Scaffold Proteins
  • Scaffold proteins suitable with the present disclosure include proteins or organic polymers containing amino acids. In an embodiment, the scaffold proteins can be modified. The scaffold proteins are inclusive of both natural amino acids and unnatural amino acids (e.g., beta-alanine, phenylglycine, and homoarginine, Gamma-carboxyglutamate (Gla)). In an embodiment, the amino acids are alpha-amino acids, which can be either of the L-optical isomer or the D-optical isomer. In an embodiment, the amino acids are D-optical isomers, as such isomers are less subject to proteolytic degradation. Such amino acids can be commonly encountered amino acids that are not gene-encoded, although preferred amino acids are those that are encodable. In an embodiment, a Near-IR functional group (e.g., Cy5.5, Cy7, Alexflour, and indocyanine green) for Near-IR detection is covalently bound to the scaffold protein, a PEG, and/or a targeting agent.
  • As mentioned previously, in some embodiments the scaffold proteins should include one or more amino acid residues (e.g., Lys, Glu, Asp, Cys, or combinations thereof) able to bond with the PEGs or otherwise modified. In this regard, the position of the amino acids on the protein should position the PEGs so that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
  • Various scaffold proteins may be used according to the disclosure, but in general they will be proteins, and organic polymers. More specifically, suitable scaffold proteins can be selected properties suitable for diagnostic applications. The scaffold protein for use with this disclosure may be of unitary construction (a particulate, a polychelant or a dendrimeric polymer). Scaffold proteins suitable with this disclosure may be selected without undue experimentation.
  • Embodiments of the present disclosure can include proteins such as CD2 proteins (a cell adhesion protein) that exhibit high stability against proteolysis, thermal conditions (Tm 67° C.), pH (2-10), and salt (0-4 M NaCl) denaturation. CD2 proteins can be suitable with this disclosure because such proteins are stable in physiological environments, have a topology suitable for the integration of at least one or multiple metal ion chelating sites, and typically have a relaxivity greater than 10 mM−1s−1 (some of them up to about 50 mM−1s−1). In addition, CD2 proteins can tolerate multiple surface mutations without unfolding the protein. In another embodiment, the CD2 proteins can be used as a host protein to design calcium binding sites. Examples using CD2 are described herein.
  • Fluorescent proteins are another class of preferred scaffold protein for this disclosure, as these proteins are stable in a physiological environment against proteolytic degradation and pH denaturation (pH 5-10). Such fluorescent proteins include an array of fluorescent proteins including those related to Aequorea. Suitable fluorescent proteins should have a useful excitation and emission spectra and may have been engineered from naturally occurring Aequorea victoria green fluorescent proteins (GFPs). Such modified GFPs may have modified nucleic acid and protein sequences and may include elements from other proteins. The cDNA of GFPs may be concatenated with those encoding many other proteins—the resulting chimerics are often fluorescent and retain the biochemical features of the partner proteins. Such proteins also are included in the disclosure.
  • One advantage of using fluorescent proteins is that contrast agents constructed from such proteins can be multi-functional probes. In this embodiment, the contrast agent constructed from fluorescent proteins can be screened using both fluorescence and MR imaging. This can be advantageous as such properties equip the contrast agent with both the fluorescence needed for fluorescence detection methods and sensitivity needed for the deep tissue detection from MRI. Such contrast agents are multifunctional contrast agents.
  • Other proteins may be used as scaffold proteins for this disclosure. In an embodiment, scaffold proteins are able to tolerate the addition of the metal ion binding site without substantial disruption to its structure. One of ordinary skill in the art can select a scaffold protein based on preferences without undue experimentation. Embodiments of this disclosure include natural calcium binding proteins with metal binding site such as calcium binding sites as scaffold protein proteins. These natural metal binding proteins such as calmodulin, calbindin D9K, troponin C, and parvalbumin, can be engineered to bind paramagnetic metal ions with very strong metal binding affinity thus are capable of enhancing image contrast by affecting water molecule proton relaxation rates. In addition, their selectivity over calcium and other physiologic metal ions such as zinc and magnesium is more than 105 fold higher, which is similar to that of clinically approved contrast agents such as DTPA or DTPA-BMA. More than one water molecule can be in the coordination shells and protein surface, and this likely contributes to their extremely high relaxivity. Furthermore, functional sites of these engineered proteins, such as binding to the target molecules by calmodulin, were altered and PEGylation of these engineered proteins increases solubility and reduced immunogenicity and increase relaxivity (See, Example 3).
  • Embodiments of scaffold protein sequences (SEQ ID Nos: 1-53) that can be included in the contrast agent are provided that include the unmodified scaffold proteins and modified scaffold proteins (insertions and/or deletions) for a variety of illustrative scaffold proteins that include metal ion binding sites. The scaffold protein sequences include one or more possible locations for attachment of PEGs, mutation sites, C-terminal sites for pegylation or conjugation of moieties (e.g., fluorescent dyes), and the like.
  • PEGs
  • As mentioned above, embodiments of the present disclosure include contrast agents where PEGs are attached to the protein via one or more amino acid residues such as Lys, Glu, Asp, Cys, carboxyl/amino terminals, or combinations thereof. In an embodiment, the PEGs are attached to amino acid residues so that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer. The PEGs can be attached to the amino acid residues through PEGylation processes known in the art. The PEGylation may, for example, be performed at a pH of about 7.5 to 9 or about 8 to 8.5.
  • The PEGs can be linear PEGs, multi-arm PEGs, branched PEGs, and combinations thereof. The molecular weight of the PEGs can be about 1 kDa to 100 kDa, about 1 kDa to 50 kDa, about 1 kDa to 40 kDa, about 1 kDa to 30 kDa, about 1 kDa to 20 kDa, about 1 kDa to 12 kDa, about 1 kDa to 10 kDa, or about 1 kDa to 8 kDa. It should be noted that the molecular weight can be any integer within any of the values mentioned above. When used in reference to PEG moieties, the word “about” indicates an approximate average molecular weight and reflects the fact that there will normally be a certain molecular weight distribution in a given polymer preparation. In an embodiment, 1 to 10 PEGs can be attached to the scaffold protein. In an embodiment, 2 to 6 PEGs can be attached to the scaffold protein. In an embodiment, 2 to 4 PEGs can be attached to the scaffold protein.
  • The PEGs can have additional functional groups to allow us to further modify the contrast agent by adding other moieties such as signal peptides (such as GRP signal peptide for targeting to prostate cancer).
  • Targeting Agent
  • In an embodiment, the contrast agent can have a specific affinity for a target by attaching (directly or indirectly, via the scaffold protein or the PEGs) a targeting agent to the contrast agent. In this regard, the term “affinity” means that the contrast agent is preferentially attracted to the target(s) as opposed to all other targets in the human subject. The contrast agent can be designed to have the affinity using one or more polypeptides (e.g., proteins) or chemical moieties on a target. If the targeting agent is attached to the scaffold protein (attached directly or indirectly), like the PEG (attached directly or indirectly), the targeting agent does not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
  • In an embodiment, a targeting agent can be attached (e.g., directly or indirectly) to the scaffold polymer or the PEG, where the targeting agent has an affinity for a target (e.g., a cell, a tissue, a protein, an antibody, an antigen, and the like). The targeting agent can include, but is not limited to, polypeptides (e.g., proteins such as, but not limited to, antibodies (monoclonal or polyclonal)), antigens, nucleic acids (both monomeric and oligomeric), polysaccharides, sugars, fatty acids, steroids, purines, pyrimidines, ligands, or combinations thereof, where the targeting agent binds or otherwise interacts with the target. In an embodiment, the targeting agent specifically interacts with a specific type of target or specific target and substantially (e.g., 90%, 95%, 99% or more specificity to the target or type of target) or completely excludes other targets. In an embodiment, the targeting agent has an affinity for one or more targets. In general, the target can include, but is not limited to, a cell type, a cell surface, extracellular space, intracellular space, a tissue type, a tissue surface, vascular, a polypeptide, a nucleic acid, a polysaccharide, a sugar, a fatty acid, a steroid, a purine, a pyrimidine, a hapten, a ligand, and the like, related to a condition, disease, or related biological event or other chemical, biochemical, and/or biological event of the sample or host. In an embodiment, the targeting agent can be selected based on the target selected and the environment the target is in and/or conditions that the target is subject to. In an embodiment, the targeting agent can include: a biomarker probe, a precancerous targeting agent, a cancer targeting agent, a tumor targeting agent, and a probe or agent that targets at least two of a biomarker, a precancerous cell, a cancer cell, and a tumor.
  • The targeting agent can be linked, directly or indirectly, using a stable physical, biological, biochemical, and/or chemical association. In an embodiment, the targeting agent can be independently linked to the scaffold polymer or the PEG using, but not limited to, a covalent link, a non-covalent link, an ionic link, a chelated link, as well as being linked through interactions such as, but not limited to, hydrophobic interactions, hydrophilic interactions, charge-charge interactions, π-stacking interactions, combinations thereof, and like interactions.
  • In an embodiment, the targeting agent can include, but is not limited to, (gastrin release peptide (GRP) that can bind to specific types of cancer receptors, i.e. GRP receptors, and, RGD peptides (Arg-Gly-Asp) (corresponding to integrin αvβ3 target). In an embodiment, molecules that can be targets include, but are not limited to, vascular receptors (e.g., Vascular endothelial growth factor receptor (VEGF-R)), extracellular matrix proteins (e.g., proteases, MMP, thrombin), cell membrane receptors (e.g., epidermal growth factor receptor (EGFR) (e.g., HER2)), intracellular proteins, enzymes (e.g., caspases and PSA), serum proteins (e.g., albumin), and the like.
  • Metal Ion Binding Sites
  • The affinity of the metal ion binding site may vary the contrast agent affinity for metal ions. Specifically, as affinity and sensitivity of the metal ion binding sites may be modified, the relaxivity and metal affinity of the contrast agent may be modified. Preferably, the metal ion binding site has optimal imaging properties including metal binding affinity, selectivity, relaxivity, NMRD profile, and water exchange rates.
  • Embodiments of the metal ion binding site of the present disclosure may be constructed using three methods:
  • (1) A computational design approach in which the metal ion binding site with a selectivity and affinity for a metal ion is engineered and rationally designed de novo based on optimal binding characteristics of a metal ion with other moieties; and
  • (2) A grafting method in which the metal ion binding site with a selectivity and affinity for a metal ion is engineered and constructed selectively by varying the primary, secondary, and tertiary of an identified binding site.
  • (3) Direct modification of natural metal binding proteins in which the metal binding affinity to the desired metal ions are significantly increased while the affinity to the physiological metal ions are decreased.
  • An engineered metal binding site can be created by a combination of more than one above mentioned methods.
  • The Computational Design Approach
  • The computational design approach focuses on designing a metal ion binding site de novo. This design approach focuses on using an algorithm to construct and engineer an optimal binding site. Preferably, the computation design approach is used to create optimal binding sites by, e.g., varying the coordination geometry of the site, the water number in the coordination shells, the ligand types, and the charges.
  • The computational design approach comprises the following steps:
  • (1) Accessing one or more databases having structural, coordination, and/or 3-dimensional structure or model on metal ion binding sites; or creating model structures based on the sequence homology;
  • (2) Generating one or more preliminary metal ion binding sites from portions of the structural data;
  • (3) Selecting rationally one or more suitable metal ion binding sites from the generated preliminary binding sites based on, e.g., coordination geometry; and
  • (4) Creating a metal ion binding site by tailoring and tuning the selected metal ion binding site.
  • The metal ion binding site may be incorporated into a scaffold protein, e.g., a fluorescent or CD2 protein. Further, such a method may be used to alter metal ion binding properties of proteins and generate new materials with various ion binding affinities.
  • More particularly, the method involves searching and accessing public and or private databases for preferred components of a metal ion binding site. Such databases that may be searched for the criteria or components may include public domain banks (e.g., NBCI or PubMed) or knowledge banks such as protein modeling structure data banks (e.g., Cambridge or RCSB Protein Data Bank Data Bank and BioMagResBank database) or data bank. Further, the database could include structural data from metal ion binding proteins whose structures have been characterized previously. One of ordinary skill in the art can identify databases and sources of material for databases suitable with this disclosure. Use of a computer obviously would greatly speed up the searching and is preferred.
  • These databases may be used to provide structural analysis of one to several thousand different small molecules or metal ions that bind to a protein. Such analysis may include local coordination properties, types of residues or atoms commonly used to bind a desired metal ion, chemical features (e.g., pKa or changes), the number of charged residues on a site, and the range or deviation of the known binding sites. Further, such analysis may include the environment, such as types of atoms, residues, hydrophobicity, solvent accessibility, shapes of the metal binding sites, electrostatic potentials, and the dynamic properties (e.g., B-factors or the order factors of the proteins) of the binding sites. Such analysis also may include whether a binding site for a particular metal ion is a continuous or discontinuous binding site.
  • Once preliminary metal ion binding sites are found, using the structural data and analysis, one or more suitable metal ion binding sites may be generated based on rational factors. Specifically, different search algorithms may be used to generate potential metal ion binding sites based on other key features in addition to, for example, the geometric descriptors. These key features include the properties of the original residues in the scaffold protein, ligand positions that are essential to protein folding, the number of the charged residues and their arrangement and number of water molecules in the coordination shell. The hydrogen bond network and the electrostatic interactions with the designed ligand residues also can be evaluated. Furthermore, the protein environments of metal ion binding sites can be analyzed according to solvent accessibility, charge distribution, backbone flexibility, and properties of scaffold proteins. Thus, one of ordinary skill in the art may rationally select a binding site based on desired parameters.
  • Once the metal ion binding sites are generated, a site may be tailored using two complementary approaches of computational design and grafting (see below). First, as discussed above, the metal ion binding site may be tailored using a grafting method in which the primary, secondary, tertiary, and/or quaternary structures are tuned. Second, the metal ion binding site may be tailored using a computational design approach. It is understood that one or both of these approaches may be used to tailor the binding site.
  • The computational design approach includes modifying the metal ion binding site by modifying residues in the scaffold of the metal ion binding site. In one embodiment, a geometric description of the ligands around a metal ion, a three-dimensional structure of the backbone of proteins, and a library of side-chain rotamers of amino acids (or atoms from the main chain) can identify a set of potential metal-binding sites using a computer. Using the geometric and graph description of a particular metal ion site, key ligand residues are carefully placed in the amino acid sequence to form the metal (metal ion) binding pocket. This binding pocket can be created automatically by the computer algorithm according to the coordination description and the user's preferred affinity.
  • The created potential metal ion binding sites can be optimized and tuned to specification. A backbone structure of the metal ion binding site with different degrees of flexibility may be used according to the need or the flexibility of the metal ion binding site. The designed metal ion binding sites are further filtered and scored based on the local factors, which may include the shape of the metal ion binding sites, locations, charge numbers, dynamic properties, the number of mutation needed, solvent accessibility, and side chain clashes. To achieve the maximums relaxivity, one to two oxygen atoms from the solvent water molecules in the coordination shell may provide additional coordination without reducing the required binding affinity and selectivity.
  • Stronger metal ion binding affinities of the designed sites may be developed based on several modeled factors that contribute to metal ion affinity. For example, the number of ligand residues is a factor to directly chelate a specific metal ion. In some cases, in order to have a strong metal ion affinity with a Kd necessary to measure a metal ion concentration, it is necessary to include residues from the protein frame for optimal metal ion binding. In other cases, the number of charged residues is able to change metal ion affinity. In still other cases, the ligand type is a factor as the binding preferences of a chelate may depend on the particular ligand type. Other factors, such as negatively charged environments, may contribute to the binding affinity of a metal ion binding protein and can be taken into account by those of ordinary skill in the art without undue experimentation. These charged residues could increase the water-exchange rate to avoid its limitation for the required relaxivity.
  • An illustrative version of this computational approach is the computerized (or otherwise automated) querying of one or more databases that comprise structural data on metal ion binding sites using selected criteria relevant to the metal ion binding site, generating at least one preliminary metal ion binding site from the database information based on compatibility with the selected criteria, and selecting one or more suitable metal ion binding sites from the preliminary metal ion binding sites based on optimal compatibility with the selected criteria. Once a suitable metal ion binding site is selected, the nucleic acid sequence of the selected metal ion binding site is obtained, tailored, and operatively linked with a scaffold protein sequence, whereby the nucleic acid sequence of the selected metal ion binding site is tailored so to achieve the metal ion binding site having a desired specificity for the metal ion. Further, a nucleic acid sequence encoding the preliminary binding sites can be generated from the structural or model data. The computational approach also can be used to produce the metal ion binding site.
  • The computational approach can be performed on or by a system comprising at least one database that comprises the structural data on metal ion binding sites, an algorithm for generating the preliminary metal ion binding sites from portions of the structural or model data using selected criteria relevant to the metal ion binding site and rating the preliminary metal ion binding sites based on specificity for a selected metal ion, and a computer for executing the algorithm so as to query the databases to generate the preliminary metal ion binding sites. The algorithm generally is a relatively simple searching algorithm that will query the databases based on inputted criteria.
  • The Grafting Method
  • The grafting method focuses on engineering and constructing a metal ion binding site by modifying the primary, secondary, tertiary, and/or quaternary structure of an identified binding site. By selectively manipulating the structure of the binding site, it is possible to obtain a metal ion binding site that can be engineered into a scaffold protein, e.g., CD2 or fluorescent protein, without significantly denaturing the protein. Using the grafting method, it is possible to achieve a binding site that has a stronger preference for one metal ion over another metal ion. Such modifications may allow for improved contrast abilities.
  • Initially, an identified binding site for use with the grafting method may be any continuous sequence site that has some affinity for a metal ion. Such binding sites may derive from either known binding peptides such as an individual EF-hand site or from short fragments that have demonstrated the ability to bind specific metal ions such as alpha-lactalbumin. Such peptides may be highly conserved in nature and prevalent throughout nature or may be unnatural but known to have an affinity for a particular metal ion. One of ordinary skill in the art is able to identify binding sites with affinity for a metal ion without undue experimentation.
  • Once the binding site has been identified, the primary structure of the metal ion binding site may be altered and tuned to achieve a metal ion binding site with improved binding characteristics. For example, more charged ligand residues such aspartate and glutamate may be engineered by inserting codon(s) into the metal ion binding site so as to tune the responsiveness of the site or the scaffold protein. The inclusion of additional charged ligands can allow the contrast agent to achieve an affinity for selected paramagnetic metal ions and to have a desired selectivity. Further, one or two water molecules can also be introduced into the coordination shell by removing or modifying ligand residues and their environments. Further other mutations to the primary structure include removing or adding amino acids to change properties such as flexibility or rigidity of the site. Adding or removing amino acids from the binding site alters the primary structure of the binding site.
  • The secondary structure of the metal ion binding site, that is the spatial arrangement of amino acid residues that are near one another in linear sequence, may be modified to tune the sensitivity and responsiveness of the metal ion binding site. The residues on the site itself, the flanking or the neighboring structures such as helices, beta strands, or turns may be modified by changing properties such as hydrophobicity, salt bridges, secondary structure propensity (e.g., helicity and β-sheets), and charge interactions with different amino acids, which all may inherently change the secondary structure.
  • The tertiary structure of the metal ion binding site may be modified to further tune the sensitivity and responsiveness of the metal ion binding site. The affinity of the metal ion binding site for the metal ion may be varied by selectively manipulating and adding helices, loops, bridges and/or linkers and chemical properties such as hydrogen bonding, electrostatic interactions and hydrophobic interactions. In fact, such variations in tertiary structure may add stability and affinity by increasing secondary structure propensity, adding charge interaction of the side chains, and by stabilizing the metal ion binding coordination chemistry. As such, it may be possible to increase or decrease the binding affinity of the continuous binding site by tuning the tertiary structure of the metal ion binding site. In addition, the dynamic properties can be modified by increasing the packing of the protein and replacing residues with amino acids or other moieties with more rigid (e.g., Pro) or flexible (e.g., Gly) properties,
  • One method of directly altering the primary, secondary, and/or tertiary structure of the metal ion binding site is by altering the charges in the site. As the charges in any binding site have a significant role in the structure of the site, changing the charges or charge ratio may have significant impact on the structure of the site. In addition, as the charged side chains exhibit a strong influence on the metal ion binding affinity even though they are not directly involved as ligands, the variation of these chains results in variations in metal ion binding affinities and selectivity. A metal ion binding site may have stronger affinities to and better selectivity for a desired metal ion over a competitive metal ion by designing or modifying the site, e.g., changing the number of charged ligand residues to form metal ion binding pockets. For example, the metal ion binding affinity of the metal ion binding site may be varied by changing the charged side chains that are present on the metal ion binding site and/or the neighboring environment. The replacement of charged residues such as aspartate or glutamate with a residue such as alanine may dramatically reduce the binding affinity for the metal ion by up to 100 times.
  • In the case of multifunctional contrast agents, e.g., where the contrast agent is a fluorescent protein, it can be a factor to induce the metal binding site without altering significantly the chromophore environment to reduce the fluorescent signal. These metal binding sites can be added at remote locations away from the chromophore or simply fusion to the fluorescent moieties. Such locations can be evident from the sequence and protein folding.
  • In another embodiment, the grafting approach may be used with the design approach to create an optimal metal binding site. For example, metal binding sites can be created by using part of continuous site and part of ligand residues created by computer design. The loops or any sequences of the proteins can be removed or modified to achieve optimal required binding affinity, metal selectivity, relaxivity and stability.
  • Thus, by varying the primary, secondary, and/or tertiary structure of the metal ion binding site, it is possible to achieve a metal ion binding site with desired specificity and affinity and more importantly contrast abilities.
  • The Modification Method of Natural Metal Binding Proteins
  • Natural metal binding proteins' their metal binding affinity can be altered by directly modifying the proteins such as the addition of metal ligand residues in the calcium binding proteins to increase metal binding affinity to lanthanides. In an embodiment, fragments and/or domains of the natural metal binding proteins encompassing metal binding sites can also serve as scaffold protein of the contrast agents if they exhibit strong metal binding affinity for Ln3+ or other paramagnetic metal ions, serum stability, and desired relaxation properties. The affinity to natural metal ions such as physiological metal ions, e.g., calcium, zinc, and magnesium, will be significantly reduced by deleting metal binding ligand residues or reducing the cooperativity between coupled metal binding sites. As noted in Example 3, the calcium binding sites in the natural calcium binding protein such as calmodulin and parvalbumin were modified so that the modified proteins have a strong metal binding affinity to lanthanides. On the other hand, the metal selectivity for lanthanides over calcium, magnesium and zinc are very high. If it is necessary, the molecular recognition sites of these natural calcium binding proteins can be altered by deletion at the active sites or PEGylation.
  • In addition, sequences for N- and C-terminal domains of calmodulin and its variants are listed that can be serve as a protein contrast agents (See sequences included herein). Additional modifications can performed to reduce their intrinsic biological function, avoid immunogenicity, increase serum stability, and targeting capability.
  • Selecting Metal ion Binding Sites in the Scaffold Protein
  • The metal ion binding sites may be selectively introduced into numerous sites of a scaffold protein without substantially impairing its secondary structure. A number of methods for identifying integration sites in proteins, such CD2 proteins, fluorescent proteins (e.g., GFP, YFP, CFP, and RFP) are known in the art, including, for example, site directed mutagenesis, insertional mutagenesis, and deletional mutagenesis. Other methods, including the one exemplified below and in the Examples, are known or easily ascertained by one skilled in art.
  • The sites of the fluorescent protein that can tolerate the insertion of a metal ion binding site also may be determined and identified by gene manipulation and screening. By generating mutant proteins and by manipulating the DNA sequence, it is possible to obtain a variety of different insertions, which then may be screened to determine whether the protein maintains its intrinsic activities. Preferably, sites that remove or interfere with the intrinsic fluorescence of the fluorescent protein are not optimal and may be screened out. Variants identified in this fashion reveal sites that can tolerate insertions while retaining fluorescence.
  • The metal ion binding sites for use with scaffold proteins may be selected by considering five criteria so to as optimize the local properties of the metal binding site, the fluorescent protein, and the protein environment. First, the geometry of the metal ion binding site should have relatively minor deviations from the desired coordination geometry. Second, negatively charged residues should be varied by no more than 3-5 charges according to the desired affinity for metal ion (Kd). Third, the water coordination shell of the metal ion chelating sites should be able to coordinate at least 1-2 water molecules. Fourth, the residues from the loops between the secondary structures with good solvent accessibility are desired for both the folding of the protein and the fast kinetics required for the contrast agent.
  • The mutation or the introduction of the metal ion binding site should not substantially interfere with the synthesis and folding of the protein. More particularly, the introduction of the metal ion binding site does not interfere with either post-translational chromophore formation or intermolecular interactions required for stabilizing the chromophores and folding of the protein frame. Furthermore, the introduced side chain should not be overpacked and should not clash with the protein frame of the scaffold protein (e.g., the fluorescent protein). The direct use of chromophore residues as chelating sites is not preferred but is within the scope of this disclosure.
  • In an embodiment, the metal binding sites in the natural metal binding proteins can be directly modified to have proper metal binding affinity to the desired metal ions.
  • Metal Ions
  • One or more metal ions are atoms and ions, including the respective isotopes and radioisotopes, that can bind to proteins or peptides. A metal ion may bind reversibly or irreversibly and such a bond may be covalent or non-covalent. While Gd3+ is used in some embodiments of this disclosure as an exemplary metal ion, it is understood that metal ions suitable with this disclosure include, but are not limited to metal ions including Group IIA metal ions, transition metal ions, and Lanthanide Series ions. Exemplary metal ions include, but are not limited to, the ion, isotope, and/or radioisotope forms of magnesium, calcium, scandium, titanium, manganese, iron, boron, chromium, cobalt, nickel, cooper, zinc, gallium, strontium, yttrium, strontium, technetium, ruthenium, indium, hafnium, tungsten, rhenium, osmium, and bismuth. Exemplary radioisotopes include, but are not limited to, 64Cu, 67Cu, 67Ga, 68Ga, 88Y, 89Sr, 90Y, 97Ru, 99mTc, 103Ru, 111In, 153Sm, 186Re, 188Re, 203Pb, 211Bi, 212Bi, 213Bi, and 214Bi.
  • The metal ions chosen to be chelated by the contrast agents depend in part on the diagnostic role of the metal ion. Metals that can be incorporated, e.g., through chelation, include lanthanides and other metal ions, including isotopes and radioisotopes thereof. For MR imaging applications, the preferred metal ion is gadolinium (III). One of ordinary skill in the art can select a metal ion for chelation, based on the intended diagnostic application, without undue experimentation.
  • As mentioned, the choice of metal ions to be held in chelate complexes by the contrast agents of the disclosure depends upon the diagnostic technique for which the agent is to be used. For MRI or MRS applications, the metal ions should be paramagnetic, and preferably non-radioactive. For X-ray and ultrasound imaging, heavy metal ions, e.g., with atomic numbers of at least 37, and in an embodiment, at least 50, should be used, again preferably non-radioactive species. For scintigraphy the metal ions should be ions of radioactive isotopes. For MR, X-ray, EIT or magnetometric imaging, one may use chelating groups to bind to heavy metal clusters (e.g., polyoxoanions and full or partial sulfur analogues) or to iron oxides or other superparamagnetic polyatomic species.
  • Methods of complexing metal ions with chelants and polychelants are known to those with ordinary skill in the art. Metal may be incorporated into the contrast agent, i.e., the tailored binding sites, by direct incorporation, template synthesis, and transmetallation. Preferably, the metal ion is chelated into the contrast by direct incorporation, which involves titration with solution of sub-stoichiometric levels up to full incorporation.
  • In an embodiment, one or two or more metal ions can bind to the contrast agent. In an embodiment, the contrast agent includes one or two or more metal ion binding sites. In an embodiment, each of the metal ion binding sites binds to the same metal ion. In an embodiment, each of the metal ion binding sites binds to a different metal.
  • Methods of Use
  • Embodiments of the contrast agents (e.g., contrast agents including PEGs or not including PEGs, and/or including targeting agents or not including targeting agents) can be used in any one of a number of methods. Embodiments of this disclosure include, but are not limited to: methods of detecting, studying, monitoring, evaluating, and/or screening, diseases, conditions, and other biological events in vivo or in vitro. The conditions can include, but are not limited to, altered growth rate of tissues, cancerous transformation of tissues, inflammation or infection of a tissue, altered volume of a tissue, altered density of a tissue, altered blood flow in a tissue, altered physiological function, altered metabolism of a tissue, loss of tissue viability, presence of edema or fibrosis in a tissue, altered perfusion in tissue, and combinations thereof. In particular, embodiments of the present disclosure include: methods of imaging tissue; methods of diagnosing the presence of a disease, precancerous cells or tissue, cancer cells or tissue cancer, and tumors, as well as related biological events; methods of monitoring the progress of a disease, precancerous cells or tissue, cancer cells or tissue cancer, and tumors, as well as related biological events; and the like.
  • Embodiments of the present disclosure include, but are not limited to, imaging, detecting, studying, monitoring, evaluating, and/or screening biological materials (e.g., organs, tissues, tumors, cells, and the like), in vivo or in vitro. The tissue types that can be studied using the methods of the present disclosure include, but are not limited to, myocardial tissues, nervous tissue, lymphoid tissue, skeletal and smooth muscle tissue, bones and cartilages, tissues of various organs (e.g., the kidney, the liver, the spleen, the prostate, the uterus, the testicles, and the ovaries), and select portions of each.
  • Embodiments of the methods can use one or more types of detecting or imaging systems such as, but not limited to, magnetic resonance imaging (MRI), SPECT, PET, ultrasound, X-ray, CAT, optical imaging, and combinations thereof. In an embodiment, the contrast agent is a multimodality contrast agent that includes a polymer having optical properties (GFP). Thus, the polymer having optical properties can be detected using optical imaging, while the metal can be detected using another technique such as a MRI system.
  • In general, embodiments of the contrast agent are administered to a host using one or more techniques or routes (e.g., oral, mucosal, parenteral, and the like). After an appropriate amount of time, the host can be introduced to an appropriate detection or imaging system. The detection or imaging system can detect the contrast agent. In particular, the detection or imaging system can detect the location(s) of the contrast agents, the concentration of the contrast agent, and the like. The information obtained from the detection or imaging system can be used to create or form an image of the host or a portion thereof. The image would include the position and/or concentration of the contrast agent in the host.
  • In an embodiment, the contrast agents can be used to study, image, diagnose the presence of, and/or treat cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors. For example, the presence and location of the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors can provide insight into the appropriate diagnosis and/or treatment. It should be noted that contrast agents could include targeting agents specific for other diseases or conditions so that other diseases or conditions can be imaged, diagnosed, and/or treated using embodiments of the present disclosure. In an embodiment, other diseases and/or conditions can be studied, imaged, diagnosed, and/or treated in a manner consistent with the discussion below as it relates to cancerous cells, precancerous cells, cancer, and/or tumors.
  • In an embodiment, the contrast agent can be used to study, image, diagnose the presence of, and/or treat cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors. In studying, imaging, diagnosing, and/or treating cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors in a host, the contrast agent is administered to the host in an amount effective to result in uptake of the contrast agent into the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors. After administration of the contrast agent, the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors that takes up the contrast agent is detected using an appropriate imaging system. Embodiments of the present disclosure can non-invasively image the cancerous cells or tissue, precancerous cells or tissue, cancer, or tumors throughout a host.
  • In an embodiment, the contrast agent includes a targeting agent having an affinity for a specific cancer. Detecting the presence of the contrast agent, in particular, the presence of the contrast agent at the typical location of the specific cancer can be used in the diagnosis of the presence of the cancer (or vice versa). Imaging the host over a time period (e.g., days, weeks, months, or years) can provide information about the progression of the cancer or other disease or condition.
  • The contrast agent or compositions including the contrast agent may be administered to a subject in an amount effective to achieve the desired result at the appropriate dosages and for the desired periods of time. An effective amount of the contrast agent or compositions may vary according to factors such as the age, body weight, general health, sex, and diet of the host; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; the existence of other drugs used in combination or coincidental with the specific compositions employed; the ability of the composition to elicit a desired response in the subject; and like factors well known in the medical arts. An effective amount is also one in which any toxic or detrimental effects (e.g., side effects) of the contrast agent or compositions are outweighed by the therapeutically or diagnostically beneficial effects. The contrast agent or compositions of the disclosure may be administered at a concentration of, for example, about 1 to 3.0 μmole/kg or about 6-20 mM.
  • Dosage Forms
  • Unit dosage forms of the contrast agents of this disclosure may be suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., intramuscular, subcutaneous, intravenous, intra-arterial, or bolus injection), topical, or transdermal administration to a patient. Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as hard gelatin capsules and soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • The composition, shape, and type of dosage forms of the contrast agents of the disclosure typically vary depending on their use. For example, a parenteral dosage form may contain smaller amounts of the active ingredient than an oral dosage form used to treat the same condition or disorder. These and other ways in which specific dosage forms encompassed by this disclosure vary from one another will be readily apparent to those skilled in the art (See, e.g., Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton, Pa. (1990)).
  • Typical compositions including the contrast agent and dosage forms of the compositions of the disclosure can include one or more excipients. Suitable excipients are well known to those skilled in the art of pharmacy or pharmaceutics, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient. For example, oral dosage forms, such as tablets or capsules, may contain excipients not suited for use in parenteral dosage forms. The suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients can be accelerated by some excipients, such as lactose, or by exposure to water. Active ingredients that include primary or secondary amines are particularly susceptible to such accelerated decomposition.
  • The disclosure encompasses compositions including the contrast agent and dosage forms of the compositions of the disclosure that can include one or more compounds that reduce the rate by which an active ingredient will decompose. Such compounds, which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers. In addition, pharmaceutical compositions or dosage forms of the disclosure may contain one or more solubility modulators, such as sodium chloride, sodium sulfate, sodium or potassium phosphate, or organic acids. An exemplary solubility modulator is tartaric acid.
  • Like the amounts and types of excipients, the amounts and specific type of active ingredient in a dosage form may differ depending on various factors. It will be understood, however, that the total daily usage of the compositions of the present disclosure will be decided by the attending physician or other attending professional within the scope of sound medical judgment. The specific effective dose level for any particular host will depend upon a variety of factors, including for example, the activity of the specific composition employed; the specific composition employed; the age, body weight, general health, sex, and diet of the host; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; the existence of other drugs used in combination or coincidental with the specific composition employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of the composition at levels lower than those required to achieve the desired effect and to gradually increase the dosage until the desired effect is achieved.
  • Kits
  • This disclosure encompasses kits, which may include, but are not limited to, a contrast agent and directions (instructions for their use (written or electronic)). The components listed above can be tailored to the particular disease or condition to be monitored. The kit can further include appropriate reagents known in the art for administering various combinations of the components listed above to the host organism or patient.
  • EXAMPLES Example 1 Rational Design of Protein Based MRI Contrast Agents Introduction:
  • This Example describes the rational design of a novel class of magnetic resonance imaging contrast agents with an engineered protein chelated with gadolinium. The design of protein based contrast agents involves creating high coordination Gd3+ binding sites in a stable host protein using amino acid residues and water molecules as metal coordinating ligands. Designed proteins show strong selectivity for Gd3+ over physiological metal ions such as Ca2+, Zn2+, and Mg2+. These agents exhibit a 20-fold increase in longitudinal and transverse relaxivity values over the current clinically used contrast agent, Gd-DTPA. They provide strong contrast enhancement in vivo with much longer vascular retention time. These protein contrast agents have good biocompatibility and potential functionalities may extend MRI applications in targeting disease markers.
  • Magnetic resonance imaging (MRI) is a non-invasive technique providing high resolution, three-dimensional images of morphological features as well as functional and physiological information about tissues in vivo. It is capable of detecting abnormalities in deep tissues and allows for whole body imaging. It has emerged as a primary diagnostic imaging technique for human diseases.1,2 Exogenous MRI contrast agents are often used to enhance the contrast between pathological and normal tissues by altering the longitudinal and transverse (i.e., T1 and T2) relaxation times of water protons.3-5 Gadolinium (Gd3+) is the most frequently used MRI contrast agent due to its high magnetic moment, asymmetric electronic ground state and potential for increased MRI intensity.6,7 The relaxivity (unit capability of the agent to change the relaxation time) of a contrast agent is dependent on several factors including the number of water molecules in the coordination shell, the exchange rate of the coordinated water with the bulk water, and the rotational correlation time τR of the contrast agent.8-10. The MRI contrast agent can have: 1) high relaxivity for high contrast-to-noise ratio (CNR) and dose efficiency, 2) thermodynamic stability, especially metal selectivity for the target ions over excess physiological metal ions, to minimize the release of toxic paramagnetic metal ions, 3) adequate vascular, tissue retention time to allow imaging, and 4) proper excretion from the body.
  • To date, the most commonly used MRI contrast agent in diagnostic imaging is Gd-DTPA, or its derivatives such as Gd-DTPA-BMA. With an intrinsic rotational correlation time, τR, of 100 picoseconds, these small molecular gadolinium contrast agents have longitudinal and transverse proton relaxivities, r1 and r2, less than 10 mM−1s−1, much lower than the theoretically maximal value (>100 mM−1s−1).9, 11 In addition, these small molecule contrast agents exhibit very short blood circulation (within several minutes) and tissue retention time, limiting some MRI applications that require longer data collection time.12 To increase correlation time, τR, small contrast agents were covalently or non-covalently conjugated to macromolecules such as linear polymers,13 dendrimers,14, 15 carbohydragates,16 proteins,17-21 viral capsids,22 and liposomes23. However, conjugation yields limited improvement due to internal mobility and restricted water exchange rate (FIG. 1.1 a).11 An increase in relaxivity was observed when Gd3+ binds to calcium binding peptides24 or proteins such as concannavalin A and bovine serum albumin (BSA).6 However, the application of these short peptides or proteins as MRI contrast agents is limited due to their weak metal binding affinity for Gd (Kd˜100 μM for Eu3+) and dynamic flexibility.24,25
  • This Example describes the development of a new class of MRI contrast agents with significantly improved relaxivity using rational design of Gd3+-binding proteins. This class of contrast agents was created by designing the metal binding sites into a stable host protein with desired dynamic properties and metal selectivity to increase relaxivity by optimizing local τR. This approach provides a new platform for developing MRI contrast agents with high relaxivity and functionality.
  • Materials and Methods
  • Determination of r1 and r2 Relaxivity Values. Relaxation times, T1 and T2, were determined at 1.5, 3, 9.4 Tesla using a Siemens whole-body MR system (1.5, 3T) or a Bruker MRI scanner (9.4T). T1 was determined using inversion recovery and T2 using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence. The contrast agent samples (200 μl) with different concentrations were placed in eppendorf tubes. The tubes were placed on a tube rack, which was placed in MRI scanners for the measurement of relaxation times. r1 and r2 were calculated based on r1(mM−1S−1)=(1/T1s−1/T1c)/C and r2(mM−1S−1)=(1/T2s−1/T2c)/C, where T1s and T2s are relaxation times with contrast agent and T1c and T2c are relaxation times without contrast agent. C is the concentration of contrast agent in mM (the measured Gd3+ concentrations by ICP-MS).
  • Measurement of Water Coordination Number by Terbium Life Time Luminescence. The number of water ligands coordinated to Gd3+-CA1.CD2 complex was determined by measuring Tb3+ luminescence decay in H2O or D2O. Tb3+ excited state lifetime was measured using a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Following excitation at 265 nm with a XenoFlash (Photon Technology International, Inc.), Tb3+ emission was monitored at 545 nm in a time series experiment in both H2O and D2O systems. Luminescence decay lifetime was obtained by fitting the acquired data with a mono-exponential decay function. H2O in CA1.CD2 solution was replaced with D2O by lypholization and re-dissolved in D2O at least three times. A standard curve correlating the Δkobs with water number under our experimental conditions was established by using well-characterized chelators, such as EDTA (q=3), DTPA (q=1), NTA (q=5), and Aquo Tb3+ (q=9) solution with R2=0.997.26,27 Water number coordinated to Tb3+-CA1.CD2 complex was then obtained by fitting the acquired Δkobs value to the standard curve.
  • Gd3+-binding Affinity Determination. Gd3+-binding affinity of CA1.CD2 was determined by a competition titration with Fluo-5N applied as a Gd3+ indicator. The fluorescence spectra of Fluo-5N were obtained with a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Fluo-5N emission spectra were acquired at 500 nm to 650 nm with an excitation at 488 nm. Gd3+-binding affinity of Fluo-5N, Kd1, was first determined by a Gd3+ titration with Gd3+ buffer system of 1 mM nitrilotriacetic acid (NTA). Free Gd3+ concentration was calculated with a NTA Gd3+-binding affinity of 2.6×10−12 M.28 Fluo-5N was mixed with Gd3+ in 1:1 ratio for a competition titration. The experiment was performed with a gradual addition of CA1.CD2. An apparent constant, Kapp, was estimated by fitting the fluorescence emission intensity of Fluo-5N at 520 nm with different CA1.CD2 concentrations as a 1 to 1 binding model. Gd3+-binding affinity of CA1.CD2, Kd2, was calculated with the following equation:
  • K d 2 = K app K d 1 K d 1 + [ Fluo - 5 N ] T ( 1 )
  • Mouse MR Imaging. Care of experimental animals was in accordance with institutional guidelines. CD-1 mice (25-30 g, four mice were imaged) were anesthetized with an isoflurane gas mixture. The anesthetized animal was positioned and stabilized with soft-supporting material (e.g., foam) in the scanner in the coil cradle and was kept warm during the MRI scan. The mice were scanned prior to the administration of any contrast agent (pre-contrast). Approximately 50 μl of Gd-CA1.CD2 (˜1.2 mM) or Gd-DTPA (˜300 mM) were injected into the animal via the tail vein. MR images were collected at different times (indicated). For T1 weighted imaging at 3T, spin echo sequence with TE/TR=15 ms/500 ms was employed. Rectangular Field of View (FOV) at 100/40 mm, an acquisition matrix of 1962 and 1.1 mm slice thickness without gap were used. Images were collected from both transverse and coronal sections. The in-plane resolution of images was less than 0.5 mm after they were reconstructed to the matrix of 1962. For T2 weighted imaging at 9.4T, MR images were recorded using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence. The data were collected and processed by Dicomworks software. The MR signal intensity in several organs was ascertained by the average intensity in ROIs or points within the organs. Signal intensity for each organ was normalized to that of the leg muscle.
  • Blood Circulation Time, Tissue Retention Time, and Bio-distribution. CD-1 mice (25-30 g, a group of four mice were tested) were anesthetized with isoflurane. Appropriate dosages of Gd-CA1.CD2 or Gd-DTPA were i.v. injected (via tail vein). Blood (˜50 μl) samples were collected via orbital sinus of the mouse at different time points. The mouse was euthanized at the final time point. Tissue samples from kidney, liver, heart, and lung, were collected. Serum samples were prepared from the collected blood. For the bio-distribution analyses, the animals were euthanized at single time point (indicated) after i.v. administration of the contrast agent (indicated). The organ/tissue samples were collected. Tissue extracts were freshly made from collected samples using commercially available tissue extracting kits (Qiagen). CA1.CD2 was detected and quantified by immunoblotting and Sandwich-ELISA using a monoclonal antibody (OX45, detecting antibody) and a home made polyclonal antibody (PabCD2, capture antibody). A series of known amounts of CA1.CD2 samples mixed with blank mouse serum or tissue extracts were used as standard in Sandwich-ELISA. ELISA signal from HRP was monitored using a Fluorstar fluorescence microplate reader. For quantification of Gd3+ in the serum and tissue samples one hour (or indicated times) after the contrast agent administration, animals were sacrificed and critical organs were collected, and the tissues were then digested with concentrated nitric acid at 120-130° C. with proper amount of 157Gd spike as an internal marker. The digested solution was analyzed by ICP-MS (Element 2) using an isotope dilution method.
  • Toxicity Analyses. The MR imaged CD-1 mice that received the i.v. administered Gd-CA1.CD2 (at a dose of ˜2.4 μmol/kg) were returned to their cages (one mouse per cage). The mice were observed for five days and were euthanized at the end of the fifth day. Tissue samples from kidney, liver, spleen, and lung were collected. Gd3+ ion contents in the tissue samples were analyzed by ICP-MS (see above paragraph).
  • Two groups of mice were used to examine potential renal and/or liver damage by Gd-CA1.CD2. One group of mice received (i.v. tail veil) 50 μl of saline-buffer (as control). Another group received (i.v. tail veil) Gd-CA1.CD2 at 4 μmole/kg. The mice were observed for 48 hours and were euthanized. Blood samples were collected from the experimental mice. Serum samples were prepared from the collected blood. Liver enzymes in serum samples, including Alanine transaminase (ALT), Alkaline phosphatase (ALP), Aspartate transaminase (AST), Gamma glutamyl transpeptidase (GGT), and bilirubin and urea nitrogen were analyzed by a commercially available source (MU Research Animal Diagnostic Laboratory). All clinical chemistry parameters were measured on an Olympus AU 400 analyzer.
  • Cytotoxicity was analyzed by MTT assay of the cells that were treated with Gd-CA1.CD2 at appropriate doses (indicated in figure). The cells were grown under normal growth medium in 96 well plates. Gd-CA1.CD2 or saline-phosphate buffer was added to the cell culture medium. The cells were incubated for appropriate times. A standard MTT assay was employed to assess the cell growth status of the treated cells.
  • Serum stability. CA1.CD2 (40 μM) in complex with Gd3+ was incubated with 75% human serum over 3 or 6 hours at 37° C. The degradation of the protein (disappearance of 12 kDa protein band) was analyzed by SDS-PAGE and visualized by coomassie blue staining. In parallel, the degradation of the protein was also analyzed by immunoblot using antibodies OX54 or PabCD2. The identities of the 12 kDa bands as CA1.CD2 were always verified by immunobloting using antibody PabCD2.
  • Results
  • Rational Design of Gd3+-binding Proteins. FIG. 1.1 shows the simulation of the dependence of r1 and r2 on the rotational correlation time, τR, of a contrast agent at different magnetic field strengths according to the theory developed by Blombergen, Solomon6,7 (for detailed simulation procedures, please see on-line supporting materials). For small molecules such as Gd-DPTA with τR at hundreds of ps, the relaxivity is <10 mM−1s−1 regardless of how the other parameters are adjusted. On the other hand, the simulation clearly suggests that contrast agents with τR of 10-50 ns have the highest r1 and r2 values at clinically relevant magnetic field strengths from 0.47-4.7 Tesla (T).11 Thus, we envisioned that high-relaxivity MRI contrast agents can be developed by directly designing Gd3+ binding sites in proteins with desired τR. Coordinating Gd3+ ions directly to the rigid protein frame eliminates the high internal mobility associated with chelator-macromolecule conjugates (FIG. 1.1 a).
  • We chose domain 1 of rat CD2 (referred to as CD2), a cell adhesion protein with a common immunoglobin fold, as a scaffold (FIG. 1.1 c). CD2 protein exhibits strong stability against pH changes and excellent tolerance against various mutations,29 which are essential features of functional protein engineering. In addition, it has a compact structure with rotational correlation time, τR, of ˜10 ns, corresponding to optimal relaxivity for the current clinically allowed magnetic field strength.30 Moreover, its molecular size (12 kDa) is suitable for good tissue penetration and easy renal exclusion.17
  • We next designed a series of Gd3+ binding sites into CD2 using computational methods.32,33 The design was based on the established structural parameters obtained from detailed analysis of metal binding sites in over 500 small chelators and metalloproteins. Gd3+, Tb3+, La3+ and other Ln3+ ions have coordination properties similar to those of Ca2+ with a strong preference for oxygen ligand atoms.34 Small chelators usually have on average 9.3 and 6.9 total coordinating atoms for Gd3+ and Ca2+, respectively. For example, DTPA has 5 oxygen ligand atoms and 2 nitrogen ligand atoms. For macromolecules such as proteins, the coordination atoms are almost always oxygen atoms, and the coordination numbers are lower than small chelators with an average of 7.2 for Ln3+ and 6.0-6.5 for Ca2+. These effects are possibly due to steric crowding and sidechain packing.34 Previously, we successfully designed Ca2+ and Ln3+ binding sites in a scaffold protein with strong selectivity over excess physiological metal ions.35 Structure determination by solution NMR revealed that the actual coordination geometry in a designed variant is the same as our design, verifying the computational methods and the design strategy of metal-binding sites in proteins.31
  • The designed proteins were named CA1.CD2-CA9.CD2 reflecting Gd-binding sites at different locations. FIG. 1.1 c shows an example of designed Gd3+-binding protein CA1.CD2 with a metal binding site formed by the six potential oxygen ligands from the carboxyl side chains of Glu15, Glu56, Asp58, Asp62 and Asp64. Based on our studies of charged residues in the coordination shell,36 we placed 5 negatively charged residues to provide these six oxygen ligand atoms in the coordination shell of CA1.CD2 to increase the selectivity for Gd3+ over Ca2+. To achieve the desired relaxation property, one position of the metal binding geometry was left open in the design to allow fast water exchange between the paramagnetic metal ion and the bulk solvent (FIG. 1.1 a). The Gd3+-binding site has minimal internal flexibility as the ligand residues originate from rigid stretches of the protein frame. To test the requirement for rigid embodiment in achieving high relaxivity, another Gd3+-binding protein, CA9.CD2, was engineered by fusing a continuous cation-binding EF-hand loop from calmodulin with flexible glycine linkers to the host protein.37,38 This protein mimics previously reported highly flexible chelate-based contrast agents conjugated to macromolecules.24,25
  • All of the designed Gd3+ binding proteins were expressed in E. coli and subsequently purified by procedures previously published from our laboratory.30,31 All of the designed proteins form the expected metal:protein complex as demonstrated by ESI-Mass spectrometry (FIG. 1.5). Since metal selectivity for Gd3+ over other physiological metal ions is for minimizing the toxicity of the agents,39,40 we measured metal binding constants using dye-competition assays with various chelate-metal buffer systems (Table 1). Low limit metal binding affinities of the proteins were also estimated based on Tb3+-sensitized FRET and competition assays. CA1.CD2 exhibited disassociation constants (Kd) of 7.0×10−13, 1.9×10−7, 6×10−3, and >1×10−2 M for Gd3+, Zn2+, Ca2+, and Mg2+, respectively. The selectivity Kd ML/Kd GdL for Gd3+ over physiological divalent cations Zn2+, Ca2+, and Mg2+ are 105.34, >109.84, and >1010.06, respectively. The metal selectivity of CA1.CD2 is significantly greater than or comparable to that of FDA approved contrast agents DTPA- and DTPA-BMA40 (Table 1). The high metal binding selectivity of CA1.CD2 was further supported by the observation that r1 and r2 of Gd-CA1.CD2 were not altered in the presence of excess Ca2+ (10 mM) (FIG. 1.2). Further assays showed that potential chelators in serum, such as phosphate (50 mM), were not able to remove the Gd3+ from the protein. This is considered for in vivo applications of the contrast agent as the phosphate concentration in serum is maintained at ˜1.3 mM.6,9 The stability of a contrast agent in blood circulation is another factor for in vivo applications. We characterized the stability by incubating Gd-CA1.CD2 with 75% human serum at 37° C. for 3 and 6 hours. The protein-Gd complex remained intact after 6 hours of incubation. The result suggests that the protein is stable in blood. Taken together, the designed protein contrast agent is comparable to the clinically used contrast agents with good metal binding stability and selectivity.6,7
  • TABLE 1
    Example 1. Metal binding constants (Log Ka) and
    metal selectivity of DTPA, DTPA-BMA and CA1.CD2
    Log Log Log
    Sample Gd3+ Zn2+ Ca2+ Mg2+ (KGd/KZn) (KGd/KCa) (KGd/KMg)
    DTPA28 22.45 18.29 10.75 18.20 4.17 11.70  4.25
    DTPA-BMA41 16.85 12.04 7.17 na* 4.81 9.68 na*
    CA1.CD2 12.06 6.72 <2.22 <2.0  5.34 >9.84 >10.06
    *na: not available.
  • The designed Gd-binding proteins exhibit high r1 and r2 relaxivity. We have determined the relaxivity values of the designed protein contrast agents at 1.5, 3.0 and 9.4 Tesla field strengths (FIG. 1.2). FIG. 1.2 a shows that, at a concentration of 50 μM, the designed contrast agents Gd-CA1.CD2 and Gd-CA2.CD2 were able to introduce contrast enhancement in T1 weighted imaging at 3.0 T while 100 μM Gd-DTPA and protein CA1.CD2 alone did not lead to significant enhancement. The in vitro relaxivity values of the designed Gd-binding proteins were measured (Table 2). Gd-CA1.CD2 exhibits r1 up to 117 mM−1s−1 at 1.5T, about 20-fold higher than that of Gd-DTPA. In contrast, Gd-CA9.CD2, which carries a flexibly-conjugated Gd3+-binding site, had significantly lower relaxivity values (3.4 and 3.6 mM−1s−1, for r1 and r2 respectively, at 3.0 T), that are comparable to those of Gd-DTPA (Table 2). These data support the concept that elimination of the intrinsic mobility of the metal binding site resulted in the desired high relaxivity values.
  • TABLE 2
    Example 1. Proton relaxivity of different classes of contrast agents
    Compounds r1 r2 B0 MW
    CA class (ligand residues) (mM−1 s−1) (mM−1 s−1) (T) (kDa)
    Designed proteins CA1 117 129 1.5 12
    (E15/E56/D58/D62/D64) 48 88 3
    6 50 9.4
    CA2
    (D15/D17/N60/D62) 35 58 3 12
    CA3 130 1.5
    (E15/E56/D58/D62/E64) 34 57 3 12
    CA9
    (Add EF-loop III from 3.5 3.6 3 12
    Calmodulin)
    Small compound GdDTPA 5.4 8 1.5
    4.2 6.8 3    0.743
    *Protein carriers Albumin 11.5 12.4 0.25 80
    Poly-lysine 13 15 0.47 52
    *Dendrimers Gadomer-17 13 1.5 35
    *Liposome ACPL 12 11 1.5 >103
    *Nanoparticle Gd-perfluorocarbon 34 50 1.5 >103
    emulsion nanoparticles
    *Based on references.17,19-21
  • The r1 and r2 of Gd-CA1.CD2 exhibited an inverse relationship with the magnetic field strength (Table 2, Example 1). In contrast, the r1 and r2 of Gd-DPTA showed weak dependence on field strengths. The magnetic field strength dependent changes in relaxivity are consistent with our simulation results based on the rotational τR of the contrast agent (FIG. 1.1 b). The results showed that the protein contrast agent offers much higher relaxivities for MRI contrast enhancement at clinical magnetic field strengths (1.5-3.0 T). Interestingly, the transverse relaxivity of designed contrast agent is very high (i.g. >50 mM−1s−1) at 9.4T compared to Gd-DTPA, making it appropriate as a T2 contrast agent (Table 2) at high fields. It should be pointed out that r2 of our protein-based contrast agent is smaller than the currently used r2 agents such as iron oxides.42 This property allows our contrast agents to fill a gap between small Gd-chelators and iron oxide nanoparticles, extending the range of MRI applications both at clinically relevant field strength and possibly higher field strength.
  • One factor that contributes greatly to the relaxivity of an MRI contrast agent is its rotational correlation time, τR.11, 30, 31 Dynamic NMR studies showed that the overall correlation time of our designed protein (CA1.CD2) is similar in the absence (9.20 ns) and presence of bound metal ions (9.08 ns), consistent with that for proteins of similar size.43 The values of the order factor S2 of the ligand residues are similar to the average value of the protein, suggesting that the metal binding pocket tumbles with the protein as a whole (FIG. 1.3 a). Therefore, the measured correlation time of the protein directly reflects the τR of the metal binding site. In contrast, the flexible metal binding loop in CA9.CD2 has an S2 order value of 0.3-0.4, which is very different from CA1.CD2 with an S2 value very close to that of the backbone of the protein.
  • The hydration number of an MRI contrast agent is another determinant for r1 and r2. The hydration number of the designed protein-based contrast agents was determined by measuring the luminescence lifetime of Tb3+.26 The free Tb3+ in H2O and D2O has a life time value of 410 μs and 2,796 μs, respectively. The formation of M-protein complex significantly increases Tb3+ life time to 859 μs. The Tb3+ life time values of CA1.CD2 were 1,679 μs in D2O, suggesting a hydration number of 2.1 (FIG. 1.3 b). Interestingly, a well-known Ca2+-binding protein troponin C exhibits a hydration number of 1.8 (FIG. 1.3 b). It was determined by the X-ray crystal structure that trponin C has only one water molecule coordinated Tb3+ in the metal binding pocket.44 Therefore, it is conceivable that the hydration water molecules either from the coordination shell or the outer shell of the protein also contribute to the observed high relaxivity of CA1.CD2.
  • Application of the Designed Gd-binding Proteins for in vivo MR Imaging. The effect of MRI contrast enhancement of the protein contrast agent was tested in mice (CD-1 mice). Gd-CA1.CD2 was administered via tail vein at a dose of ˜2.4 μmole Gd/kg body weight, about 35 fold lower than the dosage of Gd-DTPA used in diagnostic imaging. Comparison of pre- and post-contrast T1 weighted spin echo images obtained at 3T showed the contrast enhancement in several organs with the greatest enhancement of the MRI contrast observed in the kidney (FIG. 1.4 a, arrows indicated), which exhibited a time dependent change of the contrast enhancement over a period of 2 hours (FIG. 1.4 b). Careful analysis of image data showed the distribution of the contrast enhancement at different organs (FIG. 1.4 b). The tissue dependent enhancement is consistent with the biodistribution of Gd3+ analyzed at 1 hour time point using inductively coupled plasma mass spectrometry (ICP-MS) (FIG. 1.4 c). The T1 contrast enhancement in the kidney cortex diminished substantially at 18 hours after the administration of Gd-CA1.CD2, suggesting that the agent was gradually cleared from the kidney and other organs. Consistent with the simulations and relaxivity values determined in vitro, a strong T2 contrast enhancement was observed at 9.4T. T2-weighted images at the 9.4T and T1-weighted images at 3T showed very similar tissue and organ distribution patterns (FIG. 1.7). At the same concentration, Gd-DTPA failed to exhibit contrast enhancements at either 3T or 9.4T.
  • Furthermore, contrast enhancement by Gd-CA1.CD2 was sustained over 4-7 hours at multiple organs (FIG. 1.8 b), indicating much longer tissue retention time of the agent than that of Gd-DTPA. The tissue retention and blood circulation time of Gd-CA1.CD2 in mice were characterized by administering various doses of agents in mice and analyzing the collected blood samples or tissue sections from sacrificed animals using immunoblots and ELISA with monoclonal (OX45) and in-house developed polyclonal (PabCD2) antibodies. In contrast to the short blood circulation time of Gd-DPTA, Gd-CA1.CD2 exhibited a prolonged blood circulation time. No significant decrease in the CA1.CD2 levels in blood was observed until 45 minutes after i.v. administration. The protein remained in blood circulation for more than 3 hours (FIG. 1.8 a). This property is considered for imaging of biological events that require prolonged imaging time, or imaging of pathological features that require time for delivery of the agent to the targeted site. In the kidney, CA1.CD2 was first detectable at 15 minutes and peaked at 4-5 hours. There was less than 10% of the injected dose of the contrast agent remaining in the kidney 15 hours after injection (by measurements of both Gd3+ and CA1.CD2). This result, along with the observation of MRI contrast changes in the bladder, suggests a clearance of the agent by kidney.
  • Gd-CA1.CD2 did not exhibit acute toxicity at the dose (˜2.4 μmole/kg) used for MRI. All mice that received the contrast agents (>10) showed no adverse effects before euthanization five days after agent injection. The effects of Gd-CA1.CD2 on liver enzymes (ALT, ALP, AST), serum urea nitrogen, bilirubin, and total protein from CD-1 mice 48 hours post-contrast injection were negligible compared to those in the control mice (Example 1, Table 3). In addition, no cytotoxicity was observed in tested cell lines, SW620, SW480 and HEK293, that were treated with 50 μM Gd-CA1.CD2, by MTT assay (FIG. 1.8 b). Based on the preliminary characterization of toxicity, we conclude that the protein contrast agent did not possess acute toxicity at current dosages for mice.
  • TABLE 3
    Example 1. Summary of Animal Clinical Pathology Profiles
    Test micea Control miceb Normal rangc
    Urea Nitrogen (mg/L) 26.0 ± 0.2d 27.0 ± 0.2  18-31
    Total Urine Protein 5.2 ± 0.2 5.3 ± 0.2  5.9-10.3
    (g/L)
    Total Bili (mg/L) 0.4 ± 0.2 0.5 ± 0.2 0.3-0.8
    Direct Bili (mg/L) 0.0   0.0
    ALT (U/L) 49.0 ± 0.2  72.0 ± 0.2  44-87
    ALP (U/L) 115.0 ± 0.2  302.0 ± 0.2  43-71
    AST (U/L) 280.0 ± 0.2  219.0 ± 0.2  101-214
    GGT (U/L) 0.0 −3.0
    aFour mice per group were injected with Gd-CA1.CD2 at dose of 4.0 μmole/kg. All clinical chemistry parameters are measured on an Olympus AU 400 analyzer by MU Research Animal Diagnostic Laboratory (details see Material and Methods).
    bControl group mice were injected with 50 μl of phosphate buffered saline pH = 7.4.
    cNormal range values are from Quesenberry, K. E. and J. W. Carpenter; Ferrets, Rabbits, and Rodents Clinical Medicine and Surgery; W. B. Saunders: Philadelphia, 2003.
    dThe standard deviations of measurements with four animal (n = 4).
  • Discussion
  • While new developments of Gd3+ chelators4, 5, 11, 21, 46 continue to expand the applications of small molecular contrast agents, macro-molecular agents are increasingly attractive for functional and molecular imaging applications. A common approach of using small molecular GdDTPA to bind albumin in serum (e.g., MS-325) has the capability to enhance the relaxivity in vivo. However, this class of contrast agents is currently limited to imaging the vascular system47-50 with the complex pharmacokinetics.51 Conjugation or encapsulation of small Gd-chelators to or in the liposome, fullerene, and nanotube, indeed resulted in increases in relaxivity; however, several important drawbacks limit the applications of these agents. Our approach of using an engineered protein to chelate the Gd3+ for contrast enhancing effect differs fundamentally from those previous studies in several respects.
  • First, we have created a Gd3+-binding site with strong metal selectivity in a stable and potentially fully functioning host protein by de novo design. This is significantly different from using a small peptide fragment to cross-link small Gd3+-chelate in stability and rigidity of the binding site as well as biological functions of the protein. To our knowledge, this is the first example that an MRI protein contrast agent was made using an engineered Gd3+ binding protein without using existing small metal chelators. This is an achievement in protein design, involving the rational development of a metalloprotein with high coordination number and charged ligand residues in the coordination shell.
  • Second, our approach provides a new platform for developing MRI contrast agents with further improved relaxivity and metal selectivity and stability by protein engineering. Our studies reveal that three factors are key in achieving high relaxivity: 1) the longer rotational correlation time τR of the designed agents, 2) the direct coordination of Gd3+ ions to amino acid ligands at the rigid protein matrix to eliminate internal mobility, and 3) the increased hydration of water molecules. As predicted by the Solomon-Bloembergen-Morgan equation (Supplementary Equation 1), relaxivities can be significantly increased by increasing the number of hydration water molecules. Unfortunately, previous attempts to increase relaxivities by increasing the number of coordinating water molecules from 1 to 2 for BTPA and DTPA-BMA did not yield successful results. Our studies demonstrated an excellent example that it is possible to increase in relaxivity by increasing the hydration number of protein MRI contrast agent without sacrifice the metal binding properties, such as affinity and metal specificity. Presumably, the concept demonstrated in this study may be applied to the design of other macromolecule based MRI contrast agents. Using protein to chelate Gd3+ as an MRI contrast agent has several potential advantages over currently used Gd-DTPA in functional and molecular imaging applications: 1) it greatly increases the contrast-to-noise ratio (CNR); 2) it improves dose efficiency with reduced metal toxicity; 3) it prolongs the tissue retention time, which enables imaging of abnormalities that requires prolonged tissue enhancement; and 4) provide a potential functioning protein or a protein carrier that can conjugate target specific ligands to a biomarker for targeted molecular MR imaging.
  • ESI-Mass spectrometry of metal:protein complex, determination of metal binding constants, MRI imaging and its data analysis, tissue retention and blood circulation time of contrast agents, toxicity, and simulation of contrast agent relaxivities.
  • Computational Simulation of Contrast Agent Relaxivities52
  • The simulation of R1 and R2 as a function of magnetic field strength is performed based on supplementary equations 1 and 2:
  • R 1 = cq 55.5 1 T 1 m + τ m ( 1 ) R 2 = cq 55.5 T 2 m - 2 + τ m - 1 T 2 m - 1 + Δ ω m 2 τ m ( ( τ m - 1 + T 2 m - 1 ) 2 + Δ ω m 2 ) ( 2 )
  • In these equations, the water coordination number, q, is assumed to be 1 and the agent concentration is 0.001 M; τm is the dwelling time of the coordination water; and Δωm is the chemical shift difference between the bound and free water. Since Δωm 2 is much smaller than other components, equation 2 is simplified to supplementary equation 3 and used in this simulation:
  • R 2 = cq 55.5 1 T 2 m + τ m ( 3 )
  • Tim is determined by dipole-dipole (DD) and scalar or contact (SC) mechanisms as shown in supplementary equation 4:
  • 1 T im = 1 T i DD + 1 T i SC i = 1 , 2 ( 4 )
  • Because the contribution from Ti DD is much greater than that of Ti SC, only the former is used in the simulation, which is obtained using supplementary equations 5 and 6:
  • 1 T 1 DD = 2 15 γ I 2 g 2 μ B 2 r GdH 6 S ( S + 1 ) ( μ 0 4 π ) 2 [ 7 τ c 2 1 + ω s 2 τ c 2 2 + 3 τ c 1 1 + ω I 2 τ c 1 2 ] ( 5 ) 1 T 2 DD = 1 15 γ I 2 g 2 μ B 2 r GdH 6 S ( S + 1 ) ( μ 0 4 π ) 2 [ 13 τ c 2 1 + ω s 2 τ c 2 2 + 3 τ c 1 1 + ω I 2 τ c 1 2 + 4 τ c 1 ] ( 6 )
  • The following values are used in the calculation: the gyro-magnetic constant for proton γ1, 2.675×108T−1s−1; g, 2.0; Bohr magneton μB, 9.274×10−24 J T−1; S, 7/2; permeability of vacuum μ0, 1.257×10−6N A−2; and the distance between the Gd3+ and proton rGdH, 3.0×10−10 m (normally 2.7-3.3 Å). The frequency of proton ω1 equals to the γ1 multiplied by the magnetic field while the frequency of electron ωs is 658-fold of ω1. The τci is determined by the rotational correlation time τR, the water dwelling time τm, and Tie as shown in supplementary equation 7:
  • 1 τ ci = 1 τ R + 1 τ m + 1 T ie i = 1 , 2 ( 7 )
  • Tie is related to the electron frequency ωs as well as the τv (correlation time of splitting) and Δ2 (mean square zero field splitting energy) of the Gd3+ as in supplementary equations 8-10:
  • 1 T 1 e = 2 C ( 1 1 + ω s 2 τ v 2 + 4 1 + 4 ω s 2 τ v 2 ) ( 8 ) 1 T 2 e = C ( 5 1 + ω s 2 τ v 2 + 2 1 + 4 ω s 2 τ v 2 + 3 ) ( 9 )
  • Where
  • C = 1 50 Δ 2 τ v { 4 S ( S + 1 ) - 3 } ( 10 )
  • Various combinations of τR (1 ps, 10 ps, 100 ps, 1 ns, 10 ns, and 100 ns), τm (1 ps, 10 ps, 100 ps, 1 ns, 10 ns, and 100 ns), τv (1 and 10 ps), and Δ2 (1017, 1018, 1019, and 1020 s−2) have been proposed for the calculation of magnetic field-dependent relaxivities under magnetic field strengths ranging from 0.001 MHz to more than 1000 MHz. For small molecules such as DPTA with τR at hundreds of ps level, the relaxivity is <10 mM−1s−1 no matter how the other parameters are adjusted. On the other hand, for the contrast agents with τR at 10 ns level, such as the CD2 derivatives in our study, the relaxivity can reach a much higher level by adjusting other parameters such as the τm.
  • Toxicity of Contrast Agent Gd-CA1.CD2
  • The protein contrast agent Gd-CA1.CD2 did not exhibit acute toxicity at the MRI imaging dose (˜2.4 mole/kg), as demonstrated by the fact that all MR imaged mice that received the contrast agent (>10) behaved normally and remained healthy (sacrificed five days after agent injection). The effects of Gd-CA1.CD2 on liver enzymes (ALT, ALP, AST), serum urea nitrogen, bilirubin, and total protein from CD-1 mice 48 hours post-contrast injection were negligible compared to the control mice (Table 3). In addition, no cytotoxicity was observed in tested cell lines, SW620, SW480 and HEK293 that were treated with 50 μM Gd-CA1.CD2, by MTT assay (FIG. 1.7 b). Based on the preliminary characterization of toxicity, we conclude that the protein contrast agent is relatively safe. References for Example 1, each of which are incorporated herein by reference:
    • 1. Tyszka, J. M.; Fraser, S. E.; Jacobs, R. E., Curr Opin Biotechnol 2005, 16, (1), 93-99.
    • 2. Lippard, S. J., Nat Chem Biol 2006, 2, (10), 504-507.
    • 3. Louie, A. Y.; Huber, M. M.; Ahrens, E. T.; Rothbacher, U.; Moats, R.; Jacobs, R. E.; Fraser, S. E.; Meade, T. J., Nat Biotechnol 2000, 18, (3), 321-325.
    • 4. Frangioni, J. V., Nat Biotechnol 2006, 24, (8), 909-913.
    • 5. Woods, M.; Woessner, D. E.; Sherry, A. D., Chem Soc Rev 2006, 35, (6), 500-511.
    • 6. Lauffer, R. B., Chem. Rev. 1987, 87, 901-927.
    • 7. Aime, S.; Barge, A.; Cabella, C.; Crich, S. G.; Gianolio, E., Curr Pharm Biotechnol 2004, 5, (6), 509-518.
    • 8. Toth, E.; Helm, L.; Merbach, A. E., Contrast Agents I. Magnetic Resonance Imaging W. Krause, Ed. 2002, 221, 61-102.
    • 9. Merbach, A. E.; Toth, E., The chemistry of contrast agent agents in medical magnetic resonance Imaging. 2001.
    • 10. Geraldes, C. F.; Sherry, A. D.; Cacheris, W. P.; Kuan, K. T.; Brown, R. D., 3rd; Koenig, S. H.; Spiller, M., Magn Reson Med 1988, 8, (2), 191-199.
    • 11. Caravan, P., Chem Soc Rev 2006, 35, (6), 512-523.
    • 12. Weinmann, H. J.; Press, W. R.; Gries, H., Invest Radiol 1990, 25 Suppl 1, S49-50.
    • 13. Opsahl, L. R.; Uzgiris, E. E.; Vera, D. R., Acad Radiol 1995, 2, (9), 762-767.
    • 14. Langereis, S.; de Lussanet, Q. G.; van Genderen, M. H.; Meijer, E. W.; Beets-Tan, R. G.; Griffioen, A. W.; van Engelshoven, J. M.; Backes, W. H., NMR Biomed 2006, 19, (1), 133-141.
    • 15. Bryant, L. H., Jr.; Brechbiel, M. W.; Wu, C.; Bulte, J. W.; Herynek, V.; Frank, J. A., J Magn Reson Imaging 1999, 9, (2), 348-352.
    • 16. Sirlin, C. B.; Vera, D. R.; Corbeil, J. A.; Caballero, M. B.; Buxton, R. B.; Mattrey, R. F., Acad Radiol 2004, 11, (12), 1361-1369.
    • 17. Lanza, G. M.; Winter, P.; Caruthers, S.; Schmeider, A.; Crowder, K.; Morawski, A.; Zhang, H.; Scott, M. J.; Wickline, S. A., Curr Pharm Biotechnol 2004, 5, (6), 495-507.
    • 18. Karfeld, L. S.; Bull, S. R.; Davis, N. E.; Meade, T. J.; Barron, A. E., Bioconjug Chem 2007, 18, (6), 1697-1700.
    • 19. Gillies, R. J., J Cell Biochem Suppl 2002, 39, 231-238.
    • 20. Artemov, D.; Bhujwalla, Z. M.; Bulte, J. W., Curr Pharm Biotechnol 2004, 5, (6), 485-494.
    • 21. Aime, S.; Cabella, C.; Colombatto, S.; Geninatti Crich, S.; Gianolio, E.; Maggioni, F., J Magn Reson Imaging 2002, 16, (4), 394-406.
    • 22. Anderson, E. A.; Isaacman, S.; Peabody, D. S.; Wang, E. Y.; Canary, J. W.; Kirshenbaum, K., Nano Lett 2006, 6, (6), 1160-1164.
    • 23. Strijkers, G. J.; Mulder, W. J.; van Heeswijk, R. B.; Frederik, P. M.; Bomans, P.; Magusin, P. C.; Nicolay, K., MAGMA 2005, 18, (4), 186-192.
    • 24. Caravan, P.; Greenwood, J. M.; Welch, J. T.; Franklin, S. J., Chem Commun (Camb) 2003, (20), 2574-2575.
    • 25. Kim, Y.; Welch, J. T.; Lindstrom, K. M.; Franklin, S. J., J Biol Inorg Chem 2001, 6, (2), 173-181.
    • 26. Sudnick, D. R.; Horrocks, W. D., Jr., Biochim Biophys Acta 1979, 578, (1), 135-144.
    • 27. Beeby, A.; Clarkson, I. M.; Dickins, R. S.; Faulkner, S.; Parker, D.; Royle, L.; de Sousa, A. S.; Gareth Williams, J. A.; Woods, M., J. Chem. Soc., Perkin Trans. 1999, 2, 493-504.
    • 28. Martell, A. E.; Simth, R. M.; Motekaitis, R. J., NIST Standard Reference Data, Gaithersburg, Md. 1993.
    • 29. Wilkins, A. L.; Yang, W.; Yang, J. J., Curr Protein Pept Sci 2003, 4, (5), 367-373.
    • 30. Yang, W.; Wilkins, A. L.; Li, S.; Ye, Y.; Yang, J. J., Biochemistry 2005, 44, (23), 8267-8273.
    • 31. Yang, W.; Wilkins, A. L.; Ye, Y.; Liu, Z. R.; Li, S. Y.; Urbauer, J. L.; Hellinga, H. W.; Kearney, A.; van der Merwe, P. A.; Yang, J. J., J Am Chem Soc 2005, 127, (7), 2085-2093.
    • 32. Deng, H.; Chen, G.; Yang, W.; Yang, J. J., Proteins 2006, 64, (1), 34-42.
    • 33. Yang, W.; Lee, H. W.; Hellinga, H.; Yang, J. J., Proteins 2002, 47, (3), 344-356.
    • 34. Pidcock, E.; Moore, G. R., J Biol Inorg Chem 2001, 6, (5-6), 479-489.
    • 35. Yang, W.; Jones, L. M.; Isley, L.; Ye, Y.; Lee, H. W.; Wilkins, A.; Liu, Z. R.; Hellinga, H. W.; Malchow, R.; Ghazi, M.; Yang, J. J., J Am Chem Soc 2003, 125, (20), 6165-6171.
    • 36. Maniccia, A. W.; Yang, W.; Li, S. Y.; Johnson, J. A.; Yang, J. J., Biochemistry 2006, 45, (18), 5848-5856.
    • 37. Ye, Y.; Lee, H. W.; Yang, W.; Shealy, S. J.; Wilkins, A. L.; Liu, Z. R.; Torshin, I.; Harrison, R.; Wohlhueter, R.; Yang, J. J., Protein Eng 2001, 14, (12), 1001-1013.
    • 38. Ye, Y.; Lee, H. W.; Yang, W.; Shealy, S.; Yang, J. J., J Am Chem Soc 2005, 127, (11), 3743-3750.
    • 39. Wedeking, P.; Shukla, R.; Kouch, Y. T.; Nunn, A. D.; Tweedle, M. F., Magn Reson Imaging 1999, 17, (4), 569-575.
    • 40. Kumar K.; Tweedle, M. F.; Malley, M. F.; Cougoutas, J. Z., Inorg. Chem. 1995, 34, 6472-6480.
    • 41. Cacheris, W. P.; Quay, S.C.; Rocklage, S. M., Magn Reson Imaging 1990, 8, (4), 467-481.
    • 42. Bulte, J. W.; Kraitchman, D. L., NMR Biomed 2004, 17, (7), 484-499.
    • 43. Wyss, D. F.; Dayie, K. T.; Wagner, G., Protein Sci 1997, 6, (3), 534-542.
    • 44. Rao, S. T.; Satyshur, K. A.; Greaser, M. L.; Sundaralingam, M., Acta Crystallogr D Biol Crystallogr 1996, 52, (Pt 5), 916-922.
    • 45. Barnhart, J. L.; Kuhnert, N.; Bakan, D. A.; Berk, R. N., Magn Reson Imaging 1987, 5, (3), 221-231.
    • 46. van Zijl, P. C.; Jones, C. K.; Ren, J.; Malloy, C. R.; Sherry, A. D., Proc Natl Acad Sci USA 2007, 104, (11), 4359-4364.
    • 47. Lauffer, R. B.; Parmelee, D. J.; Ouellet, H. S.; Dolan, R. P.; Sajiki, H.; Scott, D. M.; Bernard, P. J.; Buchanan, E. M.; Ong, K. Y.; Tyeklar, Z.; Midelfort, K. S.; McMurry, T. J.; Walovitch, R. C., Acad Radiol 1996, 3 Suppl 2, S356-358.
    • 48. Lauffer, R. B.; Parmelee, D. J.; Dunham, S. U.; Ouellet, H. S.; Dolan, R. P.; Witte, S.; McMurry, T. J.; Walovitch, R. C., Radiology 1998, 207, (2), 529-538.
    • 49. Parmelee, D. J.; Walovitch, R. C.; Ouellet, H. S.; Lauffer, R. B., Invest Radiol 1997, 32, (12), 741-747.
    • 50. Allen, M. J.; Meade, T. J., J Biol Inorg Chem 2003, 8, (7), 746-750.
    • 51. Brasch, R.; Turetschek, K., Eur J Radiol 2000, 34, (3), 148-155.
    • 52. Toth, E.; Helm, L.; Merbach, A. E. Contrast Agents I: Magnetic Resonance Imaging W. Krause, Ed. 2002, 221, 61-102.
    Example 2 PEGylation Modification of Protein MRI Contrast Agents with Enhanced Relaxivity and Reduced Immunogenicity Introduction:
  • In this Example, we report our progress in optimizing designed protein contrast agents for in vivo imaging by pegylation. Our experimental results clearly demonstrate that PEGylation substantially increases the solubility of protein by more than 100 fold without reducing metal binding affinity and selectivity. In addition, the serum stability is significantly improved. Interestingly, PEGylation further increased the in vitro R1 and R2 relaxivities of the developed protein contrast agents by 2-3 fold, which is in contrast to the loss of functionality of protein drug by pegylation. Such increased relaxivity is a result of the addition of a hydration layer due to water retention by Poly-PEG chain on the protein surface and alteration of correlation time. The agent demonstrated a strong contrast enhancement in the animal imaging with a dose 70 fold lower than that of Gd-DTPA. Our developed contrast agent showed a much longer blood circulation time, and the blood circulation time and its distribution of our protein contrast agent in mice can be modified by using different lengths of PEG. Furthermore, the immunogencity of the contrast agent has been significantly decreased by pegylation. The optimized properties of protein contrast agents by pegylation facilitate the disease targeted tissue and molecular imaging.
  • PEGylation of protein is a method used to improve the pharmacokinetics and pharmaco-dynamics of various protein and peptide drugs. PEGylation involves modifications of Lys, Glu, Asp, or Cys residues of a protein or peptide with various sizes of polyethylene glycerol chain. The result of PEGylation modifications is the attachment of the different size of polyethylene glycerol chains on the surface of the modified protein or peptide. The protein or peptide experiences several property changes, especially in pharmacokinetics and pharmaco-dynamics. Two changes are obvious: (1) the increase in molecular size, especially in the case of small peptide, and (2) the reduction in surface charges of protein and peptide. A consequence of these two changes is the increase in blood circulation time and delay in the renal secretion. Another effect of the changes is the reduced immunogenicity of the protein or peptide drugs after PEGylation. The strategy has been successfully employed in a number of protein or peptide drugs for increased efficacy and/or reduced immuno-response of the drug. Since the polyethylene glycerol chains are strongly hydrophilic, PEGylation of protein will also result in a dramatic increase in solubility of protein or peptide drugs. However, the most significant drawback of PEGylation for a protein or peptide drug is that the bulk volume of the polyethylene glycerol chains on the surface of proteins often blocks the bio-active site(s) leading to significant decrease in bio-activity.
  • We have previously reported the development of protein-based MRI contrast agents by rational design of Gd3+ binding sites into a stable protein using amino acids residues as metal coordinating ligands. The designed protein contrast agent exhibits a 10-20-fold increase in in vitro R1 and R2 relaxivities compared to the current clinically used contrast agent Gd-DTPA. To apply this class of novel class of protein contrast agents to in vivo imaging, several additional factors must be considered. First, contrast agents need to have high solubility. Because of limitations in the sensitivity of MRI techniques and injection volumes for animals, in order to provide significant in vivo tissue contrast, requires up to a 300-500 mM injection dose for a clinically used DTPA with a relaxivity of 5 mM−1s−1. Compared to DTPA, our protein contrast agents with a 10-20 fold increase of relaxivity require a soluble concentration of 30-50 mM. Second, proper blood circulation time is required to facilitate targeted tissue or molecular MR imaging. Third, the in vivo stability of the contrast agents against degradation and kinetic stability against metal transformation is essential to reduce toxicity. Fifth, the immunogenicity of the protein contrast agents needs to be reduced.
  • In this Example, we report our progress in optimizing designed protein contrast agents for in vivo imaging by pegylation. Our experimental results clearly demonstrate that PEGylation substantially increases the solubility of protein by more than 100 fold without reducing metal binding affinity and selectivity. In addition, the serum stability is significantly improved. Interestingly, PEGylation further increased the in vitro R1 and R2 relaxivities of the developed protein contrast agents by 2-3 fold, which is contrast to the loss of functionality of protein drug by pegylation. The agent demonstrated a strong contrast enhancement in the animal imaging with a dose 70 fold lower than that of Gd-DTPA. Our developed contrast agent showed a much longer blood circulation time, and the blood circulation time and its distribution of our protein contrast agent in mice can be modified by using different lengths of PEG. Furthermore, the immunogencity of the contrast agent has been significantly decreased by pegylation. The optimized properties of protein contrast agents by pegylation facilitate the disease targeted tissue and molecular imaging.
  • 2. Material and Methods
  • All pegylation reagents were purchased. Metal and dye reagents were purchased from Molecular Probes and Sigma. Cys was added at the C-terminal of the CA1.CD2 and subcloned in pet20b for protein expression. Site-directed mutagenesis method was used to remove Lys at different locations. DNA sequences were verified by DNA sequence core facility.
  • 2.1 Protein Expression and Purification
  • Protein was expressed in E. coli as inclusion body and purified using urea refolding and ion-exchange column. N15 labeled protein was expressed in SV medium as GST fusion (Yang et al., Biochem 2006) and purification was by GST-4B affinity column and SP column. Protein was verifed by mass spectrometry. Protein concentration was calculated using extinction coefficient of w.t. CD2 of 11,000 (Ye et al., 2001). Protein solubility was determined by concentrating proteins to reach to precipitation using speed vac.
  • 2.2 Pegylation
  • We first carried out the PEGylation modifications of Lys residues on our designed protein MRI contrast agents using different preactivated NHS with PEG units of 4, 12, 40, 12K, and 20 K (FIGS. 2.2-2.4). Typically reactions were carried out using 3:1 or 5:1 PEG:protein at room temperature for 1-2 hours. Reactions at pH 6, 7 and 9 were also performed with no major change in the pegylation results. N-terminal pegylation of Lys was performed at pH 6 according to published papers (1-5).All reactions were quenched by adding free amino acids and stored at −20° C. for further purification. Specifically modification at the Cys at the C-terminal was performed by using published methods (1-5).
  • Separation of pegylated proteins were achieved by 10 fold dilution of reaction mix and loaded on sp column using a pH gradient from 2 to 7. Free PEG was not able to bind to the column and washed away before pH gradient. Free unpegylated protein fractions were eluted out at the latest fraction at high pH. Separated protein samples were then verified by mass spectrometry analysis, metal binding and NMR.
  • 2.3 Mass Spectrometry
  • ESI and MALDI spectrometry were used to identify the number of pegylation sites and metal binding stoichiometry. The pegylation sites were identified using trypsin cleavage followed by Mass analysis using TOF/TOF.
  • 2.4 Gd3+-Binding Affinity Determination.
  • Gd3+-binding affinities of CA1.CD2 and its pegylated variants were determined by a competition titration with Fluo-5N applied as a Gd3+ indicator (Yang et al., JACS, 2008). The fluorescence spectra of Fluo-5N were obtained with a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Fluo-5N emission spectra were acquired at 500 nm to 650 nm with an excitation at 488 nm. Gd3+-binding affinity of Fluo-5N, Kd1, was first determined by a Gd3+ titration with Gd3+ buffer system of 1 mM nitrilotriacetic acid (NTA). Free Gd3+ concentration was calculated with a NTA Gd3+-binding affinity of 2.6×10−12 M.28 Fluo-5N was mixed with Gd3+ in 1:1 ratio for a competition titration. The experiment was performed with a gradual addition of CA1.CD2 or its variants. An apparent constant, Kapp, was estimated by fitting the fluorescence emission intensity of Fluo-5N at 520 nm with different CA1.CD2 concentrations as a 1 to 1 binding model. Gd3+-binding affinity of CA1.CD2, Kd2, was calculated with the following equation:
  • K d 2 = K app K d 1 K d 1 + [ Fluo - 5 N ] T ( 1 )
  • Metal binding affinity for Ca2+ and Zn2+ were determined using similar competition methods and specific dyes with proper Kd values.
  • 2.5 Measurement of Water Coordination Number by Terbium Life Time Luminescence.
  • The numbers of water ligands coordinated to Gd3+-CA1.CD2 and variants complex were determined by measuring Tb3+ luminescence decay in H2O or D2O (Yang et al., 2008). Tb3+ excited state lifetime was measured using a fluorescence spectrophotometer (Photon Technology International, Inc.) with a 10 mm path length quartz cell at 22° C. Following excitation at 265 nm with a XenoFlash (Photon Technology International, Inc.), Tb3+ emission was monitored at 545 nm in a time series experiment in both H2O and D2O systems. Luminescence decay lifetime was obtained by fitting the acquired data with a mono-exponential decay function. H2O in CA1.CD2 solution was replaced with D2O by lypholization and re-dissolved in D2O at least three times. A standard curve correlating the Δkobs with water number under our experimental conditions was established by using well-characterized chelators, such as EDTA (q=3), DTPA (q=1), NTA (q=5), and Aquo Tb3+ (q=9) solution with R2=0.997.26,27 Water number coordinated to Tb3+-CA1.CD2 complex was then obtained by fitting the acquired Δkobs value to the standard curve.
  • 2.6 In Vitro MRI Relaxivity
  • Relaxivity was determined using 0.47 T (20 Hz) minispec Magnetic Relaxometer (Bruker) and 300 and 500 MHz NMR (Varian).
  • 2.7 NMR
  • Pulse field diffusion NMR was applied to measure the hydrodynamic radii of the protein (Lee et al., BBA 2003) with Protein samples CA1.CD2 and its variants of 0.2 mM in buffer. Lysozme and diaxone were used as an external and internal reference for calibration. The correlation size of the protein was measured using TauC pulse sequence developed by the Prestegard lab at UGA using 15N labeled protein.
  • 2.8 In Vivo Mouse MR Imaging.
  • Care of experimental animals was in accordance with institutional guidelines. CD-1 mice (25-30 g, four mice were imaged) were anesthetized with an isoflurane gas mixture. The anesthetized animal was positioned and stabilized with soft-supporting material (e.g. foam) in the scanner in the coil cradle and was kept warm during the MRI scan. The mice were scanned prior to the administration of any contrast agent (pre-contrast). Approximately 50 μl of Gd-CA1.CD2 (˜1.2 mM) or Gd-DTPA (˜300 mM) were injected into the animal via the tail vein. MR images were collected at different times (indicated). For T1 weighted imaging at 3T, spin echo sequence with TE/TR=15 ms/500 ms was employed. Rectangular Field of View (FOV) at 100/40 mm, an acquisition matrix of 1962 and 1.1 mm slice thickness without gap were used. Images were collected from both transverse and coronal sections. The in-plane resolution of images was less than 0.5 mm after they were reconstructed to the matrix of 1962. For T2 weighted imaging at 9.4T, MR images were recorded using a multi-echo Carr-Purcell-Meiboom-Gill (CPMG) sequence. The data were collected and processed by Dicomworks software. The MR signal intensity in several organs was ascertained by the average intensity in ROIs or points within the organs. Signal intensity for each organ was normalized to that of the leg muscle.
  • 2.9 Blood Circulation Time, Tissue Retention Time, and Bio-Distribution.
  • CD-1 mice (25-30 g) were anesthetized with isoflurane. Appropriate dosages of Gd-CA1.CD2 and its PEgylated variants (with Gd and 153Gd) or Gd-DTPA were i.v. injected (via tail vein). Blood (˜50 μl) samples were collected via orbital sinus of the mouse at different time points. The mouse was euthanized at the final time point. Tissue samples from kidney, liver, heart, and lung were collected. Serum samples were prepared from the collected blood. For the bio-distribution analyses, the animals were euthanized at single time point (indicated) after i.v. administration of the contrast agent (indicated). The organ/tissue samples were collected. Tissue extracts were freshly made from collected samples using commercially available tissue extracting kits (Qiagen). CA1.CD2 and its variants were detected and quantified by immunoblotting and Sandwich-ELISA using a monoclonal antibody (OX45, detecting antibody) and a home made polyclonal antibody (PabCD2, capture antibody). A series of known amounts of CA1.CD2 samples mixed with blank mouse serum or tissue extracts were used as standard in Sandwich-ELISA. ELISA signal from HRP was monitored using a Fluorstar fluorescence microplate reader. For quantification of Gd3+ in the serum and tissue samples one hour (or indicated times) after the contrast agent administration, animals were sacrificed and critical organs were collected, and the tissues were then digested with concentrated nitric acid at 120-130° C. with proper amount of 153Gd spike as an internal marker. The digested solution was analyzed.
  • 2.10 Immunogenicity
  • Rabbits were i.p. injected with Gd-CA1.CD2 and its pegylated variants. The agents (CA1.CD2) were mixed with adjuvant or with buffer saline and injected at a dose of 3.0 nmole/kg according to the standard protocol for the antibody production. The rabbits were subjected to double immunolizations in four weeks interval. Blood samples were taken from the immunolized rabbits 3 weeks after each injection. Production of antibodies against the protein contrast agent in each rabbit was examined by ELISA
  • 2.11 Toxicity Analyses.
  • The MR imaged CD-1 mice that received the i.v. administered Gd-CA1.CD2 and its variants (at a dose of ˜2.4 μmol/kg) spiked with 157Gd3+ were returned to their cages (one mouse per cage). The mice were observed for five days and were euthanized at the end of the fifth day. Tissue samples from kidney, liver, spleen, and lung were collected. Gd3+ contents in the tissue samples were analyzed by radioactivity counter.
  • Two groups of mice were used to examine potential renal and/or liver damage by Gd-CA1.CD2. One group of mice received (i.v. tail veil) 50 μl of saline-buffer (as control). Another group received (i.v. tail veil) Gd-CA1.CD2 and variants at 4 μmole/kg. The mice were observed for 48 hours and were euthanized. Blood samples were collected from the experimental mice. Serum samples were prepared from the collected blood. Liver enzymes in serum samples, including Alanine transaminase (ALT), Alkaline phosphatase (ALP), Aspartate transaminase (AST), Gamma glutamyl transpeptidase (GGT), and bilirubin and urea nitrogen were analyzed by a commercially available source (MU Research Animal Diagnostic Laboratory). All clinical chemistry parameters were measured on an Olympus AU 400 analyzer.
  • Cytotoxicity was analyzed by MTT assay of the cells that were treated with Gd-CA1.CD2 and its variants at appropriate doses (indicated in figure). The cells were grown under normal growth medium in 96 well plates. Gd-CA1.CD2 or saline-phosphate buffer was added to the cell culture medium. The cells were incubated for appropriate times. A standard MTT assay was employed to assess the cell growth status of the treated cells.
  • 2.12 Serum Stability.
  • CA1.CD2 (40 μM) in complex with Gd3+ was incubated with 75% human serum over 3 or 6 hours at 37° C. The degradation of the protein (disappearance of 12 kDa protein band) was analyzed by SDS-PAGE and visualized by coomassie blue staining for protein and idiol staining for PEG moiety. In parallel, the degradation of the protein was also analyzed by immunoblot using antibodies OX54 or PabCD2. The identities of CA1.CD2 and pegylated variants were verified by immunobloting using antibody PabCD2 and idiol staining.
  • 3. Results 3.1 PEGylation Modifications of Designed Protein Contrast Agent CA1.CD2
  • We previously reported the development of a novel class of protein-based MRI contrast agents. The contrast agents exhibit a 20 fold increase in R1 and R2 relaxivities and provide a strong contrast enhancement in the mouse MR imaging (Yang, et. al. JACS 2008). The developed MRI contrast agent also demonstrated a prolonged blood circulation time, which is considered for the application of the agents in disease targeted molecular imaging. In the processes of preparing the protein contrast agent for animal MR imaging, we realized that it is desirable to have a higher protein concentration up to 30-50 mM based on the in vivo application dose of DTPA (300-500 mM) per 100 ul injection. However, the solubility of the current formulation of our protein contrast CA1.CD2 is ˜0.7 mM.
  • As shown in FIG. 2.1, the designed contrast agent CA1.CD2 has eight Lys residues with different solvent accessibility and six Lys residues are well exposed. Based on the considerations of solubility, circulation, relaxivity and serum stability, we therefore carried out PEGylation of our protein contrast agents by modifying surface Lys residues and site specific pegylation. First, preactivated PEG units with varied chain length and branches of 4, 12 (Poly-PEG12), 40 (PolyPEG40k), 5K, and 12 K (see FIG. 2.1 b) were used to modify surface lys residues. Site-specific pegylation was performed at both N-terminal Lys and C-terminal Cys.
  • To identify optimal conditions for pegylation, PeG reactions were performed at different reaction time, pH 6, 7 and 9, and reaction ratio. FIGS. 2.2-2.4 shows the SDS gel of the protein contrast agent CA1.CD2 pegylated with PEGylation kits with different reaction moieties stained by both commassie blue for protein and idiol for the PEG moiety.
  • FIG. 2.2 shows that the PEGylated proteins were separated using ion exchange column, size exchange column, and a C18 reverse phase HPLC chromatography column. The modified proteins were purified by HPLC to relative homogeneity. FIG. 2.2 shows that there were several major peaks of the PEGylated proteins on the FPLC chromatography. MALD-Mass analyses of the separated protein fraction reveal that at pH 7.4 with 5:1 pEG:protein:preactivated pEG reagents, CA1.CD2 was pegylated mainly with 2, 3 with P4-P40 and 1 PEG unit with PEG 12 K to 20K.
  • 3.2 Determination of PeGylation Numbers
  • With the addition of N-terminal NH2 groups, the CA1.CD2 has total of seven potential PEGylation sites. The PEGylation sites of the purified protein were examined first by trypsin cleavage to generate the peptides/fragments and then sequenced by TOF/TOF MS. Usually 2-4 PEG units were attached to each protein depending on the ratio of PEG:protein under reaction and the reaction conditions.
  • 3.3 Conformation and Metal Binding Capabilities
  • FIG. 2.4 a of Trp fluorescence spectra shows that these PEGylated proteins (CA1.CD2-PEG12 and CA1.CD2-PEG40) maintain the native structure of the protein contrast agents with unchanged Trp emission maximum compared with CA1.CD2.
  • All of the designed Gd3+ binding proteins were expressed in E. coli and subsequently purified by procedures previously published from our laboratory.30,31 All of the designed proteins form the expected metal:protein complex as demonstrated by ESI-Mass spectrometry.
  • Tb-FRET was first used to monitor the effect of pegylation on the metal binding capabilities. As shown in FIG. 2.4 b, the pegylated A1.CD2 is able to have Tb-sensitized energy transfer similar to that of CA1.CD2. FIG. 2.5 shows that PEGylated CA1.CD2-P40 remains intact after incubate with human serum for 24 hours at 37° C. monitored by SDS Page.
  • Gd3+-binding affinity of CA1.CD2 was further determined by a competition titration with fluorescent dye applied as a Gd3+ indicator in various chelate-metal buffer systems. The PEGylated variants exhibit similar metal binding affinities to unpegylated ones for Ca2+, Gd3+ and Zn2+.
  • 3.4 The Effect of PEGylation on the Relaxivities of the Protein Contrast Agent
  • We first measured the in vitro relaxivities of the PEGylated protein contrast agent in MRI scanner such as at 0.47, 3.0, and 9.4T field strengths (FIG. 2.6). Very similar procedure used in our previous studies in measuring the relaxivities of protein MRI contrast agent {Yang, et. al., JACS 2008} was employed here to measure the relaxivities of PEGylated CA1.CD2. It was clear that the relaxivities (both R1 and R2) were increased compared to those of the same protein contrast agent before PEGylation modification. In addition, the MRI relaxivity increases with the increase of PEG length as shown in FIG. 2.6.
  • Increasing the degree of PEGylation modification increased the R1 and R2 relaxivities (FIG. 2.6). The R1 and R2 relaxivities of the PEGylated protein were also affected by the size of the PEG chains. The relaxivities of the PEGylated protein contrast agents demonstrated higher increases when the protein was modified by a longer PEG chain. Interestingly, both R1 and R2 relaxivities of the PEGylated protein contrast agent experienced the most dramatic increases at higher magnetic field. This is contrary to the case of unPEGylated protein, in which dramatic decreases in R1 and R2 relaxivities were observed at high field (Yang et al., JACS, 2008). FIG. 2.6 shows the MRI relaxivity as a function of chain dependencies. Specific modification of the N-terminal amine and Cys residues at the C-terminus have a similar effect on the relaxivity of the protein.
  • Table 2.1 of Example 2 shows that the water numbers in the coordination shell of CA1.CD2 increased from 2 to 3 upon PEGylation with PEG40. This may contribute to the increased relaxivity by PEGylation.
  • 3.6 PEGylation Improved Blood Circulation Time
  • PEGylation has been demonstrated to increase blood circulation time and reduce immunogenicity of a number of protein drugs. While longer blood circulation time is a desired property for the applications of MRI contrast agents. Elimination or reduction of immunogenicity is essential for clinical applications of the protein MRI contrast agents. We therefore examined whether PEGylation of CA1.CD2 changed the bio-distribution and immunogenicity properties of the agent. The PEGylated or unPEGylated CA1.CD2 was introduced to CD-1 mice via i.v. tail vein injection. Distributions of the administrated agent among different organs/tissues and blood circulation were analyzed at different time intervals by quantization of both the protein CA1.CD2 using immunochemical assays and the metal Gd3+ using γ-radiation counting the radio-isotope 157Gd3+. It was clear from analyses of both protein CA1.CD2 and Gd3+ in multiple organ sites and blood that the PEGylation changed the bio-distribution of the protein contrast agent. First, PEGylation increased the blood circulation time of the agent. Before PEGylation modifications, a sharp decrease in blood concentration of both Gd3+ and CA1.CD2 was observed at about 50 minutes post i.v. injection. In contrast, no significant decrease was observed with PEGylated proteins 6 hour post injection. The longer circulation time was observed with the protein with higher degree of PEGylation modifications. Serum stability was further examined by measuring the protein content incubating with serum. Gegylated proteins remain stable after 48 hours.
  • 3.7 PEGylation Reduced Immunogenicity
  • Immunogenicity is one of the main concerns on the application of our developed protein MRI contrast agent. We therefore carried out experiments to test immunogenicity in rabbits by i.p. injection. The agents (CA1.CD2) were mixed with adjuvant or with buffer saline and injected at a dose of 3.0 nmole/kg according to the standard protocol for antibody production. The rabbits were subjected to double immunolizations in four weeks interval. Blood samples were taken from the immunolized rabbits 3 weeks after each injection. Production of antibodies against the protein contrast agent in each rabbit was examined by ELISA (FIG. 2.7 Left) and Immunobloting (FIG. 2.7 Right) using our previous polyclonal antibody PabCD2 as positive control and the pre-bleed from each rabbit as negative controls. It was clear that without adjuvant, injection of CA1.CD2 alone did not lead to antibody production in rabbits even after a double dose immunization. Addition of adjuvant indeed resulted in immuno-responses. PEGylation modifications of protein dramatically reduced immuno-responses. There was almost no antibody production in the first immunization. The production of antibody was negligible three weeks after the second immunization. Our results suggest that the immunogenicity of the protein contrast agent may not be very strong, especially without addition of adjuvant. Modification of the protein by PEGylation substantially reduced the immune responses.
  • FIG. 2.7 shows that PEGylation with P40 completely eliminates the binding of CA1.CD2 by antibody OX55 and decreases by 50% binding of OX34 in Western Blot. Since the epitope binding sites of OX55 and OX34 are well known, we have shown that K66, K45, K44, and K47 exhibit a high probability for the specific PEGylation by PEG12. These results confirm that antibody recognition sites can be eliminated by PEGylation, which is essential for the reduction of immunogenicity.
  • References for Example 2, each of which is incorporated herein by reference
    • 1. Gaberc-Porekar V, Zore I, Podobnik B, Menart V. Obstacles and pitfalls in the PEGylation of therapeutic proteins. Curr Opin Drug Discov Devel. 2008 March; 11(2):242-50.
    • 2. Fishburn C S. The pharmacology of PEGylation: Balancing PD with PK to generate novel therapeutics. J Pharm Sci. 2008 Jan. 15;
    • 3. Veronese F M, Pasut G. PEGylation, successful approach to drug delivery. Drug Discov Today. 2005 Nov. 1; 10(21):1451-8. Review.
    • 4. Harris J M, Chess R B. Effect of pegylation on pharmaceuticals. Nat Rev Drug Discov. 2003 March; 2(3):214-21. Review.
    • 5 Yang, W., Wilkins, A. L., Li, S., Ye, Y., and Yang, J. J. (2005) The effects of Ca2+ binding on the dynamic properties of a designed Ca2+-binding protein. Biochemistry 44, 8267-73.
    • 6 Adair, F., and Ozanne, D. (2002) The immunogenicity of therapeutic proteins. Biopharm—the Applied Technologies of Biopharmaceutical Development 15, 30-+.
    • 7 Leyland-Jones, B., Arnold, A., Gelmon, K., Verma, S., Ayoub, J. P., Seidman, A., Dias, R., Howell, J., and Rakhit, A. (2001) Pharmacologic insights into the future of trastuzumab. Ann Oncol 12 Suppl 1, S43-7.
    • 8 Hermeling, S., Crommelin, D. J., Schellekens, H., and Jiskoot, W. (2004) Structure-immunogenicity relationships of therapeutic proteins. Pharm Res 21, 897-903.
    • 9 Lucas, A., and McFadden, G. (2004) Secreted immunomodulatory viral proteins as novel biotherapeutics. J Immunol 173, 4765-74.
    • 10 Schellekens, H. (2002) Immunogenicity of therapeutic proteins: clinical implications and future prospects. Clin Ther 24, 1720-40; discussion 1719.
    • 10 Schellekens, H. (2002) Bioequivalence and the immunogenicity of biopharmaceuticals. Nat Rev Drug Discov 1, 457-62.
    • 11 Federico, R., Cona, A., Caliceti, P., and Veronese, F. M. (2006) Histaminase PEGylation: Preparation and characterization of a new bioconjugate for therapeutic application. J Control Release 115, 168-174.
    • 12 Veronese, F. M., and Pasut, G. (2005) PEGylation, successful approach to drug delivery. Drug Discov Today 10, 1451-8.
    • 13 Bhadra, D., Bhadra, S., Jain, P., and Jain, N. K. (2002) Pegnology: a review of PEG-ylated systems. Pharmazie 57, 5-29.
    • 14 Bailon, P., Palleroni, A., Schaffer, C. A., Spence, C. L., Fung, W. J., Porter, J. E., Ehrlich, G. K., Pan, W., Xu, Z. X., Modi, M. W., Farid, A., Berthold, W., and Graves, M. (2001) Rational design of a potent, long-lasting form of interferon: a 40 kDa branched polyethylene glycol-conjugated interferon alpha-2a for the treatment of hepatitis C. Bioconjug Chem 12, 195-202.
    • 15 Basu, A., Yang, K., Wang, M., Liu, S., Chintala, R., Palm, T., Zhao, H., Peng, P., Wu, D., Zhang, Z., Hua, J., Hsieh, M. C., Zhou, J., Petti, G., Li, X., Janjua, A., Mendez, M., Liu, J., Longley, C., Zhang, Z., Mehlig, M., Borowski, V., Viswanathan, M., and Filpula, D. (2006) Structure-function engineering of interferon-beta-1b for improving stability, solubility, potency, immunogenicity, and pharmacokinetic properties by site-selective mono-PEGylation. Bioconjug Chem 17, 618-30.
    • 16 Kozlowski, A., Charles, S. A., and Harris, J. M. (2001) Development of pegylated interferons for the treatment of chronic hepatitis C. BioDrugs 15, 419-29.
    • 17 Kozlowski, A., and Harris, J. M. (2001) Improvements in protein PEGylation: pegylated interferons for treatment of hepatitis C. J Control Release 72, 217-24.
    • 18 Yang, Z., Wang, J., Lu, Q., Xu, J., Kobayashi, Y., Takakura, T., Takimoto, A., Yoshioka, T., Lian, C., Chen, C., Zhang, D., Zhang, Y., Li, S., Sun, X., Tan, Y., Yagi, S., Frenkel, E. P., and Hoffman, R. M. (2004) PEGylation confers greatly extended half-life and attenuated immunogenicity to recombinant methioninase in primates. Cancer Res 64, 6673-8.
    • 19 Nie, Y., Zhang, X., Wang, X., and Chen, J. (2006) Preparation and stability of N-terminal mono-PEGylated recombinant human endostatin. Bioconjug Chem 17, 995-9.
    • 20 Daugherty, A. L., and Mrsny, R. J. (2006) Formulation and delivery issues for monoclonal antibody therapeutics. Adv Drug Deliv Rev 58, 686-706.
    • 21 Mohs, A. M., Zong, Y., Guo, J., Parker, D. L., and Lu, Z. R. (2005) PEG-g-poly(GdDTPA-co-L-cystine): effect of PEG chain length on in vivo contrast enhancement in MRI. Biomacromolecules 6, 2305-11.
    • 22 Luciani, A., Olivier, J. C., Clement, O., Siauve, N., Brillet, P. Y., Bessoud, B., Gazeau, F., Uchegbu, I. F., Kahn, E., Frija, G., and Cuenod, C. A. (2004) Glucose-receptor MR imaging of tumors: study in mice with PEGylated paramagnetic niosomes. Radiology 231, 135-42.23 Yang J J, Yang J, Wei L, Zurkiya O, Yang W, Li S, Zou J, Zhou Y, Maniccia A L, Mao H, Zhao F, Malchow R, Zhao S, Johnson J, Hu X, Krogstad E, Liu Z R. Rational design of protein-based MRI contrast agents. J Am Chem Soc, 2008. 130(29): p. 9260-7.
    Example 3 Developing Protein-Based MRI Contrast Agents with Multiple Metal-Binding Sites by Engineering of Natural Calcium Binding Proteins
  • Natural calcium binding proteins with continuous calcium binding sites such as calmodulin, calbindin D9K, troponin C, parvalbumin and discontinuous calcium binding sites such as thermintase subtilisin have exhibited high metal binding affinity for calcium as shown in Table 3.1 (shown in FIG. 3.10). Most of these calcium binding proteins have multiple calcium binding sites and their protein stabilities against temperature and proteolysis were significantly increased. For example, calcium binding to calmodulin has increased its stability to higher than 100° C. While a great deal of research has shown that the calcium binding affinity of naturally evolved proteins can be reduced by site-directed mutagenesis, methods to increase calcium binding affinity have rarely been reported. In addition, while lanthanide ions were shown to have coordination chemistry properties similar to calcium and often were able to compete with calcium for the metal binding sites, the accurate measurement of lanthanide metal binding affinities was not reported due to the limitation of extremely high metal binding affinity. Traditional titration methods by direct addition of metal ions monitoring lanthanide emission or Tb-sensitized energy transfer only provide low estimation of the metal binding sites. Furthermore, calcium ions in the strong metal binding sites of proteins are often difficult to remove and require careful treatment.
  • In this study, we report the engineering of protein based contrast agents by modifying calcium binding pockets of natural calcium binding proteins using calmodulin (CBP1) (FIG. 3.1) and parvalbumin (CBPP) as examples. These engineered proteins exhibit strong metal binding affinity to Gd3+ and other lanthanide (FIG. 3.2). Both proteins have strong Gd3+ binding affinity dissociation constants (Kd, M) of 1.0×10−15 to 1.0×10−18M. In addition, their selectivity over calcium and other metal ions such as zinc and magnesium is more than 105 fold higher, which is similar to that of clinically approved contrast agents such as DTPA or DTPA-BMA. CBPP and its variants exhibit strong metal selectivity for Ln3+ over calcium, magnesium, zinc and copper (FIG. 3.2 c). Furthermore, the water numbers in the coordination site can be estimated by Tb-sensitized energy transfer in water and in D2O. These data suggest that more than one water molecules are in the coordination shells and protein surface, and this likely contributes to their extremely high relaxivity. FIG. 3.8 shows that calmodulin and its variants can be pegylated with peg units of 4, 12, 40, 5K, 12K, 20 K and reaction products can be well separated by size-exclusion and ion exchange columns (FIG. 3.3 and FIG. 3.4).
  • Furthermore, functional sites of these engineered proteins, such as binding to the target molecules by calmodulin and parvalbumin, were eliminated by both deleting the molecular recognition sites and creating submodulin without completed recognition sites. Moreover, pegylation of these engineered proteins further reduced the natural function of CBP with increased solubility and reduced immunogenicity.
  • The conformation and metal binding properties of PEGylated proteins were not changed as revealed by Tyr emission spectra and Tb3+ sensitized energy transfer and CD methods (FIG. 3.5). FIG. 3.6 shows that both BCBP1 and CBBP1 and their variants have strong serum stability revealed by SDS page. These proteins remain intact upon incubation with serum greater than 48 hours.
  • These engineered proteins exhibit extremely high r1 and r2 relaxivities that are about 20 fold higher than DTPA. More strikingly, a clear in vivo mice imaging shown in FIG. 3.7 and FIG. 3.9 can be achieved using one dose tail vein injection with 6 mM of engineered protein contrast agent, CBP1 and its pegylated variant CBP1-P40. This dosage is about 70 fold lower than that of DTPA, with relaxivity values at 20-40 mM−1s−1 at 0.47 T. These values are comparable to CA1.CD2 and 5-8 fold greater than that of DTPA (FIG. 3.8). Example 3, FIG. 3.9 (left) illustrates mice MRI images at slice 3 (top) and slice 4 (bottom) at 4.7T with tail vein injection of 6 mM CBP1-P40 at 0, and 13 mins post injection. Relative MRI intensity at different organs is shown on right.
  • Based on the fact that the blood circulation and biodistribution can be further optimized and the existence of ample natural calcium binding proteins with multiple sites and different molecular weights/sizes, in addition to their superior capability for targeting our reported approach opens a new way to engineer MRI contrast agents with significantly improved relaxivity and in vivo properties for both blood pool and molecular imaging.
  • Example 4 Developing PEGylated Protein Contrast Agents Capable of Target Cancer Tumor Biomarkers Both in Cells and in Mice
  • Embodiment of the PEGyation method of the present disclosure was applied to enhance the performance of developed contrast agents in imaging cancer biomarker.
  • Example 4 describes a novel protein-based MRI contrast agent for molecular targeting cancer marker HER2 has been developed by modification with pegylation. The MRI contrast agent also carries a near-IR dye Cy5.5 for dual modality imaging of HER2 in cancer (Section 4.1).
  • In addition, Example 4 describes the development of a contrast agent that demonstrates specific interaction with HER2 positive cancer cells but not HER2 negative cells (4.2). The in vitro MR imaging experiments with different cells that express different levels of HER2 have shown a close correlation between MRI contrast enhancements and HER2 levels (Section 4.3).
  • Furthermore, Example 4 describes that the MR imaging of xenograft models of human HER2 positive cell line SKOV3 and HER2 negative cell line MDA-MB-231 has indicated a HER2 level-dependent MRI contrast enhancement. This image intensity enhancement is further confirmed by NIR imaging (Section 4.4).
  • In addition, Example 4 describes immunohistochemical staining of tissue samples (including tumor tissue) collected after imaging analyses has shown that the designed contrast agent penetrated deep into the tumor mass (i.e., distant from tumor vasculature), demonstrating a good tissue penetration of the MRI contrast agent. Differential targeting of the MRI contrast agent to HER2 positive and to HER2 negative tumors was further verified by immunoblot and ELISA analyses of the tissue samples collected from imaging mice (Section 4.5).
  • 4.1 Design of HER2 Targeting MRI Contrast Agent
  • To apply this MRI contrast agent in targeted molecular MR imaging, we fused a high affinity 58 amino acid HER2 affibody at the C-terminal of the protein via a flexible linker GGSGG. We also introduced a photo-probe by conjugating a near-IR dye Cy5.5 to a Cys residue added to the C-terminal of the protein (FIG. 4.1). The designed dual probe protein was expressed in E. coli and subsequently purified by procedures described in our previous report {{Yang, et. al., JACS 2008}, which is incorporated herein by reference}.
  • PEGylation of the designed Gd-binding protein not only increases protein solubility and blood circulation time, but also decreases immunogenicity of the protein without decrease of MRI relaxivity of the protein contrast agent. Therefore the designed HER2 targeting protein contrast agent was PEGylated using PEG-40, a PEG molecule with triple-branched 12 units PEG. The resulting agent (PEG-CA1-Affi) exhibits very similar Gd3+ binding properties and its T1 and T2 relaxivity values are similar to its parent protein CA1.CD2. Modifications did not change the overall protein fold as demonstrated by circular dichroism and fluorescence spectra.
  • 4.2 The HER2 Targeting MRI Contrast Agent Targeted to HER2 Positive Cells but not HER2 Negative Cells
  • We first examined whether the designed CA1.HER2/Affi can target cancer cells by cell binding analyses. We used two cancer cell lines for the binding analyses. AU565 is derived from breast carcinoma. EMT6 is a mouse mammary tumor cell line. AU565 cells express very high levels of HER2. The EMT is regarded as a HER2 negative cell line. Binding of the Gd-CA1.HER2/Affi to the cancer cells was first analyzed by immuno-fluorescence staining using a polyclonal antibody against PEGylated parental protein CA1.CD2 (PAbPGCA1). A substantial increase in staining intensities of CA1-Affi bound to AU565 cells was observed at both 37 and 4° C. The protein did not bind to EMT6 cells. As a control, CA1.CD2 without HER2 affibody did not bind to either of the tested cultured cells (FIG. 4.2). Proteins binding to cell surface HER2 with a clear membrane staining pattern in AU565 cells was observed at 4° C. The binding of the proteins to the cells triggered receptor-mediated endocytosis at 37° C. as demonstrated by the staining of the protein inside the cells. In addition, PEGylation of the targeted contrast agent PEG-CA1-Affi does not change its target capability the positive cell as shown in FIG. 4.2 at both 4 and 37° C. Further labeling with NIR dye Cy5.5, the binding of the contrast agent (PEG-CA1-Affi-Cy5.5) to both AU565 and SKOV-3, a cell line derived from ovarian cancer with very high HER2 expression, is readily detected by our fluorescence microscope.
  • Binding of the Gd-CA1-Affi to the cancer cells was further analyzed by quantification of cell bound Gd3+ with AU565 and EMT6 cells. The amounts of Gd3+ were quantified by γ-counting the trace of isotope 153Gd3+ in the Gd-protein complexes. The quantification of cell bound Gd3+ supported our immuno-analyses that Gd-CA1-Affi exhibited targeted binding to AU565 cells but not to EMT6 cells (FIG. 4.3). The protein without the HER2 affibody targeting moiety could not bind to the cells that express HER2. Interestingly, calculating the amounts of bound Gd3+ from γ-counting revealed that the Gd3+ ions were bound to cells at ˜0.2 fmole Gd/cell. Based on the assumption that 1×107 cells occupy a volume of 50-100 μl, this binding capacity leads to the accumulation of Gd3+ at 5-15 μM in the cell pellets. This local concentration is sufficient to produce strong MRI contrast, especially with high relaxivity protein-based contrast agent. Further, the level of HER expression can be monitored by ELISA (FIG. 4.3).
  • 4.3 MRI Imaging of Different Cancer Cells by the Developed HER2 Targeting Contrast Agents.
  • Experiments demonstrated that the developed MRI contrast agent binds specifically to HER2 expression cells. To test whether our designed proteins are applicable for targeted imaging, we carried out MR imaging of cancer cells that were incubated with the designed protein contrast agents Gd-CA1-Affi using four different cancer cell lines with different expression levels of HER2. Cancer cells AU565 and SKOV3, with high numbers of HER2, exhibit a brighter imaging in the presence of our contrast agent Gd-CA1.HER2-Affi. In contrast, no significant changes in MR image were observed in the imaging pellets of AU565 and SKOV3 cells that were incubated with Gd-CA1.CD2, without the HER2 targeting moiety. The MR imaging of MDA-MB-231, another breast cancer cell line with limited HER2 expression, demonstrated slight contrast enhancement. Almost no contrast enhancement was observed with MR imaging of EMT6 cells using our HER2 targeting contrast agent. Quantification of the MR image intensity revealed a strong correlation between MR imaging intensity and the HER2 levels among the different cell lines used. These results confirm that the addition of HER2 targeting affibody to our protein contrast agent has the potential to provide molecular imaging of cancer cells via receptor-mediated recognition. The MR image contrast enhancements from the contrast agent can be correlated to the HER2 levels.
  • 4.4 The HER2 Targeting Contrast Agent Provided Strong MR Image Contrast Enhancement in Nude Mice Xenograft Model
  • We next tested whether our designed contrast agent would produce MR imaging contrast enhancement in nude mice xenograft models of two human cancer cell lines, SKOV3 and MDA-MB-231. Since AU565 did not grow tumors in nude mice, we used SKOV3 and MDA-MB-231 cell lines. The HER2 positive SKOV3 and negative MDA-MB-231 tumors were implanted on the right and left flanks, respectively, of different mice (FIG. 4.5). The contrast agent Gd-CA1.HER2/Affi (60 μl) was introduced via the tail vein at concentration of 6 mM. Pre- and post-contrast MRIs were collected at different time points using T1 weighted spin echo or gradient echo sequences. At an early time point (40 minutes), little contrast enhancement was observed with either HER2 positive or negative tumors. The contrast enhancements with both positive and negative tumors were apparent at 4 hours post-contrast agent injection. However, after 21 hours post-contrast, the image enhancements of the negative tumor decreased dramatically, while conversely there were no significant decreases in the MRI intensities in the positive tumor. In parallel, the mice were imaged using a Kodak in vivo FX-pro animal imaging system. Consistent with MRI imaging, we observed strong NIR light emission from both positive and negative tumor sites at early time points (50 minutes post-injection). After 24 hours post-contrast, the NIR intensities at the negative tumor site were almost identical with the background, while the light intensities at the positive tumor experienced only a minor decrease (FIG. 4.5). The in vivo imaging experiments were repeated with four tumor-bearing mice (four positive tumor mice and four negative tumor mice) with similar results. FIG. 4.4, shows the result of another mouse. 5 mM of contrast agent CA1.Affi-P40 (100 fold lower than clinic used DTPA) was injected via tail vein. MRI images at 4.7 T using fast spin echo were acquired before injection, 5 min, 30 min, 3 hr, 24 hr and 52 hr post injection. Positive tumor shows a strong contrast after 30 mins and peaked at 24 hour with about 35% enhancement. Contrast capability was decreased after 52 hours, suggesting that the contrast agent was secreted of out the animal. This mouse was alive and looks normal after 52 hours MRI scanning.
  • 4.5 IHC Analyses and NIR Imaging Indicate Molecular Targeting Rather than Vascular Retention
  • To further analyze the HER2 targeting properties of the protein contrast agent, tumors and organs from the imaged mice were collected after imaging at 45 minutes or 24 hours. The organs and tumors were imaged under the Kodak in vivo FX-pro animal imaging system. It was clear that there were very high levels of accumulation of Cy5.5 at the liver and HER2 positive tumor, with moderate levels of the NIR dye present in the kidney and spleen. NIR fluorescence intensities in the lung and heart were very weak. The IHC analyses are consistent with our biodistribution analyses of the contrast agent in CD1 mouse. The levels of Cy5.5 at the negative tumor were also very weak (FIG. 4.6). There was almost no detectable NIR fluorescence in the muscles.
  • Targeting of the protein contrast agent to HER2 positive tumor was further analyzed by immunoblot. To this end, protein extracts were made from the tissue samples that were collected from the imaged mice. Immunoblot experiments were performed with the protein extracts using the antibody PAbPGCA1. Consistently, we detected very high levels of PEGylated protein contrast agent in extracts made from liver, kidney, and positive tumor. The antibody detected very faint bands in the extracts made from muscle and negative tumor samples (FIG. 4.6). The results strongly suggested that our protein contrast agent led to a HER2 specific MR image enhancement.
  • In addition, we carried out immunohistochemistry (IHC) staining using the antibody PAbPGCA1 with tissue slides made from the tissue samples from the imaged mice, including HER2 positive and negative tumors. Strongest staining was observed with liver and HER2 positive tumor tissue slides (FIG. 4.7). Close examination of the staining patterns of the positive tumor slides revealed very high CA1-Affi protein levels inside the cancer cells, indicating internalization (endocytosis) of the protein contrast agent. The results also suggested that the contrast agent penetrated the tumor tissue, rather than being trapped in the tumor vasculature. This is an important property for HER2 targeting in the whole cancer mass. The kidney slides also gave strong immunostaining. Interestingly, the areas near proximal tubes showed the strongest staining in the slides made from the kidney (FIG. 4.7), indicating that the protein contrast agent was ready to be filtered through the kidney. This is consistent with observations that there were good levels of both Gd3+ (by γ-counting of Gd3+-153) and protein CA1-Affi (by NIR fluorescence) in the urine of mice that were injected with PEGylated Gd-CA1-Affi. Co-immunostaining the slides made from HER2 positive tumor with the antibody PAbPGCA1 and the antibody against CD31, an endothelial molecular marker, demonstrated that the protein contrast agent largely localized in tissue areas other than the tumor micro-vascular structure (CD31 positive areas). This staining pattern suggested that the designed protein contrast had penetrated tissue rather than simply being trapped in the blood in the micro-vasculature of the tumor tissue.
  • 4.6 HER2-Targeted Contrast Agent has been Generated by Engineering Metal Binding Sites in Natural Calcium Binding Proteins
  • We have created the second generation contrast agent by grafting affibody within the engineered multiple metal binding protein CBP1 based on calmodulin (CBP1-affi). In addition GRP/Bombesin sequence was also inserted into CBP1 to create a contrast agent to specifically target to prostate cancer with GRPR over expressed (CBP1-Bom). These targeted contrast agents were then PEGylated by PEG with different units and purified. Initial binding to HER2 has been studied. The HER2 positive cell line SKOV-3 and HER2 negative cell line MDA-MB-231 was treated with CBP1-affi at different concentration: 10 μM and 20 μM in 1 ml medium. Both of the cell numbers is about 1×105. One hour later, the medium will be carefully washed for 3 times. The non-binding proteins will be washed away. Then the cells will be lysized by RIPA buffer for 30 min and centrifuged to obtain the cell lysate. The cell lysate is used to run western blot to measure the binding of CBP1-affi with HER2 positive cells. FIG. 4.8 shows that the bands in SKOV-3 cells are much darker than those in MDA-MB-231 cells, which means the binding of CBP1-affi to HER2 is specific.
  • The two cell lines been treated with CBP1-affi are fixed in chamber by cold methanol for 5 min. Then primary antibody against CBP1-affi and secondary antibody will be applied consequently. Finally, the slides will be mounted by anti fade reagent with DAPI staining. The second day, images will be taken under microscope. The green color indicates FITC staining on CBP1-affi; the blue color indicates the nuclear staining. FIG. 4.9 shows the HER2 positive cell line SKOV-3 has been stained. The staining on the negative cell line is rarely.
  • 4.7 GRP Receptor Targeted Contrast Agents Have been Created for Both NIR Fluorescence and MRI to Image Prostate Cancer Both in Cells and in Mice
  • We have also created targeted contrast agents to against biomarker GRPR that are over-expressed in prostate, colon, breast, and pancreatic cancers by grafting Bom/GRP sequence into CA1.CD2 (CA1.CD2-52I-Bom. This protein was also pegylated by PEG40 (CA1.CD2-52I-Bom-P40). NIR dye such as Cy5.5 was also conjugated at the C-terminal Cys (CA1.CD2-52I-Bom-P40-Cy5.5). FIG. 4.9 shows the uptake and internalization of CA1.Bom (CA1.CD2-52I-Bom) specifically to GRPR positive cell lines (PC-3 and DU-145 cells) by using confocal fluorescent microscope.
  • DU-145, PC-3 and H441 cells (8×104) were seeded in 4-well chambers (BD) at 37° C. overnight. In the second day, fresh medium was changed. Contrast agents were incubated with cells at different time point at 37° C. Subsequently, the cells were washed with PBS for three times and fixed with 3% formaldehyde for 15 minutes. After cells were rinsed in PBS for three times, 0.2% Triton X-100 was added to permeablize cells for 10 minutes. The cells were washed and four drops of Image-IT Fx signal enhancer (Invitrogen, CA) was applied in each sample to block unspecific binding. After three time washes, CD2 antibody was used as primary antibody. Goat anti-mouse IgG conjugated with Alexa Fluor 488 was used as detection antibody (Invitrogen, CA). The fluorescence image was acquired using 488 nm and UV lasers.
  • FIG. 4.11 shows NIR-fluorescence imaging of nude mice xenografted with DU-145 tumor (positive control left) and H441 tumor (control, right) post injection of CA1.CD2-52Ibom-cy5.5-P40 26 hours via tail vein. 50 uM Cy5-CA1.CD2-52I was injected into mouse tail vein. After 26 hours, mouse was analyzed by Kodak Imaging System. Tissue organs were taken out and analyzed. Tumor intensity was analyzed by Image J. FIG. 4.12 shows NIR imaging (top) and NIR intensity (bottom) of CA1.CD2-52I-Bom-Cy5.5-P40 at different organs of the mice. The developed contrast agent is able to target to the GRPR expressed tumor monitored by NIR fluorescence.
  • It should be noted that ratios, concentrations, amounts, and other numerical data may be expressed herein in a range format. It is to be understood that such a range format is used for convenience and brevity, and thus, should be interpreted in a flexible manner to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. To illustrate, a concentration range of “about 0.1% to 5%” should be interpreted to include not only the explicitly recited concentration of about 0.1 wt % to about 5 wt %, but also include individual concentrations (e.g., 1%, 2%, 3%, and 4%) and the sub-ranges (e.g., 0.5%, 1.1%, 2.2%, 3.3%, and 4.4%) within the indicated range. The term “about” can include ±1%, ±2%, ±3%, ±4%, ±5%, ±6%, ±7%, ±8%, ±9%, or ±10%, or more of the numerical value(s) being modified. In addition, the phrase “about ‘x’ to ‘y’” includes “about ‘x’ to about ‘y’”.
  • The above discussion is meant to be illustrative of the principles and various embodiments of the present disclosure. Numerous variations and modifications will become apparent to those skilled in the art once the above disclosure is fully appreciated. It is intended that the following claims be interpreted to embrace all such variations and modifications.
  • Sequence Listings:
  • Listed sequences are representative examples from protein families where sequences of the same protein across different species typically share 50-98% sequence homology. As an embodiment, attributes in the cited sequence can be inferred to apply to homologous, related sequences, which would be known to someone familiar with the art.
  • K Possible PEGylated sites
    X Mutation sites, C is added at the C-terminal for specific pegylation or conjugating to fluorescence dye. Specific pegylation can also be at N-terminal
    These sequences include for different species. A SEQ ID No: (“#”) is noted by each sequence below.
  • Rat Calmodulin and its Variants (CBPO1 and Variants)
  • WT (wild type) (1) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN  60
    D93A (2) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN  60
    D129A (3) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN  60
    D56A (4) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVAADGN  60
    D20A (5) ADQLTEEQIAEFKEAFSLFAKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN  60
    Y99W (6) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN  60
    T26W (7) ADQLTEEQIAEFKEAFSLFDKDGDGWITTKELGTVMRSLGQNPTEAELQDMINEVDADGN  60
    N60D-N97D (8) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGD  60
    Deletion (9) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN  60
    D-N60D-N97D (10) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGD  60
    WT GTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDIDGNGYISAAELRHVMTNLGEKLTDEE 120
    D93A GTIDFPEFLTMMARKMKDTDSEEEIREAFRVFAIDGNGYISAAELRHVMTNLGEKLTDEE
    120
    D129A GTIDFPEFETMMARKMKDTDSEEEIREAFRVFDIDGNGYISAAELRHVMTNLGEKLTDEE 120
    D56A GTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDIDGNGYISAAELRHVMTNLGEKLTDEE 120
    D20A GTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDIDGNGYISAAELRHVMTNLGEKLTDEE 120
    Y99W GTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDIDGNGWISAAELRHVMTNLGEKLTDEE
    120
    T26W GTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDIDGNGYISAAELRHVMTNLGEKLTDEE 120
    N60D-N97D GTIDEPEFLTMMARKMKDTDSEEEIREAFRVFDKDGDGYISAAELRHVMTNLGEKLTDEE 120
    Deletion GTIDFPEFLTMMARK------EEEIREAFRVFDKDGNGYISAAELRHVMTNLGEKLTDEE 115
    D-N60D-N97D GTIDFPEFLTMMARK------EEEIREAFRVFDKDGDGYISAAELRHVMTNLGEKLTDEE 115
    WT VDEMIREADIDGDGQVNYEEFVQMMTAK 148
    D93A VDEMIREADIDGDGQVNYEEFVQMMTAK 148
    D129A VDEMIREAAIDGDGQVNYEEFVQMMTAK 148
    D56A VDEMIREADIDGDGQVNYEEFVQMMTAK 148
    D20A VDEMIREADIDGDGQVNYEEFVQMMTAK 148
    Y99W VDEMIREADIDGDGQVNYEEFVQMMTAK 148
    T26W VDEMIREADIDGDGQVNYEEFVQMMTAK 148
    N60D-N97D VDEMIREADIDGDGQVNYEEFVQMMTAK 148
    Deletion VDEMIREADIDGDGQVNYEEFVQMMTAK 143
    D-N60D-N97D VDEMIREADIDGDGQVNYEEFVQMMTAK 143
  • Rat N-Calmodulin (N-Terminal Domain of Calmodulin and Variants)
  • N-CaM (11) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGN 60
    N-N6OD (12) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADGD 60
    N-CaM GTIDFPEFLTMMARK 75
    N-N6OD GTIDFPEFLTMMARK 75
  • Rat C-Calmodulin (N-Terminal Domain of Calmodulin and Variants)
  • C-CaM (13)
    MKDTDSEEEIREAFRVFDIDGNGYISAAELRHVMTNLGEKLTDEEVDEMIREADIDGDGQ 60
    C-N97D (14)
    MKDTDSEEEIREAFRVFDIDGDGYISAAELRHVMTNLGEKLTDEEVDEMIREADIDGDGQ 60
    C-CaM
    VNYEEFVQMMTAK 73
    C-N97D
    VNYEEFVQMMTAK 73
  • Rat Calmodulin-Affibody (15)
  • ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQD
    MINEVDADGDGTIDFPEFLTMMARKMKDTGGSGGVDNKFNKEMRNAYWEI
    ALLPNLNNQQKRAFIRSLYDDPSQSANLLAEAKKLNDAQAPKGGSGGDSE
    EEIREAFRVFDKDGDGYISAAELRHVMTNLGEKLTDEEVDEMIREADIDG
    DGQVNYEEFVQMMTAK
  • Rat CaM-Bombesin (16)
  • ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQD
    MINEVDADGDGTIDFPEFLTMMARKMKDTGGNQWAVGHLMGGDSEEEIRE
    AFRVFDKDGDGYISAAELRHVMTNLGEKLTDEEVDEMIREADIDGDGQVN
    YEEFVQMMTAK
  • Calmodulin Different Species
  • Human (17) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDADDL  60
    Mouse (18) ADQLTEEQIAEFKEAFSLFDKDGDNTITTKELGTVMRSLGQNPTEAELQDMINEVDAD--  58
    Rat (19) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDAD--  58 
    Rabbit (20) ADQLTEEQIAEFKEAFSLFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDAD--  58
    Paramecium (21) AEQLTEEQIAEFKEAFALFDKDGDGTITTKELGTVMRSLGQNPTEAELQDMINEVDAD--  58
    Human PGNGTIDFPEFLTMMARKMKDTDSEEEIREAFRVEDKDGNGYISAAELRHVMTNLGEKLT 120
    Mouse -GNGTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDKDGNGYISAAELRHVMTNLGEKLT 117
    Rat -GNGTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDKDGNGYISAAELRHVMTNLGEKLT 117
    Rabbit -GNGTIDFPEFLTMMARKMKDTDSEEEIREAFRVFDKDGNGYISAAELRHVMTNLGEKLT 117
    Paramecium -GNGTIDFPEFLSLMARKMKEQDSEEELIEAFKVFDRDGNGLISAAELRHVMTNLGEKLT 117
    Human DEEVDEMIREADIDGDGQVNYEEFVQMMTAK 151
    Mouse DEEVDEMIREADIDGDGQVNYEEFVQMMTAK 148
    Rat DEEVDEMIREADIDGDGQVNYEEFVQMMTAK 148
    Rabbit DEEVDEMIREADIDGDGQVNYEEFVQMMTAK 148
    Paramecium DDEVDEMIREADIDGDGHINYEEFVRMMVSK 148
  • Rat CA1-CD2-Affibody
  • CA1-WT (22) GSRDSGTVWGALGHGIELNIPNFQMTDDIDEVRWERGSTLVAEFKRKMKPFLKSGAFEID  60
    CA1-ZHER2-4 (23) GSRDSGTVWGALGHGIELNIPNFQMTDDIDEVRWERGSTLVAEFKRKMKPFLKSGAFEID  60
    CA1-ZHER342 (24) GSRDSGTVWGALGHGIELNIPNFQMTDDIDEVRWERGSTLVAEFKRKMKPFLKSGAFEID  60
    CA1-WT ANGDLDIKNLTRDDSGTYNVTVYSTNGTRILNKALDLRILEGGSGGVDNKENKEQQNAFY 120
    CA1-ZHER2-4 ANGDLDIKNLTRDDSGTYNVTVYSTNGTRILNKALDLRILEGGSGGVDNKFNKELRQAYW 120
    CA1-ZHER342 ANGDLDIKNLTRDDSGTYNVTVYSTNGTRILNKALDLRILEGGSGGVDNKFNKEMRNAYW 120
    CA1-WT EILHLPNLNEEQRNAFIQSLKDDPSQSANLLAEAKKLNDAQAPK 164
    CA1-ZHER2-4 EIQALPNLNWTQSRAFIRSLYDDPSQSANLLAEANKLNDAQAPK 164
    CA1-ZHER342 EIALLPNLNNQQKRAFIRSLYDDPSQSANLLAEAKKLNDAQAPK 164
  • Rat CA1-CD2-Bombesin (C-Terminal) (25)
  • RDSGTVWGAL GHGIELNIPN FQMTDDIDEV RWERGSTLVA
    EFKRKMKPFL KSGAFEIDAN GDLDIKNLTR DDSGTYNVTV
    YSTNGTRILN KALDLRILEG GSGGSGNQWA VGHLM
  • Rat CA1-CD2-Bombesin (52I) (26)
  • RDSGTVWGAL GHGIELNIPN FQMTDDIDEV RWERGSTLVA
    EFKRKMKPFL KSGGSGGGNQ WAVGHLMGGS GGGAFEIDAN
    GDLDIKNLTR DDSGTYNVTV YSTNGTRILN KALDLRILE
  • Rat Parvalbumin
  • WT (27) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKSGFIE  60
    S56D (28) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    S56D-F103W(29) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    E60D (30) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKSGFID  60
    E60D-F103W (31) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKSGFID  60
    G99D (32) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKSGFIE  60
    G99D-F103W (33) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKSGFIE  60
    D53S-F103W (34) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILSKDKSGFIE  60
    D53E-F103W (36) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILEKDKSGFIE  60
    F103WC104 (37) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKSGFIE  60
    F103W (38) MSMTDELSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKSGFIE  60
    Human (39) MSMTDELNAEDIKKAVGAFSATDSFDHKKFFQMVGLKKKSADDVKKVFHMLDKDKSGFIE  60
    WT EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEFSTLVAES- 110
    S56D EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEFSTLVAES- 110
    S56D-F103W EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAES- 110
    E6OD EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEFSTLVAES- 110
    E60D-F103W EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAES- 110
    G99D EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIDVEEFSTLVAES- 110
    G99D-F103W EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIDVEEWSTLVAES- 110
    G99D-F103W EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIDVEEWSTLVAES- 110
    G99D-F103W EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIDVEEWSTLVAES- 110
    F103WC104 EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAESC 111
    F103W EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAES- 110
    Human EDELGFILKGFSPDARDLSAKETKMLMAAGDKDGDGKIGVDEFSTLVAES- 110
  • Rat-Parvalbumin Insertion Variants
  • PV_Collagen (39) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    PV_Bombsin (40) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    PV_Selectin (41) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    PV_RGD (42) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    PV_Cys (43) MSMTDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    PV_AFFIBODY (44) MSMIDLLSAEDIKKAIGAFTAADSFDHKKFFQMVGLKKKSADDVKKVFHILDKDKDGFIE  60
    PV_Collagen EDELGSILKGFSSDARDLSAKETKTLMAAGDEDGDGKIGVEEWSTLVAESGGGKKWHCYT 120
    PV_Bombsin EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAESGGGAQMAVGH
    120
    PV_Selectin EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAESGGG-KYDGDI 119
    PV_RGD EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAESGGGRGDRGDR
    120
    PV_Cys EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAESC--------- 111
    PV_AFFIBODY EDELGSILKGFSSDARDLSAKETKTLMAAGDKDGDGKIGVEEWSTLVAESGGSGGVDNKF
    120
    PV_Collagen YFPNHYCVYG------------------------------------------- 130
    PV_Bombsin LM--------------------------------------------------- 122
    PV_Selectin TWDQLWDLMK------------------------------------------- 129
    PV_RGD GDRGD------------------------------------------------ 125
    PV_Cys ------------------------------------------------------
    PV_AFFIBODY NKEMRNAYWEIALLPNLNNQQKRAFIRSLYDDPSQSANLLAEAKKLNDAQAPK 173
  • Human-Calbindin D9k
  • CalbindinD9k (45) MSTKKSPEELKRIFEKYAAKEGDPDQLSKDELKLLIQAEFPSLLKGPNTLDDLFQELDKN 60
    CalbindinD9kF67W (46) MSTKKSPEELKRIFEKYAAKEGDPDQLSKDELKLLIQAEFPSLLKGPNTLDDLFQELDKN 60
    CalbindinD9kP43M (47) MSTKKSPEELKRIFEKYAAKEGDPDQLSKDELKLLIQAEFPSLLKGMNTLDDLFQELDKN 60
    CalbindinD9kCys (48) MSTKKSPEELKRIFEKYAAKEGDPDQLSKDELKLLIQAEFFSLLKGPNTLDDLFQELDKN 60
    CalbindinD9k GDGEVSFEEFQVLVKKISQ- 79
    CalbindinD9kF67W GDGEVSFEEWQVLVKKISQ- 79
    CalbindinD9kP43M GDGEVSFEEWQVLVKKISQ- 79
    CalbindinD9kCys GDGEVSFEEFQVLVKKISQC 80
  • Human-Troponin C (49)
  • MTDQQAEARSYLSEEMIAEFKAAFDMFDADGGGDISVKELGTVMRMLGQT
    PTKEELDAIIEEVDEDGSGTIDFEEFLVMMVRQMKEDAKGKSEEELAECF
    RIFDRNADGYIDPGELAEIFRASGEHVTDEEIESLMKDGDKNNDGRIDFD
    EFLKMMEGVQ
  • Rat CA1-CD2-Bombesin-RGD (52I)-Cend (50)
  • RDSGTVWGAL GHGIELNIPN FQMTDDIDEV RWERGSTLVA
    EFKRKMKPFL KSGGSGGGNQ WAVGHLMGGS GGGAFEIDAN
    GDLDIKNLTR DDSGTYNVTV YSTNGTRILN KALDLRILE-RGD
  • Rat CA1-CD2-Bombesin-RGD (52I)-Need (51)
  • RGDRDSGTVWGAL GHGIELNIPN FQMTDDIDEV RWERGSTLVA
    EFKRKMKPFL KSGGSGGGNQ WAVGHLMGGS GGGAFEIDAN
    GDLDIKNLTR DDSGTYNVTV YSTNGTRILN KALDLRILE
  • Rat CA1-CD2-RGD-83I (52)
  • RDSGTVWGAL GHGIELNIPN FQMTDDIDEV RWERGSTLVA
    EFKRKMKPFL KSGAFEIDAN GDLDIKNLTR DDSGTYNVTV
    YSTGGSGGRGDGGSGGNGTRILN KALDLRILEG
  • Rat CA1-CD2-Bom-52I-RGD-83I (53)
  • RGDRDSGTVWGAL GHGIELNIPN FQMTDDIDEV RWERGSTLVA
    EFKRKMKPFL KSGGSGGGNQ WAVGHLMGGS GGGAFEIDAN
    GDLDIKNLTR DDSGTYNVTV YSTGGSGGRGDGGSGGNGTRILN
    KALDLRILE

Claims (46)

1. A contrast agent comprising:
a) a scaffold protein, and
b) at least one metal ion chelating site, wherein the scaffold protein includes at least one metal ion chelating site that is already present or is integrated into the scaffold protein, wherein the scaffold protein includes a metal ion bound to a metal ion chelating site,
wherein the contrast agent is stable in a physiological environment.
2. The contrast agent of claim 1, wherein the metal is Gd(III).
3. The contrast agent of claim 2, wherein the scaffold protein includes at least one polyethylene glycol (PEG) group attached to the scaffold protein.
4. The contrast agent of claim 3, further comprising a targeting agent attached to the scaffold protein.
5. The contrast agent of claim 4, wherein the targeting agent is selected from the group consisting of: a biomarker probe, a precancerous targeting agent, a cancer targeting agent, a tumor targeting agent, and a probe or agent that targets at least two of a biomarker, a precancerous cell, a cancer cell, and a tumor.
6. The contrast agent of claim 3, wherein one or more metal ion chelating sites are integrated into the scaffold protein, and wherein each of the metal ion chelating sites include one Gd(III) bound to each of the metal ion chelating sites.
7. The contrast agent of claim 6, further comprising a targeting agent attached to the scaffold protein.
8. The contrast agent of claim 6, wherein a Near-IR functional group for Near-IR detection is covalently bound to the scaffold protein.
9. The contrast agent of claim 7, wherein the targeting agent is selected from the group consisting of: a biomarker probe, a precancerous targeting agent, a cancer targeting agent, a tumor targeting agent, and a probe or agent that targets at least two of a biomarker, a precancerous cell, a cancer cell, and a tumor.
10. The contrast agent of claim 1, wherein the scaffold protein includes at least one modification to the protein sequence, wherein the modification of the metal binding site alters the metal selectivity of the binding site, optimizes binding affinity for the metal, or improves serum stability of the contrast agent.
11. The contrast agent of claim 1, wherein the scaffold protein includes at least one polyethylene glycol (PEG) group attached to the scaffold protein.
12. The contrast agent of claim 11, wherein the PEGylation of the contrast agent increased the solubility of protein by more than 100 fold without reducing the metal binding affinity and selectivity relative to a contrast agent that does not include the PEG.
13. The contrast agent of claim 11, wherein the serum stability of the contrast agent is improved relative to a contrast agent that does not include the PEG.
14. The contrast agent of claim 11, wherein PEGylation of the contrast agent increases the in vitro R1 and R2 relaxivities of the contrast agents by at least 2-fold relative to a contrast agent that does not include the PEG.
15. The contrast agent of claim 11, wherein the contrast agent exhibits a significantly-increased blood circulation time relative to a contrast agent not including the PEG.
16. The contrast agent of claim 11, wherein the immunogenicity of the contrast agent is significantly decreased relative to a contrast agent not including the PEG.
17. The contrast agent of claim 11, wherein the PEG has a molecular weight of about 1 to 100 kDa.
18. The contrast agent of claim 11, wherein the PEG has a molecular weight of about 1 to 20 kDa.
19. The contrast agent of claim 11, wherein the PEG is attached to the scaffold protein via an amino acid selected from: lysine, glutamic acid, aspartic acid, cysteine, carboxyl/amino terminals, and combinations thereof.
20. The contrast agent of claim 11, wherein the PEG is attached to the scaffold protein via lysine.
21. The contrast agent of claim 1, wherein at least one metal ion chelating site is embedded within the scaffold protein.
22. The contrast agent of claim 1, wherein at least one metal ion chelating site is substantially embedded within the scaffold protein.
23. The contrast agent of claim 22, wherein the relaxivity of the contrast agent is greater than that of the scaffold protein relative to a contrast agent not including the PEG.
24. The contrast agent of claim 1, wherein the metal is a paramagnetic metal ion selected from the group consisting of: Gd(III), Mn(II), Fe(II), Fe(III), Co(II), Co(III), Ni(III), Mo(V), and V(IV).
25. The contrast agent of claim 1, wherein the metal is an ion of a metal selected from the group consisting of Lanthanide Series metals.
26. The contrast agent of claim 1, wherein the scaffold protein is selected from immunoglobulin proteins.
27. The contrast agent of claim 1, wherein the scaffold protein is selected from proteins derived from a fluorescent protein having a chromophore.
28. The contrast agent of claim 1, further comprising a targeting agent attached to the scaffold protein.
29. The contrast agent of claim 28, wherein the targeting agent is selected from the group consisting of: a biomarker probe, a precancerous targeting agent, a cancer targeting agent, a tumor targeting agent, and a probe or agent that targets of a biomarker, a precancerous cell, a cancer cell, and a tumor.
30. The contrast agent of claim 28, wherein the targeting agent is selected from the group consisting of: a gastrin release peptide (GRP) and a RGD (Arg-Gly-Asp) peptide.
31. The contrast agent of claim 1, wherein the scaffold protein is a natural metal binding site protein, wherein the natural metal binding site protein includes a natural metal binding site, and wherein the metal ion binding site is derived from a natural metal binding site.
32. The contrast agent of claim 31, wherein the natural metal binding protein is selected from a group consisting of: calmodulin, calbindin D9K, troponin C, and parvalbumin.
33. The contrast agent of claim 1, wherein binding the metal ion causes no changes to the protein conformation or to the binding affinity under clinical conditions that would cause premature release of the metal ion, and that the contrast agent will function as a contrast agent.
34. The contrast agent of claim 1, wherein the tailored metal ion binding site is present in an unmodified protein of a scaffold protein.
35. The contrast agent of claim 1, wherein the tailored metal ion binding site is integrated into the scaffold protein.
36. The contrast agent of claim 1, wherein the scaffold protein includes a fragment or domain of a natural metal binding protein.
37. The contrast agent of claim 1, wherein the scaffold protein functions independently or in a complex with another cofactor, ligand or other biomolecule.
38. A method of imaging a sample comprising:
administering at least one of the contrast agent of claims 1-37 to the sample;
introducing the sample to an imaging system; and
imaging the sample.
39. The method as claimed in claimed 38, wherein the imaging system is selected from the group consisting of: a nuclear magnetic resonance system, a Near-IR system, a PET system, and a combination thereof.
40. The method as claimed in claimed 38, wherein the imaging system is an optical imaging system.
41. A method for preparing a contrast agent comprising the steps of:
a) selecting a scaffold protein;
b) constructing at least one metal ion chelating site;
c) operatively embedding the metal ion binding site into the protein, wherein the metal ion has contrast agent properties, and
d) attaching at least one polyethylene glycol (PEG) to the scaffold protein.
42. The method of claim 41, wherein attaching includes:
attaching the PEG to the scaffold protein via an amino acid selected from the group consisting of: lysine, glutamic acid, aspartic acid, cysteine, carboxyl/amino terminals, and a combination thereof.
43. The method of claim 41, wherein attaching includes:
attaching the PEG to the scaffold protein via lysine.
44. The method of claim 41, wherein the PEGs are attached to amino acid residues so that the PEGs do not substantially interfere with or interfere with the metal ion binding site ability to interact with the metal ion of interest or the conformation of the polymer.
45. The method of claim 41, wherein constructing at least one metal ion chelating site includes constructing at least two metal ion chelating sites.
46. The method of claim 41, further comprising:
attaching at least one targeting agent to the scaffold protein.
US12/935,413 2008-04-02 2009-04-02 Contrast agents, methods for preparing contrast agents, and methods of imaging Abandoned US20110097742A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/935,413 US20110097742A1 (en) 2008-04-02 2009-04-02 Contrast agents, methods for preparing contrast agents, and methods of imaging

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US4169308P 2008-04-02 2008-04-02
US12/935,413 US20110097742A1 (en) 2008-04-02 2009-04-02 Contrast agents, methods for preparing contrast agents, and methods of imaging
PCT/US2009/039276 WO2009146099A2 (en) 2008-04-02 2009-04-02 Contrast agents, methods for preparing contrast agents, and methods of imaging

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/039276 A-371-Of-International WO2009146099A2 (en) 2008-04-02 2009-04-02 Contrast agents, methods for preparing contrast agents, and methods of imaging

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/910,893 Continuation US10849993B2 (en) 2008-04-02 2018-03-02 Contrast agents, methods for preparing contrast agents, and methods of imaging

Publications (1)

Publication Number Publication Date
US20110097742A1 true US20110097742A1 (en) 2011-04-28

Family

ID=41377862

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/935,413 Abandoned US20110097742A1 (en) 2008-04-02 2009-04-02 Contrast agents, methods for preparing contrast agents, and methods of imaging
US15/910,893 Active 2029-04-19 US10849993B2 (en) 2008-04-02 2018-03-02 Contrast agents, methods for preparing contrast agents, and methods of imaging
US17/068,215 Active 2030-01-30 US11738098B2 (en) 2008-04-02 2020-10-12 Contrast agents, methods for preparing contrast agents, and methods of imaging
US18/346,503 Pending US20240082434A1 (en) 2008-04-02 2023-07-03 Contrast agents, methods for preparing contrast agents, and methods of imaging

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/910,893 Active 2029-04-19 US10849993B2 (en) 2008-04-02 2018-03-02 Contrast agents, methods for preparing contrast agents, and methods of imaging
US17/068,215 Active 2030-01-30 US11738098B2 (en) 2008-04-02 2020-10-12 Contrast agents, methods for preparing contrast agents, and methods of imaging
US18/346,503 Pending US20240082434A1 (en) 2008-04-02 2023-07-03 Contrast agents, methods for preparing contrast agents, and methods of imaging

Country Status (3)

Country Link
US (4) US20110097742A1 (en)
EP (2) EP3498307A1 (en)
WO (1) WO2009146099A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8252595B2 (en) 2008-05-13 2012-08-28 University Of Kansas Metal abstraction peptide (MAP) tag and associated methods
US9187735B2 (en) 2012-06-01 2015-11-17 University Of Kansas Metal abstraction peptide with superoxide dismutase activity

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8697648B2 (en) * 2009-10-20 2014-04-15 Georgia State University Research Foundation, Inc. Protein agent for diabetes treatment and β cell imaging
PL2593127T3 (en) * 2010-07-13 2018-11-30 Georgia State University Research Foundation Anti-angiogenic agent and method of using such agent
US20150218636A1 (en) * 2012-04-12 2015-08-06 Aarhus Universitet Mutations In Calmodulin Genes
EP2698169A1 (en) * 2012-08-13 2014-02-19 Universität Duisburg-Essen A contrast agent-binding polypeptide and use thereof
US10814020B2 (en) * 2015-05-11 2020-10-27 Georgia State University Research Foundation, Inc. Targeted protein contrast agents, methods of making, and uses thereof
JP2023542341A (en) * 2020-09-21 2023-10-06 エスアールアイ インターナショナル Targeted antigen delivery system and its use

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4508625A (en) * 1982-10-18 1985-04-02 Graham Marshall D Magnetic separation using chelated magnetic ions
US5679548A (en) * 1993-02-02 1997-10-21 The Scripps Research Institute Methods for producing polypeptide metal binding sites and compositions thereof
US5690903A (en) * 1991-06-06 1997-11-25 Hainfeld; James F. Loading and conjugating cavity biostructures
US5922302A (en) * 1993-05-17 1999-07-13 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin-metal chelating protein conjugates
US6197258B1 (en) * 1996-10-25 2001-03-06 University Of Maryland, Baltimore Photoluminescent sensors of chemical analytes
US20020015038A1 (en) * 2000-06-16 2002-02-07 Patel Nehal Manhar System and method for evaluating pockets in protein
US20020136692A1 (en) * 2001-01-05 2002-09-26 Zishan Haroon Contrast enhancement agent for magnetic resonance imaging
US20030149254A1 (en) * 1999-11-10 2003-08-07 Rigel Pharmaceuticals, Inc. Methods and compositions comprising Renilla GFP
US20030180222A1 (en) * 2001-07-30 2003-09-25 Zhaoda Zhang Peptide-based multimeric targeted contrast agents
US20040208827A1 (en) * 1995-02-01 2004-10-21 Epix Medical, Inc., A Delaware Corporation Diagnostic imaging contrast agents with extended blood retention
US20060029942A1 (en) * 2004-08-09 2006-02-09 Yang Jenny J Grafting method for constructing an analyte binding motif
US20060031020A1 (en) * 2004-08-09 2006-02-09 Yang Jenny J Computational approach for constructing an analyte binding motif
US20060030029A1 (en) * 2004-08-09 2006-02-09 Yang Jenny J Analyte sensors
US7405320B2 (en) * 1998-06-22 2008-07-29 Immunomedics, Inc. Therapeutic and diagnostic conjugates for use with multispecific antibodies

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
SE9400088D0 (en) 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
DE19724796A1 (en) * 1997-06-06 1998-12-10 Max Delbrueck Centrum Antitumor therapy agents
WO2001030398A2 (en) 1999-10-22 2001-05-03 Insight Neuroimaging Systems, Llc Ligand chelated paramagnetic mri contrast agents
US6565828B2 (en) * 2000-04-07 2003-05-20 Bristol-Myers Squibb Company Macrocyclic chelants for metallopharmaceuticals
JP2002253261A (en) 2001-03-05 2002-09-10 Inst Of Physical & Chemical Res Fluorescent protein
ITMI20011708A1 (en) 2001-08-03 2003-02-03 Bracco Imaging Spa CONJUGATES OF PEPTIDES, THEIR DERIVATIVES WITH METALLIC COMPLEXES AND USE FOR DIAGNOSTIC INVESTIGATION THROUGH IMAGING FOR MAGNETIC RESONANCE (M
IL162732A0 (en) 2001-12-26 2005-11-20 Immunomedics Inc Methods of generating multispecific, multivalent agents from hv and vl domains
US20050112064A1 (en) * 2003-08-12 2005-05-26 Sonya Franklin DNA-dependent MRI contrast agents
WO2005097208A2 (en) * 2004-03-26 2005-10-20 University Of Florida Research Foundation, Inc. Tissue oxygenation measurements
US20080305049A1 (en) 2005-01-31 2008-12-11 Hadassa Degani Mri Contrast Agents for Diagnosis and Prognosis of Tumors
US20080112891A1 (en) * 2005-02-15 2008-05-15 Dendritic Nanotechnologies, Inc. Encapsulated (Chelate or Ligand) Dendritic Polymers
EP1888125A2 (en) 2005-04-01 2008-02-20 Board of Regents of the University of Texas System Poly(peptide) as a chelator: methods of manufacture and uses
EP3378496A1 (en) * 2005-07-13 2018-09-26 Georgia State University Research Foundation, Inc. Contrast agents and methods for preparing contrast agents
WO2007030802A2 (en) * 2005-09-09 2007-03-15 Georgia State University Research Foundation, Inc. Targeted contrast agents and methods for targeting contrast agents
JP5377965B2 (en) 2005-09-09 2013-12-25 ジョージア ステート ユニバーシティ リサーチ ファウンデーション、インコーポレイテッド Targeted contrast agents and methods for targeting contrast agents

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4508625A (en) * 1982-10-18 1985-04-02 Graham Marshall D Magnetic separation using chelated magnetic ions
US5690903A (en) * 1991-06-06 1997-11-25 Hainfeld; James F. Loading and conjugating cavity biostructures
US5679548A (en) * 1993-02-02 1997-10-21 The Scripps Research Institute Methods for producing polypeptide metal binding sites and compositions thereof
US5922302A (en) * 1993-05-17 1999-07-13 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin-metal chelating protein conjugates
US20040208827A1 (en) * 1995-02-01 2004-10-21 Epix Medical, Inc., A Delaware Corporation Diagnostic imaging contrast agents with extended blood retention
US6197258B1 (en) * 1996-10-25 2001-03-06 University Of Maryland, Baltimore Photoluminescent sensors of chemical analytes
US7405320B2 (en) * 1998-06-22 2008-07-29 Immunomedics, Inc. Therapeutic and diagnostic conjugates for use with multispecific antibodies
US20030149254A1 (en) * 1999-11-10 2003-08-07 Rigel Pharmaceuticals, Inc. Methods and compositions comprising Renilla GFP
US20020015038A1 (en) * 2000-06-16 2002-02-07 Patel Nehal Manhar System and method for evaluating pockets in protein
US20020136692A1 (en) * 2001-01-05 2002-09-26 Zishan Haroon Contrast enhancement agent for magnetic resonance imaging
US20030180222A1 (en) * 2001-07-30 2003-09-25 Zhaoda Zhang Peptide-based multimeric targeted contrast agents
US20060029942A1 (en) * 2004-08-09 2006-02-09 Yang Jenny J Grafting method for constructing an analyte binding motif
US20060031020A1 (en) * 2004-08-09 2006-02-09 Yang Jenny J Computational approach for constructing an analyte binding motif
US20060030029A1 (en) * 2004-08-09 2006-02-09 Yang Jenny J Analyte sensors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Caravan et al. Gadolinium-binding helix-turn-helix peptides: DNA-dependent MRI contrast agents. Chem. Commun 2003, pp. 2574-2575. *
Patonay et al. Near-infrared fluorogenic labels: new approach to an old problem. Analytical chemistry 1991, Vol. 63, No. 6, pp. 321-327. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8252595B2 (en) 2008-05-13 2012-08-28 University Of Kansas Metal abstraction peptide (MAP) tag and associated methods
US8278111B2 (en) 2008-05-13 2012-10-02 University Of Kansas Metal abstraction peptide (MAP) tag and associated methods
US8975082B2 (en) 2008-05-13 2015-03-10 University Of Kansas Metal abstraction peptide (MAP) tag and associated methods
US9096652B2 (en) 2008-05-13 2015-08-04 University Of Kansas Metal abstraction peptide (MAP) tag and associated methods
US9187735B2 (en) 2012-06-01 2015-11-17 University Of Kansas Metal abstraction peptide with superoxide dismutase activity

Also Published As

Publication number Publication date
WO2009146099A3 (en) 2010-02-18
EP2257316A4 (en) 2014-10-22
EP3498307A1 (en) 2019-06-19
EP2257316A2 (en) 2010-12-08
WO2009146099A2 (en) 2009-12-03
US20180250426A1 (en) 2018-09-06
US20240082434A1 (en) 2024-03-14
US10849993B2 (en) 2020-12-01
EP2257316B1 (en) 2018-11-07
US11738098B2 (en) 2023-08-29
US20210038745A1 (en) 2021-02-11

Similar Documents

Publication Publication Date Title
US11738098B2 (en) Contrast agents, methods for preparing contrast agents, and methods of imaging
US9956304B2 (en) Contrast agents, methods for preparing contrast agents, and methods of imaging
US9333272B2 (en) Fibrin binding peptide conjugates for diagnostic and therapeutic applications
US10525150B2 (en) Targeted contrast agents and methods for targeting contrast agents
CN109476721B (en) CD 8-specific capture agents, compositions and methods of use and preparation
US8367040B2 (en) Contrast agents and methods for preparing contrast agents
US9200017B2 (en) Multimodal imaging of fibrin
Matharu et al. Gadolinium-complexed Aβ-binding contrast agents for MRI diagnosis of Alzheimer's Disease
US8367039B2 (en) Compounds for the diagnosis of diseases associated with VCAM expression
WO2001030398A2 (en) Ligand chelated paramagnetic mri contrast agents
US9889213B2 (en) Multimodal imaging of fibrin
US20170224833A1 (en) Self-assembling molecules that accumulate in acidic tumor microenvironments
EP1928507B1 (en) Targeted contrast agents and methods for targeting contrast agents
Salarian Development of Novel Collagen-targeted Protein-based MRI Contrast Agent for Imaging of Chronic Liver and Heart Diseases

Legal Events

Date Code Title Description
AS Assignment

Owner name: GEORGIA STATE UNIVERSITY RESEARCH FOUNDATION, GEOR

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:YANG, JENNY JIE;LIU, ZHIREN;LI, SHUNYI;AND OTHERS;SIGNING DATES FROM 20090527 TO 20101012;REEL/FRAME:025552/0418

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE