Multi DTPA conjugated tetrapyrollic compounds for phototherapeutic contrast agents

Information

  • Patent Grant
  • 7897140
  • Patent Number
    7,897,140
  • Date Filed
    Friday, June 30, 2006
    17 years ago
  • Date Issued
    Tuesday, March 1, 2011
    13 years ago
Abstract
Novel tetrapyrollic water soluble photosensitizing and imaging compounds and the methods of treating and imaging hyperproliferative tissue, e.g. tumors and hypervacularized tissue such as found in macular degeneration. Broadly, the compounds are tetrapyrollic photosensitizer compounds where the tetrapyrollic compound is a chlorin, bacteriochlorin, porphyrin, pyropheophorbide, purpurinimide, or bacteriopurpurinimide having 3 to 6 —CH2CONHphenylCH2CH(N(CH2COOH)2))(CH2N(CH2COOH)(CH2CH2N(CH2COOH)2)) groups or esters thereof or complexes thereof with gadolinium(III).
Description
BACKGROUND OF THE INVENTION

As described in above priority patent application Ser. No. 10/390,438, an effective tetrapyrollic photosensitizer, e.g. HPPH (a chlorophyll-a derivative) was conjugated with Gd(III)-aminophenyl-DTPA, an imaging agent. In vivo reflection spectroscopy confirmed that tumor uptake of the HPPH-aminophenylDTPA Gd (III) conjugate was higher than that of HPPH alone in the radiation-induced fibrosarcoma (RIF) tumor of C3H mice. The subcutaneously-implanted Ward colon carcinoma in rats showed markedly increased MRI signal at twenty-four hours after intravenous injection of the conjugate. Both in vitro (RIF tumor cells) and in vivo (mice bearing RIF tumors) the conjugate produced significant efficacy. We have synthesized a molecule [two Gd (III) atoms per HPPH molecule] that also remained tumor-avid, PDT-active, and with improved MRI enhancing ability than the related mono-Gd(III) analog. Unfortunately, at the MRI dose (10 μmole/kg), these conjugates produced severe skin phototoxicity. However, replacing the hexyl-group of the pyropheophorbide-a with a PEG group, produced remarkable tumor enhancing at 8 hour postinjection, significant tumoricidal activity. The poor water-solubility problem of these conjugates was resolved by liposomal formulation.


For many years, in vivo imaging of human organs was largely dependent upon the intravenous administration of radioactive molecules for nuclear scanning or non-radioactive iodinated chemicals for radiography. However, over the last decade magnetic resonance imaging (MRI) has assumed a critical role in imaging. Unlike nuclear scanning, conventional radiography, or even computed tomography, MRI uses contrast enhancers (“contrast media”) that contain paramagnetic ions, particularly gadolinium [Gd(III)]. They are not themselves “seen” by the MRI scanner. Rather, they affect the water in body tissue so as to increase its “signal” when placed in a magnetic field. At present, three similar gadolinium(III)-derived MRI contrast agents have been approved for human clinical use in the United States, the bis-N-methylglucamine salt of Gd(III)diethylenetriaminepentaacetic acid (DTPA) (Magnavist), the bis-N-methylamide of Gd(III) DTPA (Omniscan), and the Gd(III) chelate of 20-(2-hydroxypropyl) derivative of 1,4,7,10-tetraazacyclododecane-N,N′,N″,N′″-1,4,7-tetraacetic acid (Prohance). All three of these agents are carboxylate containing, water-soluble complexes. After intravenous injection they result in a transient signal increase in the vascular space and penetrate the “leaky” capillary bed of many tumors, but are rapidly excreted through the kidneys by glomerular filtration. Although several liver-specific contrast media have also been created, other organs have not been successfully targeted, and no specific tumor-avid MRI contrast agent is available to date.


Signal Intensity in MRI:


Signal intensity in MRI stems largely from the local value of the longitudinal and transverse relaxation rates of water protons, 1/T1, and the transverse rate, 1/T2. Signal tends to increase with increasing 1/T1 and decrease with increasing 1/T2. Pulse sequences that emphasize changes in 1/T1 are referred to as “T1-weighted,” and the opposite is true for T2-weighted scans. Contrast agents increase both 1/T1 and 1/T2 to varying degrees, depending on their nature as well as the applied magnetic field. Agents like gadolinium (III) that increase 1/T1 and 1/T2 by roughly similar amounts are best visualized using T1-weighted images, because the percentage change in 1/T1 in tissue is much greater than that in 1/T2. The longitudinal and transverse relaxivity values, r1, and r2, refer to the amount of increase in 1/T1 and 1/T2, respectively, per milimole of agent (often given as per mM of Gd). T1 agents usually have r2/r1 ratios of 1-2.


Advances in MRI have strongly favored T1 agents and thus gadolinium(III). Faster scans with higher resolution require more rapid radio-frequency pulsing and are thus generally T1-weighted, because MR signal in each voxel becomes saturated. T1 agents relieve this saturation by restoring a good part of the longitudinal magnetization between pulses. At the same time, a good T1 agent would not significantly affect the bulk magnetic susceptibility of the tissue compartment in which it is localized, thus minimizing any inhomogeneities that can lead to image artifacts and/or decreased signal intensity.


The effect of these agents is to increase signal on T1-weighted images that are negatively affected by proton density. The effect on T2-weighed images is to decrease signal, but this effect is minimal, because most of the T2 signal comes from the influence of proton density. Signal Intensity for the Spin Echo Imaging is expressed as:

S(TE,TR)=N(H)[1−2e−(TR−TE/2)/T1=e−TE/T1]e−TE/T2


Conventional clinical MRI units produce static, cross sectional images. Newer “interventional MRI” units allow the operator to continuously image an organ while performing surgery or other manipulations.


Gd(III) is a logical choice for MRI contrast media because of its superior performance compared with other lanthanide ions. Dysprosium(III) and holmium(III) have larger magnetic moments than that of Gd(III), but the asymmetry of their electronic states leads to very rapid electron spin relaxation. The symmetric S-state of Gd(III) is a more hospitable environment for electron spins, leading to a much slower electronic relaxation rate. In the process that gives rise to relaxivity, water protons hardly feel the effects of ions like Dy(III), much like a leaf near the rapid wings of hummingbird; Gd(III) electrons, on the other hand, are more closely in tune with the proton's frequency.


A key biological factor that influences the selection of gadolinium compounds for human use is that its ligands like DTPA circulate and are excreted intact. The metal ion is “buried” in the chelation cage and will not bind to donor groups of proteins and enzymes. This in vivo stability markedly reduces the potential for toxicity from free gadolinium.


Tetrapyrrole-Based Compounds as MRI Agents:


The porphyrins and related tetrapyrrolic systems are among the most widely studied of all macrocyclic compounds. In fact, in one capacity or another these versatile molecules have influenced nearly all disciplines in chemistry. The concentration of certain porphyrins and related tetrapyrrolic or expanded porphyrin-type compounds is much higher in malignant tumors than in most normal tissues. A few years ago Sessler and coworkers discovered a new class of expanded porphyrins that is based on the Schiff base condensation between a diformyl-tripyrrane and an aromatic 1,2-diamine. This new class of expanded porphyrins has come to be known as the “texaphyrins”. Compared to the natural porphyrin system, the texaphyrins possess a larger core size and thus have the capability to form complexes with certain lanthanides, including gadolinium(III). Gd(III) texaphyrin is currently under phase I/II human clinical trials as a tumor-avid MRI contrast agent.


Some tetrapyrrole-based compounds are effective photosensitizers for cancer treatment by photodynamic therapy [PDT]. Although PDT is sometimes considered a novel, idiosyncratic therapy, it has in fact been effective in a wide variety of malignancies, including skin, lung, bladder, head and neck, breast, and esophagus. The precise mechanism(s) of PDT are unknown; however, in vitro studies suggest that singlet oxygen production is phototoxic when the photosensitizing agent encounters light. In vivo animal data suggest that tumor vasonecrosis may be the direct cause of tumor kill.


Effective PDT requires delivery of light to tumor that has absorbed a photosensitizer previously delivered by the systemic circulation after peripheral intravenous injection. Superficial visible lesions, or those that are endoscopically accessible—e.g., endobronchial or esophageal—are easily treated, but the vast majority of malignant lesions are too deep to be reached by light of the wavelength required to trigger singlet oxygen production in the current generation of photosensitizers. Although the technology to deliver therapeutic light to deep lesions via thin transmission fibers “capped” by a terminal diffuser is well-developed, a deep lesion is by definition not visible from the skin surface, and its uptake of a peripherally-injected photosensitizer is unknown; therefore, PDT of deep tumors thus far been impractical.


A relatively long-wavelength absorbing photosensitizer, the 3-(1-hexyloxy)ethyl derivative of pyropheophorbide-a 1 [HPPH], developed in our laboratory, is tumor-avid and currently in Phase VIII clinical trials at The Roswell Park Cancer Institute. We investigated this compound as a “vehicle” for delivering gadolinium complexes to tumor, with the goal of creating the first single compound that would function both as an MRI tumor-avid contrast medium and a photosensitizer for cancer therapy. [Gd(III) texaphrin is not a photosensitizer, because it does not produce singlet oxygen when exposed to light].





BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS


FIG. 1A shows MRI images of tumors as compared to muscle using conjugate 3.



FIG. 1B shows MRI images of tumors as compared to muscle using conjugate 7.



FIG. 2 is a bar graph showing tumor to muscle contrast of the conjugates.



FIG. 3 is a bar graph showing tumor to fat contrast of the conjugates.





BRIEF DESCRIPTION OF THE INVENTION

The present invention deals with the synthesis of “higher pay-load’ contrast agents in which the photosensitizers are conjugated with three- and six Gd(III)DTP molecules. In contrast to conjugates with mono- and di-DTPA conjugates which were formulated in Tween 80/water and liposomal formulation, the conjugates with three- and six DTPA molecules can be formulated in phosphate buffer at 7.4 pH and show the potential for tumor imaging ability and phototoxicity. The development of a tumor-avid contrast medium for MRI would by itself represent an important step in the diagnosis of cancer, but a dual function agent presents the potential for a diagnostic body scan followed by targeted photodynamic therapy, combining two modalities into a single cost-effective “see and treat” approach.


The invention includes both the novel tetrapyrollic water soluble photosensitizing and imaging compounds and the methods of treating and imaging hyperproliferative tissue, e.g. tumors and hypervacularized tissue such as found in macular degeneration. Broadly, the compounds are tetrapyrollic photosensitizer compounds where the tetrapyrollic compound is a chlorin, bacteriochlorin, porphyrin, pyropheophorbide, purpurinimide, or bacteriopurpurinimide having 3 to 6 —CH2CONHphenylCH2CH(N(CH2COOH)2))(CH2N(CH2COOH)(CH2CH2N(CH2COOH)2)) groups or esters thereof or complexes thereof with gadolinium(III).


Preferably, the compound has at least one pendant —CH2CH2CONHC(CH2CH2CONHphenylCH2CH(N(CH2COOH)2))(CH2N(CH2COOH)(CH2CH2N(CH2COOH)2)))3 group or esters thereof or complexes thereof with gadolinium(III).


Preferred compounds of the invention have the formula:




embedded image



or a pharmaceutically acceptable derivative thereof, wherein:


R1 and R2 are each independently substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, —C(O)Ra or —COORa or —CH(CH3)(OR) or —CH(CH3)(O(CH2)nXR) where Ra is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, or substituted or unsubstituted cycloalkyl where R2 may be CH═CH2, CH(OR20)CH3, C(O)Me, C(═NR21)CH3 or CH(NHR21)CH3;


where X is an aryl or heteroaryl group;


n is an integer of 0 to 6;


R and R′ are independently H or lower alkyl of 1 through 8 carbon atoms;


where R20 is methyl, butyl, heptyl, docecyl or 3,5-bis(trifluoromethyl)-benzyl; and


R21, is 3,5,-bis(trifluoromethyl)benzyl;


R1a and R2a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;


R3 and R4 are each independently hydrogen or substituted or unsubstituted alkyl;


R3a and R4a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;


R5 is hydrogen or substituted or unsubstituted alkyl;


R6 and R6a are each independently hydrogen or substituted or unsubstituted alkyl, or together form ═O;


R7 is a covalent bond, alkylene, azaalkyl, or azaaraalkyl or ═NR20 where R20 is hydrogen or lower alkyl of 1 through 8 carbon atoms or —CH2-3,5-bis(tri-fluoromethyl)benzyl or —CH2X—R1 or —YR1 where Y is an aryl or heteroaryl group;


R8 and R8a are each independently hydrogen or substituted or unsubstituted alkyl or together form ═O;


R9 is a pendant group containing 3 through 6


—CH2CONHphenylCH2CH(N(CH2COOH)2))—(CH2N(CH2COOH)(CH2CH2N(CH2COOH)2)) groups or esters thereof or complexes thereof with gadolinium(III).


R10 is hydrogen, or substituted or unsubstituted alkyl and;


each of R1-R10, when substituted, is substituted with one or more substituents each independently selected from Q, where Q is alkyl, haloalkyl, halo, pseudohalo, or —COORb where Rb is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, araalkyl, or ORc where Rc is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl or CONRdRe where Rd and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or NRfRg where Rf and Rg are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or ═NRh where Rh is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or is an amino acid residue;


each Q is independently unsubstituted or is substituted with one or more substituents each independently selected from Q1, where Q1 is alkyl, haloalkyl, halo, pseudohalo, or —COORb where Rb is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, araalkyl, or ORc where Rc is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl or CONRdRe where Rd and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or NRfRg where Rf and Rg are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or ═NRh where Rh is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or is an amino acid residue.


R9 is preferably a —CH2CH2CONHC(CH2 CH2CONHphenylCH2CH(N(CH2COOH)2))—(CH2N(CH2COOH)(CH2CH2N(CH2COOH)2)))3 group or esters thereof or complexes thereof with gadolinium(III).


DETAILED DESCRIPTION OF THE INVENTION

A compound in accordance with the present invention that effectively functions both as an MRI contrast medium and a photosensitizer creates an entirely new paradigm for tumor diagnosis and therapy. After peripheral intravenous injection of this compound, a patient could be scanned with interventional MRI. The tumor site(s) could thus be defined, and, while the patient remains in the scanner, an interventional radiologist could transcutaneously insert ultra-slim needles acting as introducers for light-transmission fibers into the lesion(s). Because such fiber diameters can be small, e.g. only 400 microns, the introducer needles would produce negligible tissue damage. A light source can be coupled to the fibers, and PDT of the lesion(s) can commence, without any significant injury to other organs. Because the same molecule represents the contrast medium and the therapeutic medium, the lesion(s) can be continuously imaged during needle/fiber placement, without any ambiguity in location or “misregistration” of separate diagnostic/therapeutic images. This paradigm makes the low-toxicity and high efficacy of PDT available to virtually any location from the skull base to the floor of the pelvis.


Examples of compounds for use in accordance with the present invention were prepared and tested. Synthetic intermediates and the final products were characterized by NMR (400 MHz), mass spectrometry (EIMS & HRMS) and elemental analyses. 1H NMR spectra were recorded on a Bruker AM-400 spectrometer. Chemical shifts are expressed in ppm. All photo physical experiments were carried out using spectroscopic grade solvents. The reactions were monitored by TLC and/or spectrophotometrically. Column chromatography was performed either over Silica Gel 60 (70-230 mesh) or neutral Alumina (Brockmann grade III, 50 mesh). UV-visible spectrums were recorded on Varian Cary 50 Bio UV-visible spectrophotometer using dichloromethane as solvent unless other wise specified.


Compound No. 1:


HPPH (100.0 mg, 0.157 mmol), amine A (97.8 mg, 0.235 mmol), EDCI (60.2 mg, 0.314 mmol) and DMAP (38.36 mg, 0.314 mmol) were taken in a dry RBF (100 ml). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over silica gel using 1-3% Methanol/Dichloromethane mixture as eluent to give product 1. Yield: 130.0 mg (80.0%). UV-vis (λmax cm−1, dichloromethane): 318, 412, 506, 537, 604 & 660. 1HNMR (400 MHz, CDCl3): δ 9.78 (s, 1H, H-5), 9.41 (s, 1H, H-10), 8.54 (s, 1H, H-20), 5.96-5.91 (m, 2H, CH3CHOhexyl, NH), 5.33 (d, 1H, CH-151, J=19.6 Hz), 5.14 (d, 1H, CH-151, J=19.6 Hz), 4.53 (q, 1H, H-17, J=7.2 Hz), 4.32 (m, 1H, H-18), 3.71-3.67 (m, 2H, —OCH2-hexyl), 3.63-3.60 (m, 2H, 8-CH2CH3), 3.52 (s, 3H, 7-CH3), 3.39 (s, 3H, 2-CH3), 3.27 (s, 3H, 12-CH3), 2.67 (m, 1H, CH-172), 2.37 (m, 1H, H-172), 2.26 (m, 1H, H-171), 2.15-2.13 (m, 9H, 3CH2-chain & CH3CHOhexyl), 1.97 (m, 1H, H-171), 1.92 (t, 6H, 3CH2-chain, J=7.6 Hz), 1.80 (d, 3H, 18-CH3, J=7.2 Hz), 1.73 (m, 2H, CH2hexyl), 1.66 (t, 3H, 8-CH2CH3, J=7.6 Hz), 1.44 (m, 2H, CH2hexyl), 1.30 (s, 27H, 3CO2tBu), 1.24 (m, 4H, 2CH2hexyl), 0.78 (t, 3H, CH3hexyl, J=6.8 Hz), 0.04 (brs, 1H, NH), −1.68 (brs, 1H, NH).


EIMS: 1035 (MH+).


Compound No. 2:


Compound 1 (73.0 mg, 0.07 mmol) was stirred with 70% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. To this crude were added, amino-benzyl-DTPA-penta-tert-butyl ester (219.0 mg, 0.282 mmol), EDCI (67.0 mg, 0.352 mmol) and DMAP (43.0 mg, 0.352 mmol). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over alumina G (III) using 1-3% Methanol/Dichloromethane mixture as eluent to give product 2. Yield: 130.0 mg (58.47%). UV-vis (λmax cm−1, dichloromethane): 319, 411, 506, 537, 604, 661. 1HNMR (400 MHz, CDCl3): δ 9.74 (splitted s, 1H, H-5), 9.36 (splitted s, 1H, H-10), 8.54 (splitted s, 1H, H-20), 8.17 (brs, 1H, NH), 7.74 (m, 1H, Ph-DTPA), 7.61-7.56 (m, 2H, Ph-DTPA), 7.43 (m, 4H, Ph-DTPA), 7.12 (m, 5H, Ph-DTPA), 6.96 (brs, 1H, NH), 5.96 (m, 1H, CH3CHOhexyl), 5.29 (d, 1H, CH-151, J=19.2 Hz), 5.11 (d, 1H, CH-151, J=19.2 Hz), 4.47 (m, 1H, H-17), 4.26 (m, 1H, H-18), 3.64 (m, 2H, & OCH2hexyl), 3.54 (m, 2H, 8-CH2CH3), 3.45-3.38 (m, 30H, 15CH2-DTPA), 3.36 (s, 3H, 7-CH3), 3.32 (s, 3H, 2-CH3), 3.22 (s, 3H, 12-CH3), 3.12 (m, 3H, CH-DTPA), 2.84-2.70 (m, 19H, 9CH2-DTPA & CH-172), 2.59 (m, 6H, 6CH2-benzyl), 2.47-2.41 (m, 8H, 3CH2-chain, CH-172 & CH-171), 2.19-2.15 (m, 9H, 3CH2-chain, CH3CH-Ohexyl), 2.06 (d, 3H, 18-CH3, J=7.6 Hz) 2.00 (m, 1H, CH-171), 1.74 (m, 4H, 2CH2-hexyl), 1.66 (t, 3H, 8-CH2CH3, J=7.2 Hz), 1.60 (m, 135H for 15 CO2tBu), 1.26 (m, 4H, 2CH2-hexyl), 0.77 (m, 3H, CH3-Ohexyl), 0.55 (brs, 1H, NH), −0.24 (brs, 1H, NH). HRMS: Calculated for C172H267N17O36: 3149.053, found: 3150.10 (MH+).


Compound No. 3:


Compound 2 (110.0 mg, 0.034 mmol) was stirred with 70% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. The crude thus obtained was dissolved in pyridine (10 ml) and put under stirring, to this stirring solution GdCl3.6H2O (77.9 mg, 0.21 mmol) in 1 ml of water was added slowly and resultant mixture was stirred for 16 hr. Reaction mixture was concentrated to dryness under high vacuum. Residue was washed with water (10 ml×3), acetone (10 ml×3) and finally dried under high vacuum using P2O5 as drying agent. Yield: 75.0 mg (77.55%). UV-vis (λmax cm−1, MeOH): 620, 408, 504, 537, 604 & 660. Elemental analysis: Calculated for C113H151Gd3N17O36: C, 48.55; H, 5.44; Gd, 16.88; N, 8.52; O, 20.61. found: C, 48.68; H, 5.49; N, 8.57.


Compound No. 5:


Acid 4 (100.0 mg, 0.143 mmol), amine A (89.1 mg, 0.214 mmol), EDCI (54.9 mg, 0.28 mmol) and DMAP (34.9 mg, 0.28 mmol) were taken in a dry RBF (100 ml). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over silica gel using 1-2% Methanol/Dichloromethane mixture as eluent to give product 5. Yield: 134.0 mg (85.3%). UV-vis (λmax cm−1, dichloromethane): 318, 411, 506, 536, 604 & 661. 1HNMR (400 MHz, CDCl3): δ 9.76 (splitted s, 1H, H-5), 9.38 (splitted s, 1H, H-10), 8.55 (s, 1H, H-20), 6.02 (d, 1H, CH3CHOOTEG, J=6.4 Hz), 5.99 (brs, 1H, NH), 5.34 (d, 1H, CH-151, J=20.0 Hz), 5.15 (d, 1H, CH-151, J=20.0 Hz), 4.58 (q, 1H, H-17, J=6.8 Hz), 4.33 (m, 1H, H-18), 3.88-3.75 (m, 4H, 2CH2—O-TEG), 3.70-3.62 (m, 6H, 3CH2—O-TEG), 3.55 (m, 2H, 8-CH2CH3), 3.47-3.44 (m, 2H, CH2—O-TEG), 3.40 (s, 3H, 7-CH3), 3.39 (s, 3H, 2-CH3), 3.29 (s, 3H, 12-CH3), 3.27 (s, 3H, CH3—O-TEG), 2.69 (m, 1H, CH-172), 2.39 (m, 1H, CH-172), 2.31 (m, 1H, CH-171), 2.14 (m, 8H, 4CH2-chain), 2.00 (m, 1H, CH-171), 1.92 (m, 7H, 2CH2-chain, CH3CHOTEG), 1.82 (d, 3H, 18-CH3, J=7.2 Hz), 1.68 (t, 3H, 8-CH2CH3, J=7.6 Hz), 1.31 (s, 27H, 3CO2tBu), 0.42 (brs, 1H, NH), −1.69 (brs, 1H, NH). EIMS: 1097 (MH+)


Compound No. 6:


Compound 5 (100.0 mg, 0.091 mmol) was stirred with 80% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. To this crude were added, amino-benzyl-DTPA-penta-tert-butyl ester (285.0 mg, 0.365 mmol), EDCI (105.0 mg, 0.54 mmol) and DMAP (66.8 mg, 0.54 mmol). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over alumina G (III) using 1-3% Methanol/Dichloromethane mixture as eluent to give product 6. Yield: 250.0 mg (85.3%). UV-vis (λmax cm−1, dichloromethane): 319, 411, 506, 537, 606, 661. 1HNMR (400 MHz, CDCl3): δ 9.70 (splitted s, 1H, H-5), 9.44 (splitted s, 1H, H-10), 9.37 (brs, 1H, NH), 9.20 (brs, 1H, NH), 8.56 (m, 1H, NH), 8.47 (s, 1H, H-20), 7.77 (m, 1H, Ph-DTPA), 7.59 (m, 2H, Ph-DTPA), 7.44 (m, 2H, Ph-DTPA), 7.10 (m, 6H, Ph-DTPA), 6.81 (m, 1H, Ph-DTPA), 5.97 (m, 1H, CH3CHOTEG), 5.20 (m, 2H, CH2-151), 4.60 (m, 1H, H-17), 4.22 (m, 1H, H-18), 3.81-3.64 (m, 4H, 2CH2—OTEG), 3.57-3.50 (m, 4H, 8-CH2CH3, CH2—OTEG), 3.60 (m, 6H, 3CH2OTEG), 3.38 (s, 30H, 15CH2-DTPA), 3.35 (s, 3H, 7-CH3), 3.23 (m, 6H, 12-CH3, OCH3-TEG), 3.04 (m, 3H, 3CH-DTPA), 2.70 (m, 19H, 9CH2-DTPA, CH-172), 2.55 (m, 7H, 3CH2-benzyl & CH-172), 2.32 (t, 6H, 3CH2-chain, J=6.8 Hz), 2.23 (m, 1H, CH-171), 2.10 (d, 3H, CH3CH-OTEG, J=6.4 Hz), 2.01 (m, 1H, CH-171), 1.77 (d, 3H, 18-CH3, J=7.2 Hz), 1.68 (t, 3H, 8-CH2CH3, J=7.6 Hz), 1.59 (t, 6H, 3CH2-chain, J=6.4 Hz), 1.44 (m, 135H, 15CO2tBu), −1.75 (brs, 1H, NH).


HRMS: Calculated for C173H269N17O39: 3211.077, found: 3212.20 (MH+).


Compound No. 7:


Compound 6 (226.0 mg, 0.07 mmol) was stirred with 80% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. The crude thus obtained was dissolved in pyridine (10 ml) and put under stirring, to this stirring solution GdCl3.6H2O (156.9 mg, 0.422 mmol) in 1 ml of water was added slowly and resultant mixture was stirred for 16 hr. Reaction mixture was concentrated to dryness under high vacuum. Residue was washed with water (10 ml×3), acetone (10 ml×3) and finally dried under high vacuum using P2O5 as drying agent. Yield: 165.0 mg (82.5%). UV-vis (λmax cm−1, MeOH): 320, 408, 505, 537, 605 & 660. Elemental analysis: Calculated for C113H149Gd3N17O39: C, 47.77; H, 5.29; Gd, 16.60; N, 8.38; O, 21.96. found: C, 47.85; H, 5.30; N, 8.43.


Compound No. 9:


Acid 8 (82.0 mg, 0.118 mmol), amine A (73.6 mg, 0.177 mmol), EDCI (45.3 mg, 0.236 mmol) and DMAP (28.8 mg, 0.236 mmol) were taken in a dry RBF (100 ml). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over silica gel using 1-2% Methanol/Dichloromethane mixture as eluent to give product 9. Yield: 90.0 mg (69.8%). UV-vis (λmax cm−1, dichloromethane): 365, 418, 509, 545 & 699. 1HNMR (400 MHz, CDCl3): δ 9.75 (splitted s, 1H, meso-H), 9.64 (s, 1H, meso-H), 8.55 (s, 1H, meso-H), 6.25 (m, 1H, CONH), 5.79 (q, 1H, CH3CHObutyl, J=6.4 Hz), 5.34 (m, 1H, H-17), 4.52 (t, 2H, —NCH2butyl, J=7.2 Hz), 4.42 (m, 1H, H-18), 3.84 (s, 3H, ring-CH3), 3.70-3.59 (m, 4H, —OCH2-butyl, 8-CH2CH3), 3.32 (s, 3H, ring-CH3), 3.17 (s, 3H, ring-CH3), 2.61 (m, 1H, CH-172), 2.43 (m, 1H, H-172), 2.27 (m, 1H, H-171), 2.20 (t, 6H, 3CH2-chain, J=7.2 Hz), 2.06 (m, 3H, CH3CHObutyl), 2.01 (t, 6H, 3CH2-chain, J=7.6 Hz), 1.82 (m, 1H, H-171), 1.75 (d, 3H, 18-CH3, J=6.0 Hz), 1.68 (t, 3H, 8-CH2CH3, J=7.6 Hz), 1.62 (m, 8H, 4CH2butyl), 1.34 (s, 27H, 3CO2tBu), 1.10 (t, 3H, CH3-obutyl, J=7.2 Hz), 0.87 (t, 3H, CH3-Nbutyl, J=7.2 Hz), 0.40 (brs, 1H, NH), −0.06 (brs, 1H, NH). EIMS: 1092 (MH+).


Compound No. 10:


Compound 9 (80.0 mg, 0.073 mmol) was stirred with 70% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. To this crude were added, amino-benzyl-DTPA-penta-tert-butyl ester (286.0 mg, 0.366 mmol), EDCI (84.4 mg, 0.44 mmol) and DMAP (53.7 mg, 0.44 mmol). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over alumina G (III) using 1-3% Methanol/Dichloromethane mixture as eluent to give product 10. Yield: 140.0 mg (59.57%). UV-vis (λmax cm−1, dichloromethane): 365, 418, 509, 546, 699. 1HNMR (400 MHz, CDCl3): δ 9.75 (splitted s, 1H, meso-H), 9.63 (splitted s, 1H, meso-H), 9.33 (brs, 1H, NH), 8.60 (splitted s, 1H, meso-H), 7.61 (m, 2H, Ph-DTPA), 7.58 (m, 1H, Ph-DTPA), 7.37 (m, 3H, Ph-DTPA), 7.12 (m, 5H, Ph-DTPA), 6.84 (m, 1H, Ph-DTPA), 5.76 (m, 1H, CH3CHObutyl), 5.39 (m, 1H, H-17), 4.45 (m, 3H, H-18 & NCH2butyl), 3.82 (s, 3H, ring-CH3), 3.65 (m, 4H, 8-CH2CH3 & OCH2butyl), 3.38 (m, 22H, 11CH2-DTPA), 3.31 (m, 11H, 4CH2-DTPA & ring-CH3), 3.17 (s, 3H, ring-CH3), 3.03 (m, 3H, CH-DTPA), 2.84-2.61 (m, 19H, 9CH2-DTPA & CH-172), 2.47 (m, 8H, 6CH2-benzyl & CH-172 & CH-171), 2.20 (m, 6H, 3CH2-chain), 2.04 (d, CH3CHObutyl, J=6.8 Hz), 1.96 (m, 6H, 3CH2-chain), 1.84 (m, 1H, CH-171), 1.73 (s, 3H, 17-CH3), 1.67 (t, 3H, 8-CH2CH3, J=7.2 Hz), 1.60 (m, 4H, 2CH2—Obutyl), 1.41 (m, 135H for 15 CO2tBu), 1.37 (m, 4H, 2CH2—N-butyl), 1.03 (t, 3H, CH3—Obutyl, J=6.8 Hz), 0.86 (t, 3H, CH3—N-butyl, J=6.8 Hz), −0.09 (brs, 1H, NH). HRMS: Calculated for C174H270N18O37: 3206.104, found: 3207.250 (MH+).


Compound No. 11:


Compound 10 (130.0 mg, 0.04 mmol) was stirred with 70% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. The crude thus obtained was dissolved in pyridine (10 ml) and put under stirring, to this stirring solution GdCl3.6H2O (90.3 mg, 0.243 mmol) in 1 ml of water was added slowly and resultant mixture was stirred for 16 hr. Reaction mixture was concentrated to dryness under high vacuum. Residue was washed with water (10 ml×3), acetone (10 ml×3) and finally dried under high vacuum using P2O5 as drying agent. Yield: 80.0 mg (69.9%). UV-vis (λmax cm−1, MeOH): 364, 415, 546 & 700. Elemental analysis: Calculated for C114H150Gd3N18O37: C, 48.28; H, 5.33; Gd, 16.63; N, 8.89; O, 20.87. found: C, 48.14; H, 5.40; N, 8.93.


Compound No. 12:


HPPH (100.0 mg, 0.157 mmol), Di-tert-butyl iminodiacetate (77.0 mg, 0.314 mmol), EDCI (60.2 mg, 0.314 mmol) and DMAP (38.36 mg, 0.314 mmol) were taken in a dry RBF (100 ml). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over silica gel using 1-1.5% Methanol/Dichloromethane mixture as eluent to give product 12. Yield: 120.0 mg (88.3%). UV-vis (λmax cm−1, dichloromethane): 317, 411, 506, 538, 605 & 660. 1HNMR (400 MHz, CDCl3): δ 9.80 (s, 1H, H-5), 9.51 (s, 1H, H-10), 8.55 (s, 1H, H-20), 5.94 (q, 1H, CH3CHOhexyl, J=6.4 Hz), 5.33 (d, 1H, CH-151, J=20.0 Hz), 5.17 (d, 1H, CH-151, J=20.0 Hz), 4.52 (q, 1H, H-17, J=7.6 Hz), 4.41 (m, 1H, H-18), 4.04 (m, 2H, CH2chain), 3.75 (m, 2H, —OCH2-hexyl), 3.67 (s, 3H, 7-CH3), 3.62 (m, 2H, 8-CH2CH3), 3.42 (s, 3H, 2-CH3), 3.37 (m, 2H, CH2chain), 3.29 (s, 3H, 12-CH3), 2.77 (m, 1H, CH-172), 2.46 (m, 1H, CH-172), 2.16 (m, 1H, CH-171), 2.13 (m, 3H, & CH3CHOhexyl), 1.97 (m, 1H, CH-171), 1.84 (d, 3H, 18-CH3, J=7.2 Hz), 1.78 (m, 2H, CH2hexyl), 1.72 (t, 3H, 8-CH2CH3J=7.6 Hz), 1.49 (s, 9H, CO2tBu), 1.46 (m, 2H, CH2hexyl), 1.45 (s, 9H, CO2tBu), 1.25 (m, 4H, 2CH2hexyl), 0.8 (t, 3H, CH3hexyl, J=6.8 Hz), 0.42 (brs, 1H, NH), −1.7 (brs, 1H, NH). EIMS: 865 (MH+).


Compound No. 13:


Compound 12 (120.0 mg, 0.139 mmol) was stirred with 70% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. To this crude were added, amine A (144.2 mg, 0.34 mmol), EDCI (106.6 mg, 0.556 mmol) and DMAP (67.8 mg, 0.556 mmol). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over silica gel using 2-6% Methanol/Dichloromethane mixture as eluent to give product 13. Yield: 190.0 mg (88.3%). UV-vis (λmax cm−1, dichloromethane): 318, 411, 506, 537, 605 and 660. 1HNMR (400 MHz, CDCl3): δ 9.78 (s, 1H, H-5), 9.52 (s, 1H, H-10), 8.3 (s, 1H, H-20), 8.27 (brs, 1H, NH), 8.17 (brs, 1H, NH), 6.59 (brs, 1H, NH), 5.90 (m, 1H, CH3CHOhexyl), 5.34 (d, 1H, CH-151, J=20.0 Hz), 5.15 (d, 1H, CH-151, J=20.0 Hz), 4.53 (q, 1H, H-17, J=6.0 Hz), 4.36 (m, 1H, H-18), 3.77 (m, 2H, —OCH2-hexyl), 3.69 (m, 2H, 8-CH2CH3), 3.67 (s, 3H, 7-CH3), 3.61 (m, 4H, 2CH2chain), 3.36 (s, 3H, 2-CH3), 3.26 (s, 3H, 12-CH3), 2.77 (m, 1H, CH-172), 2.66 (m, 1H, CH-172), 2.52 (m, 1H, CH-171), 2.23 (m, 12H, 6CH2-chain), 2.11 (d, 3H, CH3CHOhexyl, J=6.4 Hz), 2.07 (m, 6H, 3CH2-chain), 1.95 (m, 6H, 3CH2-chain), 1.93 (m, 1H, CH-171), 1.81 (d, 3H, 18-CH3, J=7.2 Hz), 1.75 (m, 2H, CH2hexyl), 1.71 (t, 3H, 8-CH2CH3, J=8.0 Hz), 1.43 (m, 2H, CH2hexyl), 1.41 (s, 27H, 3CO2tBu), 1.32 (s, 27H, 3CO2tBu), 1.24 (m, 4H, 2CH2hexyl), 0.77 (t, 3H, CH3hexyl, J=6.8 Hz). EIMS: 1548 (MH+).


Compound No. 14:


Compound 13 (100.0 mg, 0.064 mmol) was stirred with 80% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. To this crude were added, amino-benzyl-DTPA-penta-tert-butyl ester (503.5 mg, 0.64 mmol), EDCI (123.9 mg, 0.64 mmol) and DMAP (78.8 mg, 0.64 mmol). Dry N, N-dimethylformamide (15 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was concentrated under vacuum, added dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over alumina G (III) using 1-3% Methanol/Dichloromethane mixture as eluent to give product 14. Yield: 250.0 mg (70.0%). UV-vis (λmax cm−1, Dichloromethane): 318, 413, 507, 539, 606 & 660. Elemental analysis: Calculated for C309H491N31O71: C, 64.25; H, 8.57; N, 7.52; O, 19.67. found: C, 64.30; H, 8.59; N, 7.56.


Compound No. 15:


Compound 14 (225.0 mg, 0.038 mmol) was stirred with 70% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. The crude thus obtained was dissolved in pyridine (10 ml) and put under stirring, to this stirring solution GdCl3.6H2O (173.7 mg, 0.46 mmol) in 1 ml of water was added slowly and resultant mixture was stirred for 16 hr. Reaction mixture was concentrated to dryness under high vacuum. Residue was washed with water (10 ml×3), acetone (10 ml×3) and finally dried under high vacuum using P2O5 as drying agent. Yield: 170.0 mg (86.7%). UV-vis (λmax cm−1, MeOH): 320, 411, 507, 539, 606 & 660. Elemental analysis: Calculated for C189H251, Gd6N31O71: C, 45.07; H, 5.02; Gd, 18.73; N, 8.62; O, 22.55. found: C, 45.15; H, 5.10; N, 8.58.


Compound No. 16:


Acid 4 (150.0 mg, 0.214 mmol), Di-tert-butyl iminodiacetate (105.0 mg, 0.429 mmol), EDCI (82.3 mg, 0.429 mmol) and DMAP (52.0 mg, 0.429 mmol) were taken in a dry RBF (100 ml). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over silica gel using 1-1.5% Methanol/Dichloromethane mixture as eluent to give product 16. Yield: 165.0 mg (82.9%). UV-vis (λmax cm−1, dichloromethane): 318, 411, 506, 536, 605 and 661. 1HNMR (400 MHz, CDCl3): δ 9.75 (splitted s, 1H, H-5), 9.52 (splitted s, 1H, H-10), 8.53 (s, 1H, H-20), 6.01 (m, 1H, CH3CHOTEG), 5.31 (d, 1H, CH-151, J=20.0 Hz), 5.13 (d, 1H, CH-151, J=20.0 Hz), 4.50 (q, 1H, H-17, J=7.2 Hz), 4.36 (m, 1H, H-18), 4.02 9m, 2H, CH2chain), 3.85 (m, 2H, CH2—O-TEG), 3.79 (m, 2H, CH2—O-TEG), 3.73 (m, 4H, 3CH2—O-TEG), 3.68 (s, 3H, 7-CH3), 3.66 (m, 2H, 8-CH2CH3), 3.55 (m, 2H, CH2—O-TEG), 3.39 (s, 3H, 2-CH3), 3.27 (s, 6H, 12-CH3 & OCH3-TEG), 2.75 (m, 1H, CH-172), 2.44 (m, 1H, CH-172), 2.41 (m, 1H, CH-171), 2.16 (m, 1H, CH-171), 2.14 (d, 3H, CH3CHOTEG, J=6.4 Hz), 1.81 (d, 3H, 18-CH3, J=7.6 Hz), 1.71 (t, 3H, 8-CH2CH3, J=7.6 Hz), 1.44 (splitted s, 9H, CO2tBu), 1.06 (splitted s, 9H, CO2tBu), 0.39 (brs, 1H, NH), −1.80 (brs, 1H, NH). EIMS: 927 (MH+).


Compound No. 17:


Compound 16 (140.0 mg, 0.151 mmol) was stirred with 70% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. To this crude were added, amine A (188.2 mg, 0.453 mmol), EDCI (115.9 mg, 0.604 mmol) and DMAP (73.7 mg, 0.604 mmol). Dry dichloromethane (30 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was diluted with dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over silica gel using 3-7% Methanol/Dichloromethane mixture as eluent to give product 17. Yield: 210.0 mg (86.4%). UV-vis (λmax cm−1, dichloromethane): 318, 411, 506, 536, 604 & 661. 1HNMR (400 MHz, CDCl3): δ 9.76 (splitted s, 1H, H-5), 9.53 (s, 1H, H-10), 8.63 (s, 1H, H-20), 8.31 (splitted s, 1H, CONH), 6.56 (splitted s, 1H, CONH), 6.00 (m, 1H, CH3CHOTEG), 5.35 (d, 1H, CH-151, J=20.0 Hz), 5.16 (d, 1H, CH-151, J=20.0 Hz), 4.53 (q, 1H, H-17, J=7.6 Hz), 4.36 (d, 1H, H-18, J=10.4 Hz), 3.85-3.80 (m, 4H, 2CH2—O-TEG), 3.74-3.71 (m, 6H, 2CH2chain, CH2—O-TEG), 3.67 (s, 3H, 7-CH3), 3.66 (m, 4H, 8-CH2CH3, CH2—O-TEG), 3.53 (m, 2H, CH2—O-TEG), 3.42-3.39 (m, 5H, CH2—O-TEG, 2-CH3), 3.27-3.26 (m, 6H, 12-CH3, OCH3TEG), 2.75 (m, 1H, CH-172), 2.67 (m, 1H, CH-172), 2.52 (m, 1H, CH-171), 2.24-2.22 (m, 13H, 6CH2-chain, CH-171), 2.14 (d, 3H, CH3CHOTEG, J=6.8 Hz), 2.09-2.04 (m, 6H, 3CH2-chain), 1.96 (m, 6H, 3CH2-chain), 1.81 (d, 3H, 18-CH3, J=7.2 Hz), 1.71 (t, 3H, 8-CH2CH3, J=8.0 Hz), 1.41 (s, 27H, 3CO2tBu), 1.33 (s, 27H, 3CO2tBu), 0.39 (brs, 1H, NH), −1.79 (brs, 1H, NH). EIMS: 1610 (MH+).


Compound No. 18:


Compound 17 (100.0 mg, 0.06 mmol) was stirred with 80% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. To this crude were added, amino-benzyl-DTPA-penta-tert-butyl ester (471.0 mg, 0.60 mmol), EDCI (115.9 mg, 0.60 mmol) and DMAP (73.8 mg, 0.60 mmol). Dry N, N-dimethylformamide (15 ml) was added to it and reaction mixture was stirred at RT for 16 hr under N2 atm. Reaction mixture was concentrated under vacuum, added dichloromethane (100 ml), washed with brine solution, organic layer separated, dried over sodium sulfate and concentrated. Crude mixture was chromatographed over alumina G (III) using 1-3% Methanol/Dichloromethane mixture as eluent to give product 18. Yield: 250.0 mg (69.0%). UV-vis (λmax cm−1, Dichloromethane): 319, 412, 508, 538, 606 & 661. Elemental analysis: Calculated for C309H491N31O74: C, 63.72; H, 8.50; N, 7.46; O, 20.33. found: C, 63.67; H, 8.57; N, 7.46.


Compound No. 19:


Compound 18 (215.0 mg, 0.0369 mmol) was stirred with 80% TFA/DCM (5.0 ml) at RT for 3 hr. Resultant mixture was concentrated and dried under high vacuum to remove trace of TFA. The crude thus obtained was dissolved in pyridine (10 ml) and put under stirring, to this stirring solution GdCl3.6H2O (164.6 mg, 0.44 mmol) in 1 ml of water was added slowly and resultant mixture was stirred for 16 hr. Reaction mixture was concentrated to dryness under high vacuum. Residue was washed with water (10 ml×3), acetone (10 ml×3) and finally dried under high vacuum using P2O5 as drying agent. Yield: 160.0 mg (85.0%). UV-vis (λmax cm−1, MeOH): 320, 410, 507, 539, 606 & 661. Elemental analysis: Calculated for C190H253Gd6N31O74: C, 44.76; H, 5.00; Gd, 18.50; N, 8.52; O, 23.22. found: C, 44.80; H, 5.07; N, 8.51.




embedded image


embedded image




embedded image


embedded image




embedded image


embedded image




embedded image


embedded image


embedded image




embedded image


embedded image


embedded image




embedded image


embedded image


embedded image


embedded image


Relaxivity measurements for examples of the compounds as presented herein were acquired on a General Electric 4.7T/33 cm horizontal bore magnet (GE NMR instruments, Fremont, Calif.) incorporating AVANCE digital electronics (Bruker BioSpec platform with ParaVision version 3.0.2 acquisition software, Bruker Medical, Billerica, Mass.).


T1 relaxation rates (R1) were acquired for a range of contrast agent concentrations (0.02 mM to 0.10 mM) with a saturation recovery spin-echo (SE) sequence with a fixed TE=10 ms and TR times ranging from 75 to 8000 ms. Additional MR acquisition parameters are as follows: (FOV) 32×32 mm, slice thickness=1 mm, slices=3, interslice gap=2 mm, matrix=192×192, NEX=1. Signal intensities at each repetition time was obtained by taking the mean intensity within regions of interest (ROI's) using Analyze 5.0 (Biomedical Imaging Resource, Mayo Foundation, Rochester, Minn.), and R1 and SMAX were determined by nonlinear fitting of the equation: S(TR)=SMAX(1−e−(R1*TR))+Background Noise using Matlab's Curve Fitting Toolbox (Matlab 7.0, MathWorks Inc., Natick, Mass.). The T1 relaxivity was then determined by obtaining the slope of concentration vs. R1 via linear regression fitting. Similarly, T2 relaxation rates (R2) were acquired with multi-echo, CPMG SE sequence with a fixed TR of 2500 ms and TE times ranging from 15 to 300 ms, and the number of averages=2. R2 and SMAX were determined as described above using the equation: S(TE)=SMAX(e−(R2*TE))+Background Noise. As before, the T2 relaxivity was then determined by obtaining the slope of concentration vs. R2 via linear regression fitting.


Results are shown in Table 1


Results:









TABLE 1







T1/T2 relaxivity of photosensitizer-Gd(III)DTPA conjugates










T1 Relaxivity
T2 Relaxivity


Compound
(mM · s)−1
(mM · s)−1













593 (HPPH-3Gd)
3
23.9
62.81


601 (HPPH-6Gd)
15
24.72
66.54


604 (PP-Dibutyl-3Gd)
11
20.11
84.58


611 (Pyro-OTEG-3Gd)
7
13.58
40.11


612 (Pyro-OTEG-6Gd)
19
25.09
69.19









On a T1-weighted scan and at low intratumoral concentrations of the agents (<0.1 mM), shortened T1 times will dominate the effect on signal intensity. All five of the compounds exhibit much higher T1 relaxivity values (Table 1) than conventional MR contrast-enhancing agents, which have T1 relaxivities˜3 to 4 (mM·s)−1. The increased relaxivities of our compounds allow for reduced doses with similar enhancement when compared to conventional compounds. These compounds were tested in vivo (next section) a injection doses of 10 μmoles/kg vs. 100 μmoles/kg prescribed for conventional MR contrast agents.


Examples of compounds of the invention were tested in vivo. Baseline MR images were acquired prior to injection of the compounds to serve as a baseline comparison. Animals were then re-scanned 8 and 24 hours after injection. Two spin-echo imaging protocols were used, all utilizing the same geometry (5-6 axial slices, 1.5 mm slice thickness, 6×6 cm FOV). The first scanning protocol was a moderately T1-weighted scan acquired with a TE/TR of 10/1200 ms. The second protocol was a heavily T1-weighted scan acquired with Chemical Shift Selective (CHESS) fat suppression with a TE/TR of 10/356 ms.


Regions of interest (ROI's) for tumor, muscle, fat, and noise were defined, and the mean intensity and standard deviation for each ROI was sampled. Tumor conspicuity was measured by determining the contrast-to-noise ratios (CNR's), which is defined as the difference in signal between two tissues divided by the standard deviation of the noise. Enhancement of tumor as compared to muscle was determined using the first scanning protocol (TE/TR=10.3/1200 ms). Due to the inherent hyperintensity of fat on T1-weighted MR scans, enhancement of the tumor as compared to fat was determined by analyzing the fat-suppressed images. Results are outlined in Tables 2 & 3, and sample images show in FIGS. 1A and 1B.









TABLE 2







Tumor to fat contrast/noise ratio of the Photosensitizer-Gd(III) conjugates


Tumor to Fat Contrast to Noise Ratio (CNR)










CNR












Base-

CNR Improvement














Compound
line
4 hr
8 hr
24 hr
4 hr
8 hr
24 hr

















593 (HPPH-3Gd)-3
10.42
xx
15.88
19.34
xx
54%
86%


601 (HPPH-6Gd)-15
15.61
xx
23.80
22.66
xx
52%
45%


604
5.36
xx
15.78
17.84
xx
194%
233%


(PP-Dibutyl-3Gd)-11


611
6.20
xx
20.30
11.50
xx
227%
85%


(Pyro-OTEG-3Gd)-7


612
9.10
18.60
13.40
11.20
104%
47%
23%


(Pyro-OTEG-6Gd)-19
















TABLE 3







Tumor to muscle contrast/noise ratio of the


photosensitizer Gd(III) conjugates


Tumor to Muscle Contrast to Noise Ratio (CNR)










CNR
CNR Improvement














Compound
Baseline
4 hr
8 hr
24 hr
4 hr
8 hr
24 hr

















593-3
8.69
xx
15.31
19.17
xx
66%
96%


601-15
6.36
xx
13.40
18.65
xx
111%
193%


604-11
9.93
xx
10.40
20.48
xx
5%
106%


611-7
9.39
20.00
21.30
14.40
113%
127%
53%


612-19
11.50
16.30
20.90
14.20
 42%
82%
23%









Additionally, tumor avidity of each formulation was investigated by comparing the increase in contrast, i.e. difference in normalized signal, between tumor and normal tissues. The HPPH-Gd compounds demonstrated a much higher contrast between tumor and normal tissue than the Pyro-OTEG compounds shown in FIGS. 2 & 3. The PP-Dibutyl compound showed peak contrast at 24 hours. In the Figures, compounds coded as 593=3; 601=15; 604=11, 611=7 and 612=19. FIGS. 2 and 3 show tumor to muscle and tumor to fat contrast of the conjugates.


The relatively low increase in contrast between tumors and normal tissues with the Pyro-OTEG compounds is indicative that the increase in the CNR's is predominately a global increase of signal-to-noise in the images than a preferential accumulation of the compound within the tumor.


HPPH-formulations (593 (3) and 601 (15)) showed a continuation of increasing CNR of tumor to fat and muscle from 8 hours to 24 hours. This is indicative of a longer circulation time of these compounds as compared to the Pyro-OTEG from. Conversely, the Pyro-OTEG formulations (611 (7) & 612 (19)) also showed large increases in CNR at 4 and 8 hours, but then decreased 24 hours later. This is indicative of a much shorter circulation time, which may limit efficacy of PDT treatments if not performed within 8 hours of the administration of the agent. Furthermore, the relative increase in contrast with the Pyro-OTEG compounds [611 (7) & 612 (19)] was much lower than that of the HPPH and PP-Dibutyl compounds (593 (3), 601 (15) & 604 (11)), which is exhibited in FIGS. 2 and 3. The large increases in contrast-to-noise with these compounds is attributed more to a global increase in signal to noise (due to the presence of the agents) than tumor avidity. In “real world” application, an increase in CNR is most effective when the CNR approaches a threshold of detection, which is within the range of CNR=2 to 5 for humans. Due to the bi-functionality of these agents, serving as both PDT agents as well as MR contrast-enhancing agents, the greater tumor avidity of compounds 593 (3), 601 (15), & 604 (11) would be preferred over the Pyro-OTEG compounds so that the effect of the PDT therapy is more specific to the tumor tissue rather than host tissue. However, it is foreseeable that the more rapidly clearing Pyro-OTEG compounds would be seen as beneficial in clinical applications.

Claims
  • 1. A tetrapyrollic photosensitizer compound said tetrapyrollic compound being a chlorin, bacteriochlorin, porphyrin, pyropheophorbide, purpurinimide, or bacteriopurpurinimide having 3 to 6 —CH2CONHphenylCH2CH(N(CH2COOH)2)(CH2N(CH2COOH)(CH2CH2N(CH2COOH)2)) groups or esters thereof or complexes thereof with gadolinium(III).
  • 2. The compound of claim 1 having at least one pendant —CH2CH2CONHC(CH2CH2CONHphenylCH2CH(N(CH2COOH)2(CH2N(CH2COOH)—(CH2CH2N(CH2COOH)2)))3 group or esters thereof or complexes thereof with gadolinium(III).
  • 3. A compound of the formula:
  • 4. The compound of claim 3 where R9 is —CH2CH2CONHC(CH2CH2CONHphenylCH2CH(N(CH2COOH)2)(CH2N(CH2COOH)(CH2CH2N(CH2COOH)2)))3 group or esters thereof or complexes thereof with gadolinium(III).
  • 5. A compound according to claim 3 having the formula:
  • 6. A compound according to claim 3 having the formula:
  • 7. A compound according to claim 2 having the formula:
  • 8. The compound of claim 3, wherein: R1 is substituted or unsubstituted alkyl;R2 is substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, or —C(O)Ra,where Ra is substituted or unsubstituted alkyl;R1a and R2a together form a covalent bond;R3 and R4 are each independently substituted or unsubstituted alkyl;R3a and R4a are each independently hydrogen, or together form a covalent bond;R5 is substituted or unsubstituted alkyl;R6 and R6a together form ═O;R7 is azaalkyl, or azaaralkyl;R8 and R8a together form ═O;R9 and R10 are each independently substituted or unsubstituted alkyl;each of R1-R10, when substituted, is substituted with one or more substituents each independently selected from Q, where Q is halo, haloalkyl, COORb where Rb is hydrogen or alkyl, ORc where Rc is alkyl or aralkyl, NRfRg where Rf and Rg are each independently hydrogen, alkyl or aralkyl, or ═NRh where Rh is aralkyl;each Q is independently unsubstituted or is substituted with one or more substituents each independently selected from Q1, where Q1 is halo, or haloalkyl.
  • 9. The compound of claim 3, wherein: R1 is unsubstituted alkyl;R2 is substituted or unsubstituted alkyl, unsubstituted alkenyl, or —C(O)Ra, where Ra is unsubstituted alkyl;R1a and R2a together form a covalent bond;R3 and R4 are each independently unsubstituted alkyl;R3a and R4a are each independently hydrogen, or together form a covalent bond;R5 is unsubstituted alkyl;R6 and R6a together form ═O;R7 is azaalkyl, or azaaralkyl;R8 and R8a together form ═O;R10 is unsubstituted alkyl;each of R1-R10, when substituted, is substituted with one or more substituents each independently selected from Q, where Q is halo, haloalkyl, COORb where Rb is hydrogen or alkyl, ORc where Rc is alkyl or aralkyl, NRfRg where Rf and Rg are each independently hydrogen, alkyl or aralkyl, or ═NRh where Rh is aralkyl;each Q is independently unsubstituted or is substituted with one or more substituents each independently selected from Q1, where Q1 is halo, or haloalkyl.
  • 10. The compound of claim 3, wherein: R1 is methyl;R1a and R2a together form a covalent bond;R3 is methyl;R4 is ethyl;R3a and R4a are each independently hydrogen, or together form a covalent bond;R5 is methyl; andR10 is methyl.
  • 11. The compound claim 3, wherein: R2 is CH═CH2, CH(OR20)CH3, C(O)Me, C(═NR21)CH3 or CH(NHR21)CH3;where R20 is methyl, butyl, heptyl, dodecyl or 3,5-bis(trifluoromethyl)-benzyl; andR21 is 3,5-bis(trifluoromethyl)benzyl.
  • 12. The compound of claim 3, wherein: R7 is ═NR20, where R20 is methyl, butyl, heptyl, dodecyl or 3,5-bis(trifluoromethyl)-benzyl.
  • 13. A pharmaceutical composition, comprising a compound of claim 1 in a pharmaceutically acceptable carrier.
  • 14. A pharmaceutical composition, comprising a compound of claim 3 in a pharmaceutically acceptable carrier.
  • 15. An article of manufacture, comprising packaging material and a compound of claim 1 contained within the packaging material, and the packaging material includes a label that indicates that the compound is used in a photodynamic therapy treatment for hyperproliferative tissue.
  • 16. An article of manufacture, comprising packaging material and a compound of claim 3 contained within the packaging material, and the packaging material includes a label that indicates that the compound is used in a photodynamic therapy treatment for hyperproliferative tissue.
  • 17. A method for administering a therapy to hyperproliferative tissue, comprising: (i) administering to a subject the compound of claim 1 that selectively interacts with the hyperproliferative tissue relative to normal tissue, and(ii) irradiating the hyperproliferative tissue with light of a wavelength to kill or impair hyperproliferative tissue.
  • 18. The method of claim 17, wherein the hyperproliferatve tissue is selected from the group consisting of: a vascular endothelial tissue, a neovasculature tissue, a neovasculature tissue present in an eye, an abnormal vascular wall of a tumor, a solid tumor, a tumor of a head, a tumor of a neck, a tumor of an eye, a tumor of a gastrointestinal tract, a tumor of a liver, a tumor of a breast, a tumor of a prostate, a tumors of a lung, a nonsolid tumor, and malignant cells of one of a hematopoietic tissue and a lymphoid tissue.
  • 19. A method for administering a therapy to a hyperproliferative tissue, comprising: (i) administering to a subject the compound of claim 3 that selectively interacts with hyperproliferative tissue relative to normal tissue, and(ii) irradiating the subject with light of a wavelength to kill or impair hyperproliferative tissue.
  • 20. The method of claim 19, wherein the hyperproliferative tissue is selected from the group consisting of: a vascular endothelial tissue, a neovasculature tissue, a neovasculature tissue present in an eye, an abnormal vascular wall of a tumor, a solid tumor, a tumor of a head, a tumor of a neck, a tumor of an eye, a tumor of a gastrointestinal tract, a tumor of a liver, a tumor of a breast, a tumor of a prostate, a tumors of a lung, a nonsolid tumor, and malignant cells of one of a hematopoietic tissue and a lymphoid tissue.
  • 21. The method of claim 17, further comprising the step of allowing time for any of the compound that is not selectively interacted with the hyperproliferative tissue to clear from normal tissue of the subject prior to the step of irradiating.
  • 22. A method of photodynamic therapy for treating hyperproliferative tissue in a subject, comprising: (i) administering to the subject the compound of claim 1 that selectively interacts with the hyperproliferative tissue relative to normal tissue, and(ii) irradiating the subject with light of a wavelength to activate the compound, whereby the hyperproliferative tissue is destroyed or impaired.
  • 23. A method of photodynamic therapy for treating hyperproliferative tissue in a subject, comprising: (i) administering to the subject the compound of claim 3 that selectively interacts with the hyperproliferative tissue relative to normal tissue, and(ii) irradiating the subject with light of a wavelength to activate the compound, whereby the hyperproliferative tissue is destroyed or impaired.
  • 24. A method for detecting the presence of a hyperproliferative tissue in a subject comprising: (i) administering to the subject an effective quantity of the compound of claim 1, said compound being fluorescent and that selectively interacts with the hyperproliferative tissue relative to normal tissue; and(ii) visualizing the compound within the patient by fluorescent spectroscopy.
  • 25. A method for detecting the presence of a hyperproliferative tissue in a subject comprising: (i) administering to the subject an effective quantity of the compound of claim 3, said compound being fluorescent and that selectively interacts with the hyperproliferative tissue relative to normal tissue; and(ii) visualizing the compound within the patient by fluorescent spectroscopy.
  • 26. A method for detecting the presence of a hyperproliferative tissue in a subject comprising: (i) administering to the subject an effective quantity of the compound of claim 1, said compound being magnetically resonant and that selectively interacts with the hyperproliferative tissue; and(ii) visualizing the compound within the patient by MRI imaging.
  • 27. A method for detecting the presence of a hyperproliferative tissue in a subject comprising: (i) administering to the subject an effective quantity of the compound of claim 3, said compound being magnetically resonant and that selectively interacts with the hyperproliferative tissue relative to normal tissue; and(ii) visualizing the compound within the patient by MRI imaging.
CROSS REFERENCE TO RELATED APPLICATIONS

This is a continuation-in-part of U.S. patent application Ser. No. 10/390,438 filed Mar. 17, 2003 now U.S. Pat. No. 7,078,014 entitled METHOD FOR USING CHLORIN AND BACTERIOCHLORIN-BASED AMINOPHENYL DTPA AND N2S2 CONJUGATES FOR MR CONTRAST MEDIA AND RADIOPHARMACEUTICALS which in turn is a continuation-in-part of U.S. patent application Ser. No. 09/739,155 filed Dec. 18, 2000 now U.S. Pat. No. 6,534,040 entitled CHLORIN AND BACTERIOCHLORIN-BASED AMINOPHENYL DTPA AND N2S2 CONJUGATES FOR MR CONTRAST MEDIA AND RADIOPHARMACEUTICALS which in turn claims priority from U.S. Provisional Patent Application 60/171,961 filed Dec. 23, 1999 entitled CHLORIN AND BACTERIOCHLORINE-BASED AMINOPHENYL DTPA AND N2S2 CONJUGATES FOR MR CONTRAST MEDIA AND RADIOPHARMACEUTICALS; And, this is a continuation-in-part of U.S. patent application Ser. No. 11/452,511 to Pandey et al. filed Jun. 14, 2006 now U.S. Pat. No. 7,501,509 entitled WATER SOLUBLE TETRAPYROLLICPHOTOSENSITIZERS FOR PHOTODYNAMIC THERAPY, which is a continuation-in-part of U.S. application Ser. No. 10/607,922 to Pandey et al. filed Jun. 27, 2003 now U.S. Pat. No. 7,166,719 entitled FLUORINATED PHOTOSENSITIZERS RELATED TO CHLORINS AND BACTERIOCHLORINS FOR PHOTODYNAMIC THERAPY which in turn claims priority from Provisional Application Ser. No. 60/392,473 to Pandey et al. filed Jun. 27, 2002 entitled FLUORINATED PHOTOSENSITIZERS RELATED TO CHLORINS AND BACTERIOCHLORINS FOR PHOTODYNAMIC THERAPY. The above applications are incorporated herein by reference in their entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

This invention was made with government support under Grant Numbers NIH CA 55792 and NIH R21 CA109914 awarded by the National Institute of Health. The government has certain rights in this invention.

US Referenced Citations (283)
Number Name Date Kind
3710795 Higuchi et al. Jan 1973 A
3817837 Rubenstein et al. Jun 1974 A
3927193 Hansen et al. Dec 1975 A
RE28819 Thompson May 1976 E
4044126 Cook et al. Aug 1977 A
4328245 Yu et al. May 1982 A
4331647 Goldenberg May 1982 A
4348376 Goldenberg Sep 1982 A
4358603 Yu Nov 1982 A
4361544 Goldenberg Nov 1982 A
4364923 Cook et al. Dec 1982 A
4374925 Litman et al. Feb 1983 A
4409239 Yu Oct 1983 A
4410545 Yu et al. Oct 1983 A
4414209 Cook et al. Nov 1983 A
4444744 Goldenberg Apr 1984 A
4468457 Goldenberg et al. Aug 1984 A
4474893 Reading Oct 1984 A
4479895 Auditore-Hargreaves Oct 1984 A
4521762 Kapral Jun 1985 A
4522811 Eppstein et al. Jun 1985 A
4577636 Spears Mar 1986 A
4624846 Goldenberg Nov 1986 A
4649151 Dougherty et al. Mar 1987 A
4656186 Bommer et al. Apr 1987 A
4675338 Bommer et al. Jun 1987 A
4693885 Bommer et al. Sep 1987 A
4753958 Weinstein et al. Jun 1988 A
4818709 Primus et al. Apr 1989 A
4861876 Kessel Aug 1989 A
4866168 Dougherty et al. Sep 1989 A
4878891 Judy et al. Nov 1989 A
4889129 Dougherty et al. Dec 1989 A
4916221 Kumadaki et al. Apr 1990 A
4925736 Shikowitz May 1990 A
4932934 Dougherty et al. Jun 1990 A
4935498 Sessler et al. Jun 1990 A
4946778 Ladnar et al. Aug 1990 A
4957481 Gatenby Sep 1990 A
4968715 Dougherty et al. Nov 1990 A
4997639 Aizawa et al. Mar 1991 A
5002962 Pandey et al. Mar 1991 A
5004811 Bommer et al. Apr 1991 A
5015463 Dougherty et al. May 1991 A
5028594 Carson Jul 1991 A
5028621 Dougherty et al. Jul 1991 A
5033252 Carter Jul 1991 A
5041078 Matthes et al. Aug 1991 A
5051415 Moran et al. Sep 1991 A
5052558 Carter Oct 1991 A
5053006 Watson Oct 1991 A
5059415 Neuwelt Oct 1991 A
5062431 Potter Nov 1991 A
5066274 Bommer et al. Nov 1991 A
5066291 Stewart Nov 1991 A
5074632 Potter Dec 1991 A
5093349 Pandey et al. Mar 1992 A
5095030 Levy et al. Mar 1992 A
5111821 Potter May 1992 A
5145863 Dougherty et al. Sep 1992 A
5171741 Dougherty Dec 1992 A
5173504 Dougherty Dec 1992 A
5190536 Wood et al. Mar 1993 A
5190966 Dougherty et al. Mar 1993 A
5198460 Pandey et al. Mar 1993 A
5205291 Potter Apr 1993 A
5216012 Morgan et al. Jun 1993 A
5219345 Potter Jun 1993 A
5222795 Hed Jun 1993 A
5225433 Dougherty et al. Jul 1993 A
5257970 Dougherty Nov 1993 A
5263925 Gilmore, Jr. et al. Nov 1993 A
5298018 Narciso, Jr. Mar 1994 A
5308861 Aizawa et al. May 1994 A
5314905 Pandey et al. May 1994 A
5323907 Kalvelage Jun 1994 A
5330741 Smith et al. Jul 1994 A
5344928 Masuya et al. Sep 1994 A
5368841 Trauner et al. Nov 1994 A
5403308 Wood et al. Apr 1995 A
5418130 Platz et al. May 1995 A
5430051 Aizawa et al. Jul 1995 A
5441531 Zarate et al. Aug 1995 A
5459159 Pandey et al. Oct 1995 A
5482698 Griffiths Jan 1996 A
5484803 Richter Jan 1996 A
5496308 Brown et al. Mar 1996 A
5498710 Pandey et al. Mar 1996 A
5500009 Mendes et al. Mar 1996 A
5503637 Kyricos et al. Apr 1996 A
5506255 Smith et al. Apr 1996 A
5514669 Selman May 1996 A
5525338 Goldenberg Jun 1996 A
5532171 Motsenbocker Jul 1996 A
5534506 Morgan et al. Jul 1996 A
5549660 Mendes et al. Aug 1996 A
5556612 Anderson et al. Sep 1996 A
5567409 Aizawa et al. Oct 1996 A
5571152 Chen et al. Nov 1996 A
5580896 Horwell et al. Dec 1996 A
5591847 Pandey et al. Jan 1997 A
5594136 Sessler et al. Jan 1997 A
5599923 Sessler et al. Feb 1997 A
5622983 Horwell et al. Apr 1997 A
5624798 Yamamoto et al. Apr 1997 A
5631281 Horwell et al. May 1997 A
5648485 Dolphin et al. Jul 1997 A
5665328 Horan et al. Sep 1997 A
5667998 Dougherty et al. Sep 1997 A
5671317 Weishaupt et al. Sep 1997 A
5686280 Dougherty et al. Nov 1997 A
5688486 Watson et al. Nov 1997 A
5697902 Goldenberg Dec 1997 A
5698405 Goldenberg Dec 1997 A
5702432 Chen et al. Dec 1997 A
5703230 Boyle et al. Dec 1997 A
5705518 Richter et al. Jan 1998 A
5709874 Hanson et al. Jan 1998 A
5715837 Chen Feb 1998 A
5716595 Goldenberg Feb 1998 A
5736563 Richter Apr 1998 A
5741316 Chen et al. Apr 1998 A
5756541 Strong et al. May 1998 A
5759542 Gurewich Jun 1998 A
5766234 Chen et al. Jun 1998 A
5770619 Richter et al. Jun 1998 A
5770730 Pandey et al. Jun 1998 A
5773977 Dougherty Jun 1998 A
5776093 Goldenberg Jul 1998 A
5776094 Goldenberg Jul 1998 A
5776095 Goldenberg Jul 1998 A
5782896 Chen et al. Jul 1998 A
5800478 Chen et al. Sep 1998 A
5814008 Chen et al. Sep 1998 A
5824080 Lamuraglia Oct 1998 A
5827186 Chen et al. Oct 1998 A
5829448 Fisher et al. Nov 1998 A
5831088 Dolphin et al. Nov 1998 A
5832931 Wachter et al. Nov 1998 A
5840674 Yatvin et al. Nov 1998 A
5851225 Lawandy Dec 1998 A
5860957 Jacobsen et al. Jan 1999 A
5864035 Pandey et al. Jan 1999 A
5865840 Chen Feb 1999 A
5876427 Chen et al. Mar 1999 A
5885557 Lentini Mar 1999 A
5886173 Hemmi et al. Mar 1999 A
5900252 Calanchi et al. May 1999 A
5906928 Dougherty et al. May 1999 A
5913884 Trauner et al. Jun 1999 A
5921244 Chen et al. Jul 1999 A
5942534 Trauner et al. Aug 1999 A
5944748 Mager et al. Aug 1999 A
5945762 Chen et al. Aug 1999 A
5948433 Burton et al. Sep 1999 A
5952366 Pandey et al. Sep 1999 A
5957960 Chen et al. Sep 1999 A
5972366 Haynes et al. Oct 1999 A
5976535 Fritzberg et al. Nov 1999 A
5983134 Ostrow Nov 1999 A
5985307 Hanson et al. Nov 1999 A
5985317 Venkateshwaran et al. Nov 1999 A
5997569 Chen et al. Dec 1999 A
5997842 Chen Dec 1999 A
5998597 Fisher et al. Dec 1999 A
6004534 Langer et al. Dec 1999 A
6010715 Wick et al. Jan 2000 A
6015897 Theodore et al. Jan 2000 A
6022961 Yamamoto et al. Feb 2000 A
6024975 D'Angelo et al. Feb 2000 A
6028099 de Juan, Jr. Feb 2000 A
6036941 Bottiroli et al. Mar 2000 A
6039975 Shah et al. Mar 2000 A
6048359 Biel Apr 2000 A
6048736 Kosak Apr 2000 A
6051207 Klaveness et al. Apr 2000 A
6051702 Bird et al. Apr 2000 A
6060082 Chen et al. May 2000 A
6063108 Salansky et al. May 2000 A
6063777 Hikida et al. May 2000 A
6071495 Unger et al. Jun 2000 A
6080160 Chen et al. Jun 2000 A
6084717 Wood et al. Jul 2000 A
6090788 Lurie Jul 2000 A
6092531 Chen et al. Jul 2000 A
6096066 Chen et al. Aug 2000 A
6096289 Goldenberg Aug 2000 A
6100893 Ensz et al. Aug 2000 A
6103751 Pandey et al. Aug 2000 A
6107466 Hasan et al. Aug 2000 A
6117862 Margaron et al. Sep 2000 A
6120751 Unger Sep 2000 A
6123923 Unger et al. Sep 2000 A
6124342 Okamoto et al. Sep 2000 A
6131570 Schuster et al. Oct 2000 A
6138681 Chen et al. Oct 2000 A
6139865 Friend et al. Oct 2000 A
6152951 Hashimoto et al. Nov 2000 A
6156506 Yamamoto et al. Dec 2000 A
6162213 Stewart Dec 2000 A
6162242 Peyman Dec 2000 A
6167301 Flower et al. Dec 2000 A
6176842 Tachibana et al. Jan 2001 B1
6187030 Gart et al. Feb 2001 B1
6210425 Chen Apr 2001 B1
6217869 Meyer et al. Apr 2001 B1
RE37180 Mori et al. May 2001 E
6232295 Kayyem et al. May 2001 B1
6238426 Chen May 2001 B1
6242477 Okamoto et al. Jun 2001 B1
6253872 Neumann Jul 2001 B1
6256533 Yuzhakov et al. Jul 2001 B1
6261595 Stanley et al. Jul 2001 B1
6264914 Klaveness et al. Jul 2001 B1
6267983 Fujii et al. Jul 2001 B1
6268120 Platz et al. Jul 2001 B1
6271359 Norris et al. Aug 2001 B1
6273904 Chen et al. Aug 2001 B1
6274552 Tamarkin et al. Aug 2001 B1
6281611 Chen et al. Aug 2001 B1
6307147 Bird et al. Oct 2001 B1
6316652 Steliou Nov 2001 B1
6319273 Chen et al. Nov 2001 B1
6319488 Licha et al. Nov 2001 B1
6331175 Goldenberg Dec 2001 B1
6331744 Chen et al. Dec 2001 B1
6344050 Chen Feb 2002 B1
6350431 Snow et al. Feb 2002 B1
6387350 Goldenberg May 2002 B2
6406297 Raymond et al. Jun 2002 B1
6416531 Chen Jul 2002 B2
6454789 Chen et al. Sep 2002 B1
6482517 Anderson Nov 2002 B1
6489314 Ashley et al. Dec 2002 B1
6495585 Bellnier et al. Dec 2002 B2
6498945 Alfheim et al. Dec 2002 B1
6500816 Ekimoto et al. Dec 2002 B1
6511971 Gorun Jan 2003 B1
6514995 Zaleski et al. Feb 2003 B1
6515113 Raymond et al. Feb 2003 B2
6520669 Chen et al. Feb 2003 B1
6524552 Klaveness et al. Feb 2003 B2
6525088 Nagano et al. Feb 2003 B1
6527759 Tachibana et al. Mar 2003 B1
6534040 Pandey et al. Mar 2003 B2
6540980 Blumenthal et al. Apr 2003 B1
6554853 Chen Apr 2003 B2
6559374 Lindsey et al. May 2003 B2
6566517 Miura et al. May 2003 B2
6569846 Scherz et al. May 2003 B1
6572839 Sugita et al. Jun 2003 B2
6580228 Chen et al. Jun 2003 B1
6602274 Chen Aug 2003 B1
6624187 Pandey et al. Sep 2003 B1
6657351 Chen et al. Dec 2003 B2
6849607 Pandey et al. Feb 2005 B2
6899723 Chen May 2005 B2
6986782 Chen et al. Jan 2006 B2
RE38994 Pandey et al. Feb 2006 E
7018395 Chen Mar 2006 B2
RE39094 Pandey et al. May 2006 E
7053210 Pandey et al. May 2006 B2
7078014 Pandey et al. Jul 2006 B2
7097826 Pandey et al. Aug 2006 B2
7147840 Pandey et al. Dec 2006 B2
7166719 Pandey et al. Jan 2007 B2
7501509 Pandey et al. Mar 2009 B2
20010022970 Dees et al. Sep 2001 A1
20020033192 Lindsey et al. Mar 2002 A1
20020049247 Chen Apr 2002 A1
20020087205 Chen Jul 2002 A1
20020127224 Chen Sep 2002 A1
20020127230 Chen Sep 2002 A1
20020128303 Bellnier et al. Sep 2002 A1
20020198576 Chen et al. Dec 2002 A1
20030018371 Chen Jan 2003 A1
20030030342 Chen et al. Feb 2003 A1
20030109813 Chen Jun 2003 A1
20030114434 Chen et al. Jun 2003 A1
20030167033 Chen et al. Sep 2003 A1
20030208249 Chen Nov 2003 A1
20040044197 Pandey et al. Mar 2004 A1
20040044198 Pandey et al. Mar 2004 A1
Foreign Referenced Citations (86)
Number Date Country
0120054 Mar 1984 EP
0161606 Nov 1985 EP
0243929 Nov 1987 EP
0423195 Apr 1991 EP
0425566 May 1991 EP
0450149 Oct 1991 EP
0468997 Feb 1992 EP
0510007 Oct 1992 EP
0682956 Nov 1995 EP
1110963 Jun 2001 EP
1131100 Sep 2001 EP
1146046 Oct 2001 EP
1164136 Dec 2001 EP
1238666 Sep 2002 EP
1256586 Nov 2002 EP
1334748 Aug 2003 EP
4218002 Jul 1992 JP
6105921 Apr 1994 JP
2001335578 Apr 2001 JP
2002020389 Jan 2002 JP
2002325853 Nov 2002 JP
2003146989 May 2003 JP
8401382 Apr 1984 WO
9000392 Jan 1990 WO
9000895 Feb 1990 WO
9012573 Nov 1990 WO
9110474 Jul 1991 WO
9313769 Jul 1993 WO
9409851 May 1994 WO
9505214 Feb 1995 WO
9532206 Nov 1995 WO
9637255 Nov 1996 WO
9732520 Sep 1997 WO
9732885 Sep 1997 WO
9804317 Feb 1998 WO
9806456 Feb 1998 WO
9808565 Mar 1998 WO
9814243 Apr 1998 WO
9824371 Jun 1998 WO
9824510 Jun 1998 WO
9832491 Jul 1998 WO
9832492 Jul 1998 WO
9832493 Jul 1998 WO
9846130 Oct 1998 WO
9850034 Nov 1998 WO
9856302 Dec 1998 WO
9918879 Apr 1999 WO
9920346 Apr 1999 WO
9939769 Aug 1999 WO
9952565 Oct 1999 WO
9968149 Nov 1999 WO
9966988 Dec 1999 WO
9967248 Dec 1999 WO
9967249 Dec 1999 WO
WO9967248 Dec 1999 WO
WO9967249 Dec 1999 WO
0015296 Mar 2000 WO
0036983 Jun 2000 WO
0041725 Jul 2000 WO
0041726 Jul 2000 WO
0041727 Jul 2000 WO
0041768 Jul 2000 WO
0061584 Oct 2000 WO
0103770 Jan 2001 WO
0105316 Jan 2001 WO
0115694 Mar 2001 WO
0143825 Jun 2001 WO
0151087 Jul 2001 WO
0174398 Oct 2001 WO
0176216 Oct 2001 WO
0178458 Oct 2001 WO
0198708 Dec 2001 WO
0217690 Feb 2002 WO
02098882 Dec 2002 WO
03029494 Apr 2003 WO
03050082 Jun 2003 WO
03052793 Jun 2003 WO
WO03052793 Jun 2003 WO
03056407 Jul 2003 WO
03061695 Jul 2003 WO
WO03061696 Jul 2003 WO
2004002476 Jan 2004 WO
2004005289 Jan 2004 WO
WO2004002476 Jan 2004 WO
WO2004002486 Jan 2004 WO
WO2004005289 Jan 2004 WO
Related Publications (1)
Number Date Country
20070053840 A1 Mar 2007 US
Provisional Applications (2)
Number Date Country
60392473 Jun 2002 US
60171961 Dec 1999 US
Continuation in Parts (5)
Number Date Country
Parent 10390438 Mar 2003 US
Child 11479524 US
Parent 09739155 Dec 2000 US
Child 10390438 US
Parent 11479524 US
Child 10390438 US
Parent 11452511 Jun 2006 US
Child 11479524 US
Parent 10607922 Jun 2003 US
Child 11452511 US