US20050226810A1 - Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis - Google Patents

Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis Download PDF

Info

Publication number
US20050226810A1
US20050226810A1 US11/059,557 US5955705A US2005226810A1 US 20050226810 A1 US20050226810 A1 US 20050226810A1 US 5955705 A US5955705 A US 5955705A US 2005226810 A1 US2005226810 A1 US 2005226810A1
Authority
US
United States
Prior art keywords
mono
group
residue
alkyl
aryl
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
US11/059,557
Inventor
Byron Robinson
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.)
Miravant Pharmaceuticals Inc
Original Assignee
Miravant Pharmaceuticals 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 Miravant Pharmaceuticals Inc filed Critical Miravant Pharmaceuticals Inc
Priority to US11/059,557 priority Critical patent/US20050226810A1/en
Publication of US20050226810A1 publication Critical patent/US20050226810A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • A61K47/546Porphyrines; Porphyrine with an expanded ring system, e.g. texaphyrine
    • 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
    • A61K49/085Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier conjugated systems
    • 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
    • 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
    • A61K49/101Organic compounds the carrier being a complex-forming compound able to form MRI-active complexes with paramagnetic metals
    • A61K49/106Organic compounds the carrier being a complex-forming compound able to form MRI-active complexes with paramagnetic metals the complex-forming compound being cyclic, e.g. DOTA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/041Heterocyclic compounds
    • A61K51/044Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins
    • A61K51/0446Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K51/0451Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having four such rings, e.g. phorphine derivatives, bilirubin, biliverdine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0474Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group
    • A61K51/0485Porphyrins, texaphyrins wherein the nitrogen atoms forming the central ring system complex the radioactive metal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • This invention is directed to substituted porphyrin and azaporphyrin derivatives with various substituents in the 13- and 17-positions of the porphyrin skeleton suitable as pharmaceutical agents for use in photodynamic therapy, MRI diagnosis, and radiodiagnostics.
  • the invention is also directed to pharmaceutical agents that contain these compounds, as well as a process for the production of these compounds and agents.
  • Photodynamic therapy is a new modality for the treatment of malignancies, diseased tissue, hyperproliferating tissues, pathogens or unwanted normal tissues.
  • PDT involves a localized or systemic administration of a photosensitizing compound followed by exposure of target tissue to photoactivating light.
  • the photoactivating light excites the photosensitizer which, in turn, interacts with singlet oxygen causing the production of cytotoxic oxygen species.
  • the interaction of the cytotoxic oxygen species with tissues in which the photosensitizer is localized causes a modification of the tissue, resulting in a desired clinical effect.
  • the tissue specificity of the resultant phototoxic damage is determined largely, although not entirely, by the relative concentrations of the photosensitizer in each tissue at the time of its exposure to the photoactivating light.
  • Porphyrins and azaporphyrins and their metallated derivatives belong to a family of substances that are suitable for PDT. These compounds accumulate in target tissues and absorb light in a range in which living tissue is still fairly permeable, namely between 380-680 nm. Moreover, porphyrins, azaporphyrins and their photoactive metallated derivatives exhibit high yields of the excited triplet state, a long lifetime in this state, and good energy transfer to oxygen with concomittent production of singlet oxygen. Of the porphyrins and their derivatives, several photosensitizers have been developed largely for use in oncological applications, but have also been examined in other disease areas in the PDT field in humans.
  • Such photosensitizers include Photofrin (U.S. Pat. No. 4,882,234), 5-aminolevulinic acid (protoporphyrin IX precursor), SnET2, Visudyne® (Benzoporphyrin derivative), Antrin®, Optrin® (Lutetium texaphyrin) and mono-aspartyl chlorin e6 (MACE). All of these compounds were designed specifically for the treatment of solid tumors.
  • these compounds were designed to have large absorptions in the 620-740 nm range so as to optimize the photoactivation of the drug with a wavelength that will penetrate to the greatest depths possible all tissue types.
  • these drugs were designed to absorb outside of the blood absorption profile, thus ensuring efficient photoactivation in most tissue types.
  • the excitation light source (usually diode lasers or dye lasers) has historically been matched to the far-red absorption bandwidth of the photosensitizer in order to maximize light penetration through tissues. Indeed, the present inventor believes that all the tetrapyrrolic photosensitizers used have been designed for long wavelength absorption of light (>630 nm) to address this perceived issue. Surprisingly, it has been found that short wavelength photosensitizers (with activation absorptions ⁇ 600 nm) are capable of delivering effective localized therapy to many disease indications where historically long wavelength photosensitizers (with activation absorptions >600 nm) have shown ineffective clinical outcomes. One such example is in coronary artery disease.
  • photosensitizers described above have been used to treat atheromatous plaques and some are able to display some inhibition of intimal hyperplasia in animal models, many if not all have characteristics that will limit the usefulness of these drugs in a clinical setting.
  • One particular concern is the half-life of the photosensitizer.
  • a photosensitizer delivered systemically with a long half-life may have phototoxic side effects if exposed to direct light, within days of the procedure.
  • wavelengths of light lower than 600 nm offer significant advantages in PDT because such wavelengths have penetration characteristics that deliver the PDT effect to the target sites (media and adventicia layers of the vessel) and not to myocardial tissue. Thus, effective therapy can be afforded at the target site, while deeper tissues are shielded from a PDT response by blood absorption within these tissues.
  • Previously reported cardiovascular experiments performed to date on tetrapyrrolic molecules have been done at wavelengths >620 nm. Experiments that we have performed in pig arteries with new photosensitizer candidates at light activation >600 nm have resulted in unacceptable levels of damage to myocardial or cardiac muscle tissue surrounding the treatment area.
  • chlorins, phthalocyanines and texaphyrin type photosensitizers in general have little absorption in the 500-600 nm regions, and thus may be suboptimal with regard to light activation at green and yellow wavelengths in cardiovascular tissues.
  • protoporphyrin IX and photofrin do not display absorption maximas at 532 nm, thus they may be inefficient at absorbing treatment light at this wavelength and have very low molar extinction coefficients at 575 nm ( ⁇ 7000 cm ⁇ 1 /M ⁇ 1 ).
  • long wavelength photosensitizers by design have red absorption peaks, operating room lighting in an emergency situation may cause serious photosensitivity in light exposed tissues.
  • hematoporphyrin complex compounds used in diagnosis and treatment are described in patent application EP 0 355 041. While these compounds show a good concentration behavior in various target organs, the described compounds used as NMR diagnostic agents are not satisfacatory because they require a dose necessary for optimal imaging that is too close to the lethal dose. Hematoporphyrin derivatives have the drawback that they can eliminate both pseudobenzylic OH groups in the hydroxyethyl side chains.
  • Sakata's porphyrin-based PDT/MRI/radiodiagnostic compounds are based on a naturally occurring asymmetrical porphyrin ring system shown in FIG. 1.
  • Sakata has linked polyfunctional carboxyl groups that are capable of binding radioactive metals or MRI active metals to a) the R 1 and R 2 positions as shown via ether—alcohol linkages; and b) to positions R 4 or R 5 via ether linking units. This synthetic approach carries with it significant manufacturing problems.
  • the linking of one metal chelating moiety to an asymmetrical porphyrin at R 1 or R 2 , R 3 or R 4 (where R 1 and R 2 can be vinyl, ethyl, —CH(O-lower alkanoyl)CH 3 , —CH(OR)CH 3 or —CH(O-loweralklene-OR)CH_,) generates at least two new chemical porphyrinic entities in the synthesis process if R 1 and R 2 (or R 3 and R 4 ) are the same linking moiety. This is outlined in scheme 1.
  • Naturally occurring porphyrins like hematoporphyrin or protoporphyrin cannot be chemically modified such that only one position, either R 1 or R 2 , (or R 3 or R 4 ) is selectively modified to form a molecule with a single linking unit as the only product.
  • R 1 or R 2 , (or R 3 or R 4 ) is selectively modified to form a molecule with a single linking unit as the only product.
  • two compounds are always formed which must be separated to obtain a pure single molecule with which to link the metal chelating moiety.
  • the separation of the two porphyrins is often difficult (if not impossible) and complicates both the manufacturing process and the cost of the final product. If one chooses not to separate the isomers, the isomeric components will each have their own toxicities, and pharmacokinetic and distribution profiles.
  • Niedballa and Platzek's approach also has the same synthetic manufacturing problems as explained for Sakata (except when R 1 ⁇ H), i.e., multiple compounds are produced when a single linking moiety is attached to the molecule. These molecules may offer enhanced stability over Sakata's due to the use of amine linkages.
  • the limitation of R 1 ⁇ H symmetry does not, however, allow for modification of this molecule with other functionality that may enhance localization or uptake in tissues or target organelle, or changes in pharmacokinetic or elimination profiles for singly linked molecules. Compounds with high water solubility are often not taken up efficiently by tumors or cells. The ability to enhance the lipophilicity of the molecule is thus very important.
  • the theoretical efficacy of a photosensitizer largely correlates to the molar extinction coefficient of the photosensitizer's absorption peak and its ability to absorb light. This is due primarily to the fact that the ability of a photosensitizer to absorb incidental light is a function of the cross sectional area of the molecule's absorption profile. Hence, photosensitizers with low molar extinction coefficients capture photons less efficiently than molecules with high molar extinction coefficients and are thus less efficient.
  • the present inventor has found novel metal-free or metallated functionalized phototherapeutic agents that may be used for imaging (MRI or radiodiagnostic) before or after photodynamic therapy.
  • These novel phototherapeutic agents are based on tetrapyrrolic ring systems such as the porphyrins and azaporphyrins that can be covalently linked by stable linkages to metal complexing agents. These new photosensitizers are useful in short wavelength applications in photodynamic therapy.
  • the present invention in one aspect provides phototherapeutic compositions of metallo-tetrapyrrolic compounds of formula I which may be used as MRI, radiodiagnostic and PDT agents:
  • R 1 -R 12 can be the same or different and can be selected from:
  • M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga 3+ , Pt 2+ , Pd 2+ , Sn 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ either with or without a physiologically acceptable charge balancing counter ion.
  • R 1 -R 7 can be the same or different and can be selected from:
  • M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably scheduled from Ga 3+ , Pt 2+ , Pd 2+ , Sn 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ either with or without a physiologically acceptable charge balancing counter ion.
  • R 1 and R 2 can be the same or different and can be selected from H, NO 2 , CN, CHO, CO-alkyl, SO 3 H, SO 3 alkyl, SO 3 alkylether, SO 3 heteroalkyl, SO 3 Na, SO 3 K, SO 2 NHalkyl, SO 2 N(alkyl) 2 , SO 2 NHheteroalkyl, SO 2 N(heteroalkyl) 2 , SO 2 NHhaloalkyl, SO 2 N(haloalkyl) 2 , SO 2 NHhaloalkylether, SO 2 N(haloalkylether) 2 , SO 2 NHalkylether, SO 2 N(haloalkylether) 2 , SO 2 NHalkylether, SO 2 N(haloalkylether) 2 , CO-haloalkyl, haloalkyl, heteroalkyl, hydroxyhaloalkyl, haloalkyl ether
  • M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga 3+ ; Pt 2+ , Pd 2+ , Sn 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ either with or without a physiologically acceptable charge balancing counter ion.
  • R 1 -R 11 can be the same or different and can be selected from:
  • M is 2H or a diamagnetic or paramagnetic metal ion that can be radioactive, photoactive metals being preferably selected from Ga 3+ , Pt 2+ , Pd 2+ , Sn 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ , either with or without a physiologically acceptable charge balancing counter ion.
  • R 1 -R 6 can be the same or different and can be selected from:
  • M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga 3+ , Pt 2+ , Pd 2+ , S 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ , either with or without a physiologically acceptable charge balancing counter ion.
  • R 1 -R 10 can be the same or different and can be selected from:
  • M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga 3+ , Pt 2+ , Pd 2+ , Sn 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ either with or without a physiologically acceptable charge balancing counter ion.
  • R 1 -R 4 can be the same or different and are selected from:
  • M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga 3+ , Pt 2+ , Pd 2+ , Sn 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ either with or without a physiologically acceptable charge balancing counter ion.
  • phototherapeutic compositions of metallo-tetrapyrrolic compounds of formula IV that may be used as MRI, radiodiagnostic, or PDT agents:
  • R 1 -R 8 can be the same or different and are selected from:
  • M is selected from 2H or a diamagnetic or paramagnetic metal ion that can be radioactive, photoactive metals being preferably selected from Ga 3+ , Pt 2+ , Pd 2+ , S 4+ , In 3+ , Ge 4+ , Si 4+ , Al 3+ , Zn 2+ , Mg 2+ the appropriate number of physiologically acceptable charge balancing counter ions.
  • the metallo-tetrapyrrolic compounds of the invention can be derived by various procedures from naturally occuring cyclic tetrapyrroles.
  • the naturally occurring cyclic tetrapyrrolic molecules can have the basic ring structure of compounds I, II, III, and IV, whose substituents are outlined in Table 1, and are particularly preferred as starting materials for the synthesis of the compounds of structures I-IV.
  • tetrapyrroles derived from naturally occuring ring systems that have one linking group are particularly preferred. These are shown in scheme 1.
  • the tetrapyrrole is derived by the coupling of suitably substituted dipyrromethane, dipyrromethenes, biladienes, builirubins, pyrroles and functionalized aldehydes, or functionalized maleonitriles.
  • the cyclic tetrapyrroles that have the basic ring structure of compounds I-IV, whose substituents are outlined in Table 2, are particularly preferred as starting materials for the synthesis of compounds of structures I-IV.
  • tetrapyrrole tetrapyrrolic molecule and “porphyrin” are used here to designate compounds of the cyclic structure where four pyrrolic ring systems are linked via either carbon or nitrogen bonds.
  • Compounds within the scope of the invention include porphyrins, mono-, di-, tri- and tetraazaporphyrins, and porphyrin isomers such as porphycenes, isoporphycenes, hemiporphycenes, corroles, corrphycenes, and the like.
  • porphyrins Included in the first class of metallated tetrapyrrolic compounds of the invention are those of the porphyrins.
  • Scheme 1 outlines an example of the synthesis of porphyrins that are derived from plants. Particularly advantageous are the porphyrins based on chloroporphyrin e6 (9), chloroporphyrin e4 (10), phylloporphyrin (11), rhodoporphyrin (7), pyrroporphyrin (8), pheoporphyrin a5 (13) and phylloerythrin (12) and compounds having similar ring systems.
  • Such compounds can be synthesized with single linking groups, which can be modified according to the invention to increase their biological activity and MRI and radiodiagnostic capacity.
  • the propionic esters of (2), (3), (5) and (6) can be selectively hydrolized to form carboxylic acids, which can then be linked to the metal coordinating moiety.
  • the carboxylic acids can be converted to amides with a free amine linking unit, which can then be linked to the metal co-ordinating moiety.
  • Porphyrin amide derivatives like (4) (R 2 ⁇ NHR 3 ) may be synthesized from phylloporphyrin such that an amine linking group is present.
  • Examples include where R 2 ⁇ NHCH 2 CH 2 NH 2 , NHCH 2 CH 2 CH 2 NH 2 , NHCH 2 CH 2 NH 2 , NH(CH 2 ) 2 O(CH 2 ) 2 NH 2 and similar compounds. These amine groups can then be linked to the metal co-ordinating moiety.
  • porphyrins While plant derived porphyrins are preferred as starting materials in the invention due to their abundant availability, a very large number of synthetic porphyrins are generally applicable to the invention.
  • Such porphyrins can be made by synthetic methods known to those skilled in the art, via coupling of pyrrolic precursors, dipyrromethanes, dipyrromethenes and biladienes to give the required porphyrins with widely ranging functionality at both the ⁇ and meso positions.
  • the synthesis of porphyrins via the coupling of pyrrolic intermediates is outlined in detail in chapters 1, 2, 3 in “The Porphyrin Handbook” Editors, K. M. Kadish, K. M. Smith, R. Guilard, Volume 1, Academic press, 2000, p.
  • a second preferred class of compounds according to the invention are the mono-, di-, tri- and tetra-azaporphyrins.
  • Schemes 3-7 outline the synthesis of mono-, di-, and tetra-azaporphyrins, examples of which are listed in Table 2.
  • Schemes 3-7 outline synthetic routes to novel tetrapyrrolic molecules that possess a linking group with terminal amine groups. Such compounds can be linked to a metal complexing reagent (MCR) and subsequently modified to be phototherapeutic and diagnostic compounds.
  • MCR metal complexing reagent
  • Mono-azaporphyrins are synthesized efficiently via the coupling of dibromobiladienes with sodium azide or via the reaction of oxyporphyrins with ammonia. Copper and metal free diazaporphyrins can be obtained via the coupling of 5,5′-dibromopyrromethenes with sodium azide. Tetra-azaporphyrins are synthesized most efficiently via the treatment of substituted maleonitriles with magnesium powder or magnesium alcoxides. Such reactions are well known in the art and are outlined in detail by N. Kobayashi in “The Porphyrin Handbook,” K. M. Kadish, K. M. Smith, R. Guilard, Editors, Volume 2, Chapter 13, Academic Press, 2000, p. 301-360, the disclosure of which is hereby incorporated by reference herein.
  • peripheral functionality of these compounds is important with respect to further derivatization to achieve the desired coupling to the metal complexing reagent (MCR) and the desired biological effect (both therapeutic and diagnostic).
  • MCR metal complexing reagent
  • desired biological effect both therapeutic and diagnostic.
  • the types of peripheral functionality applicable to the invention are described in detail below. It is recognized that small changes in the peripheral functionality can have pronounced effects on the biological efficacy of the molecules as does metal co-ordination to the compounds.
  • Schemes 4-7 outline synthetic routes to the novel tetrapyrrolic molecules of the invention.
  • the new compounds of the invention are based on the porphyrin, mono-, di-, tri- and tetra-azaporphyrin ring systems that bear peripheral functionality on the ring system.
  • Such functionality includes esters, alcohols, amides, amines, ethers, and phosphates.
  • Such derivatives may also have at least one hydroxylated residue present, or an amine group with which to couple the metal co-ordination compound.
  • the new porphyrins themselves may be photodynamically active as metal free analogs and therefore useful as PDT agents.
  • metallated derivatives of these compounds are also of particular interest for treatment and diagnosis of disorders of the cardiovascular system, normal or abnormal conditions of the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system including bone, connective tissue, cartilage and skeletal muscle, pulmonary system, gastrointestinal system including the liver, reproductive system, skin, immune system, cardiovascular system, urinary system, ocular system, auditory or olfactory system, where shorter wavelengths of light are necessary or advantageous to effect a desired therapy.
  • Scheme 4 outlines chemistry that has been undertaken to produce photosensitizing or diagnostic agents (based on non-naturally occuring porphyrin systems and azaporphyrins) that possess pendant terminal amine moieties and is exemplary only and is not intended to limit the invention. It should be noted that the functionality and position of the N and C meso atoms can be varied to produce analogs different from those shown. Additionally, the R groups in these schemes constitute functional groups that can be symmetrically substituted and can also, if desired, be modified by techniques known to those skilled in the art based on the chemistry described herein without departing from the spirit or scope of the invention.
  • the ester functionality of porphyrins or azaporphyrins can be hydrolyzed to yield both mono- and di-acid compounds. It is preferred that the synthesis of mono-acid compounds via this method occurs on compounds that are symmetrical in their substitution pattern of R 1 -R 4 , such that isomers are not formed.
  • the mono- or di-acid can then be converted to the desired amide via standard techniques, to produce pendant arm groups with terminal amine moieties.
  • R 5 possesses a functional group that can be modified to produce a reactive linking moiety (for example C 6 H 4 SO 3 H, C 6 H 4 CO 2 H and the like), these may alternatively be chemically modified to produce compounds with pendant arm groups having terminal amine moieties. Such compounds can, if desired, be reacted with: metals to produce metallotetrapyrrolic complexes.
  • Scheme 6 outlines the synthesis route of metal-free or metallated mono- or di-amine porphyrins based on rhodoporphyrin or pyrroporphyrin.
  • rhodoporphyrin R ⁇ CO 2 Me
  • both groups can be hydrolyzed with KOH/H 2 O to give the di-acid derivative.
  • the acid groups can then be converted to the acid chloride derivatives and reacted with the appropriate amine to give compounds possessing one or two pendant arm terminal amine moieties. These compounds can, if desired, be reacted with metals to produce metallo-tetrapyrrolic mono or di-amine complexes.
  • Scheme 7 outlines the synthesis of metal-free or metallated mono or di-amine di-azaporphyrins.
  • the di-azaporphyrins themselves can be synthesized via the coupling of appropriate brominated dipyrromethanes.
  • the peripheral functionality can be modified by similar chemistry as outlined in schemes 4 and 5.
  • R 1 -R 4 should possess a symmetrical substitution pattern. This preference does not apply if di-amine substitution is desired.
  • the introduction of the desired metals (e.g., Zn, Ga, Al, Sn, In, Mg, Mn, Fe, etc) into the porphyrins or azaporphyrins can be carried out according to methods that are known in the literature (e.g., The Porphyrins, ed. D. Dolphin, Academic Press, New York 1980, Vol. V, p. 459; DE 4232925).
  • metal substitution of pyrrolic NH's can be carried out by heating the metal-free ligand with the corresponding metal salt, preferably acetate or halide, optionally with the addition of acid-buffering agents, such as, for example, sodium acetate in a polar solvent.
  • substitutions can be carried out by metal exchange in which a metal that is already complexed by the porphyrin or azaporphyrin is displaced by the desired metal.
  • a metal is cadmium.
  • the preferred solvent is a polar solvent, such as, for example, methanol, glacial acetic acid, dimethylformamide, chloroform or water.
  • the metal is difficult to remove under acid conditions (Pt, Pd)
  • the introduction of a diamagnetic or paramagnetic metal M into the porphyrin system can be carried out before or after linkage of the metal complexing agent radical (MCR).
  • MCR metal complexing agent radical
  • MCA metal complexing agent
  • an activated MCR can be reacted with the amine porphyrin or azaporphyrin derivatives such that a covalent link between the two compounds occurs.
  • the nature of the linking amine moiety on the porphyrins or azaporphyrins (P) to the MCR compounds may be varied.
  • Preferable examples include: P-(CH 2 ) n NH 2 , where n is an integer from 1 to 10; P-(aryl)NH 2 , P-CONHNH 2 , or P-(CH 2 ) n CONHNH 2 , where n is an integer from 1 to 10; P-CONH(CH 2 ) n NH 2 , where n is an integer from 1 to 10; P-CONH(CH 2 ) 2 O(CH 2 ) 2 NH 2 , or P-CONH[(CH 2 ) 2 ] n O n/2 [(CH 2 ) 2 ] n NH 2 , where n is an integer from 1 to 10; P-SO 2 NHNH 2 , or P-SO 2 NH(CH 2 ) n NH 2 , where n is an integer from 1 to 10; P-SO 2 NH(CH 2 ) 2 O(CH 2 ) 2 NH 2 , or P-SO 2 NH[(CH 2 ) 2 ] n
  • porphyrin and azaporphyrin units can be generated that possess alcohol terminal linking groups.
  • Preferred examples of such groups include P-(CH 2 ) n OH, where n is an integer from 1 to 10; P-(aryl)OH, P-CONHOH, or P-(CH 2 ) n CONHOH, where n is an integer from 1 to 10; P-CONH(CH 2 ) n OH, where n is an integer from 1 to 10; P-CONH(CH 2 ) 2 O(CH 2 ) 2 OH, or P-CONH[(CH 2 ) 2 ] n O n/2 [(CH 2 ) 2 ] n OH, where n is an integer from 1 to 10; P-SO 2 NHOH, or P-SO 2 NH(CH 2 ) n OH, where n is an integer from 1 to 10; P-SO 2 NH(CH 2 ) 2 O(CH 2 ) 2 OH, or P-SO 2
  • the MCR linking group Q is an organic group that when linked to the amine or alcohol porphyrin or azaporphyrin results in a product that is an ester, an amide, an amine, an ether, or a thiolate.
  • Preferable reactive MCR's are described in, for example, U.S. Pat. No. 4,885,363, U.S. Pat. No. 5,730,956, U.S. Pat. No. 6,136,841, and U.S. Pat. No. 5,275,801 the disclosures which are hereby incorporated herein by reference. Examples are illustrated in FIG. 2.
  • linker groups Q include —CO—, —CS—, —COCH 2 NH—, —CO(CH 2 ) 2 NH, —CO(CH 2 ) 2 —, —COCH 2 OC 6 H 4 CO, —COC 6 H 4 NH—, —COCH 2 OCH 2 NH—, —COC 6 H 4 —, —COCH 2 NHCOCH 2 CH(CH 2 COOH)C 6 H 4 NH—, phenyleneoxy, a C1-C12 alkylene or a C7-C12 aralkylene group by one or more oxygen atoms.
  • the porphyrin or azaporphyrin linked MCR compounds can then be modified to produce PDT/MRI radiodiagnostic compounds.
  • paramagnetic metal ions must be present in the complex. These are preferably divalent or trivalent ions of the elements of atomic numbers 21-29, 42-44 and 57-71.
  • the paramagnetic metal may be coordinated to either the inner pyrrolic core of the porphyrin or azaporphyrin, or in the MCR pendant arm, or in both.
  • Suitable ions include, for example, chromium, gadolinium, dysprosium, manganese, iron, cobalt, cobalt, nickel, copper, praseodymium, neodymium, samarium, terbium, holmium, erbium and ytterbium ions. Because of their high magnetic moment, the gadolinium, dysprosium, manganese, terbium, holmium, erbium and iron ions are especially preferred.
  • the metal ions must be radioactive.
  • radioisotopes of the elements copper, cobalt, gallium, zinc, germanium, yttrium, strontium, technetium, indium, ytterbium, gadolinium, samarium, thallium, and iridium.
  • the radioactive isotope may be coordinated to the porphyrin or azaporphyrin ligand or within the MCR, or both.
  • Metal chelation to the MCR group can be carried out by techniques known in the literature (see, e.g., DE3401052) by the metal oxide or metal salt (e.g., nitrate, acetate, carbonate, chloride or sulfate) of the metal that is desired.
  • the metal oxide or salt can be suspended or dissolved in polar solvents such as water or aqueous alcohols and then reacted with the corresponding amount of the complexing ligand.
  • acidic hydrogen atoms or acid groups that are present can be substituted by cations of inorganic and/or organic bases or amino acids.
  • neutralization can be carried out with the aid of inorganic bases, such as, e.g., alkali or alkaline-earth hydroxides, carbonates or bicarbonates and/or organic bases such as, for example, primary, secondary and tertiary amines, such as, e.g., ethanolamine, morpholine, glucamine, N-methyl- and N,N-dimethylglucamine, as well as basic amino acids, such as, e.g., lysine, arginine and ornithine or amides of originally neutral or acidic amino acids.
  • inorganic bases such as, e.g., alkali or alkaline-earth hydroxides, carbonates or bicarbonates and/or organic bases
  • organic bases such as, for example, primary, secondary and tertiary amines, such as, e.g., ethanolamine, morpholine, glucamine, N-methyl- and N,N-dimethylglucamine, as
  • a sufficient amount of the desired bases can be added to, for example, the acidic complex salts in aqueous solution or suspension to ensure that the neutral point is reached.
  • the solution that is obtained can then be evaporated to the dry state in a vacuum.
  • water-miscible solvents such as, for example, lower alcohols (e.g., methanol, ethanol, isopropanol, acetonitrile), lower ketones (e.g., acetone), polar ethers (e.
  • the acidic complex compounds contain several free acid groups, it is often advantageous to produce neutral mixed salts that contain both inorganic and organic cations as counter-ions.
  • This can be achieved, for example, by reacting the complexing ligands in aqueous suspension or solution with the oxide or salt of the element that yields the central ion and half of the amount of an organic base that is required for neutralization.
  • the complex salt that is formed can then be isolated, optionally purified, and then mixed for complete neutralization with the required amount of inorganic base.
  • the sequence in which the base is added can also be reversed.
  • Another way of obtaining neutral complex compounds consists of converting the remaining acid groups in the complex completely or partially into esters. This can be achieved by subsequent reaction on the finished complex, e.g., by exhaustive reaction of free carboxy groups with dimethylsulfate.
  • Pharmaceutical agents of the invention can be produced by adding to the complex compounds of the invention certain additives that are commonly used in the pharmaceutical industry to suspend or dissolve the compounds in an aqueous medium, and then the suspension or solution can be sterilized by techniques known in the art.
  • suitable additives include, for example, physiologically harmless buffers (such as, e.g., trimethamine), small additions of complexing agents (such as, e.g., diethylenetriaminepentaacetic acid) or, if necessary, electrolytes such as, e.g., sodium chloride or antioxidants such as, e.g., ascorbic acid, butylate hydroxy toluene, or tocopherol.
  • suspensions or solutions of the agents according to the invention in water or in physiological salt solution are desired for enteral administration or other purposes, they can be mixed with one or more adjuvants that are commonly used in galenicals (e.g., methylcellulose, lactose, mannitol) and/or surfactant(s) (e.g., lecithins, Tween, and/or flavoring substances for taste correction (e.g., ethereal oils).
  • adjuvants e.g., methylcellulose, lactose, mannitol
  • surfactant(s) e.g., lecithins, Tween, and/or flavoring substances for taste correction (e.g., ethereal oils).
  • the compounds and agents according to the invention should be stored and handled as much as possible in a light-free environment.
  • the pharmaceutical agents according to the invention preferably contain from about 20 ⁇ mol/L to about 200 mmol/L of the complex salt and are generally dosed in amounts of 0.01 ⁇ mol to 2 mmol/kg of body weight, both for their use in PDT and for therapy monitoring using MRI diagnosis. They are intended for enteral and parenteral administration or are administered with the methods of interventional radiology.
  • the agents according to the invention are especially suitable for PDT and as MRI contrast media. After administration, they can enhance the informational value of the image that is obtained from a nuclear spin tomograph by increasing the signal intensity. They are effective without burdening the body with large amounts of foreign substances.
  • the high water-solubility of the agents according to the invention allows the production of highly concentrated solutions, so as to keep the volume burden of the circulation within acceptable limits and to compensate for dilution by bodily fluid.
  • the agents according to the invention show not only a high stability in vitro but also a surprisingly high stability in vivo, so that a release or an exchange of the ions, which are inherently toxic and not covalently bonded in the complexes, will not be harmful within the time that it takes for the contrast media to be completely excreted.
  • any porphyrinic molecule may be modified according to the invention to form the desired photoactive compounds with widely differing functionality as described in the literature (for example see “Porphyrins and Metalloporphyrins” Ed. K. Smith, Elsevier, 1975, N.Y., “The Porphyrins”, Ed. D. Dolphin, Vol I-V, Academic Press, 1978, and “The Porphyrin Handbook”, Eds. K. Kadish, K. M. Smith, R. Guilard, Academic Press, 2000). These compounds contain various and ranging substituents on the ⁇ -pyrrole positions or meso-positions of the porphyrin ring, either symmetrically or asymmetrically substituted on the ring.
  • Examples of such functionality include functional groups having a molecular weight less than about 100,000 daltons and can be a biologically active group or organic. Examples include, but are not limited to: (1) hydrogen; (2) halogen, such as fluoro, chloro, iodo and bromo (3) lower alkyl, such as methyl, ethyl, n-propyl, butyl, hexyl, heptyl, octyl, isopropyl, t-butyl, n-pentyl and the like groups; (4) lower alkoxy, such as methoxy, ethoxy, isopropoxy, n-butoxy, t-pentoxy and the like; (5) hydroxy; (6) carboxylic acid or acid salts, such as —CH 2 COOH, —CH 2 COONa, —CH 2 CH 2 COOH, —CH 2 CH 2 COONa, —CH 2 CH 2 CH(Br)COOH, —CH 2 CH 2 CH(CH
  • biologically active group can be any group that selectively promotes the accumulation, elimination, binding rate, or tightness of binding in a particular biological environment.
  • one category of biologically active groups is the substituents derived from sugars, specifically: (1) aldoses such as glyceraldehyde, erythrose, threose, ribose, arabinose, xylose, lyxose, allose, altrose, glucose, mannose, gulose, idose, galactose, and talose; (2) ketoses such as hydroxyacetone, erythrulose, rebulose, xylulose, psicose, fructose, sorbose, and tagatose; (3) pyranoses such as glucopyranose; (4) furanoses such as fructo-furanose; (5) O-acyl derivatives such as penta-O-acetyl- ⁇ -glucose;
  • Amino acid derivatives are also useful biologically active substituents, such as those derived from valine, leucine, isoleucine, threonine, methionine, phenylalanine, tryptophan, alanine, arginine, aspartic acid, cystine, cysteine, glutamic acid, glycine, histidine, proline, serine, tyrosine, asparagine and glutamine.
  • peptides particularly those known to have affinity for specific receptors, for example, oxytocin, vasopressin, bradykinin, LHRH, thrombin and the like.
  • nucleosides for example, ribonucleosides such as adenosine, guanosine, cytidine, and uridine; and 2′-deoxyribonucleosides, such as 2′-deoxyadenosine, 2′-deoxyguanosine, 2′-deoxycytidine, and 2′-deoxythymidine.
  • ribonucleosides such as adenosine, guanosine, cytidine, and uridine
  • 2′-deoxyribonucleosides such as 2′-deoxyadenosine, 2′-deoxyguanosine, 2′-deoxycytidine, and 2′-deoxythymidine.
  • ligand specific for a biological receptor refers to a moiety that binds a receptor at cell surfaces, and thus contains contours and charge patterns that are complementary to those of the biological receptor.
  • the ligand is not the receptor itself, but a substance complementary to it. It is well understood that a wide variety of cell types have specific receptors designed to bind hormones, growth factors, or neurotransmitters. However, while these embodiments of ligands specific for receptors are known and understood, the phrase “ligand specific for a biological receptor”, as used herein, refers to any substance, natural or synthetic, that binds specifically to a receptor.
  • ligands examples include: (1) the steroid hormones, such as progesterone, estrogens, androgens, and the adrenal cortical hormones; (2) growth factors, such as epidermal growth factor, nerve growth factor, fibroblast growth factor, and the like; (3) other protein hormones, such as human growth hormone, parathyroid hormone, and the like; (4) neurotransmitters, such as acetylcholine, serotonin, dopamine, and the like; and (5) antibodies. Any analog of these substances that also succeeds in binding to a biological receptor is also included within the invention.
  • the steroid hormones such as progesterone, estrogens, androgens, and the adrenal cortical hormones
  • growth factors such as epidermal growth factor, nerve growth factor, fibroblast growth factor, and the like
  • other protein hormones such as human growth hormone, parathyroid hormone, and the like
  • neurotransmitters such as acetylcholine, serotonin, dopamine, and the like
  • antibodies Any analog of
  • substituents tending to increase the amphiphilic nature of the compounds include, but are not limited to: (1) short or long chain alcohols, such as, for example, —C 12 H 24 —OH; (2) fatty acids and their salts, such as, for example, the sodium salt of the long-chain fatty acid oleic acid; (3) phosphoglycerides, such as, for example, phosphatidic acid, phosphatidyl ethanolamine, phosphatidyl choline, phosphatidyl serine, phosphatidyl inositol, phosphatidyl glycerol, phosphatidyl 3′-O-alanyl glycerol, cardiolipin, or phosphatidyl choline; (4) sphingolipids, such as, for example, sphingomyelin; and (5) glycolipids, such as, for example, glycosyldiacylglycerols, cerebrosides, sulfate
  • the compounds of the present invention can be administered to the host in a variety of forms adapted to the chosen route of administration, e.g., orally, intravenously, topically, intramuscularly or subcutaneously.
  • the active compound may be orally administered, for example with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets, or it may be incorporated directly with food.
  • the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least about 0.1% of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may, for example, conveniently be between about 2 to about 60% of the weight of the administered product.
  • the amount of active compound in such therapeutically useful compositions is can be selected so that a suitable dosage will be obtained.
  • Preferred compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains between about 50 and 300 mg of active compound.
  • the tablets, troches, pills, capsules and the like may also contain the following: a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; a sweetening agent such as sucrose, lactose or saccharin; or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring.
  • a binder such as gum tragacanth, acacia, corn starch or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin
  • a flavoring agent such as peppermint, oil of winter
  • tablets, pills, or capsules may be coated with shellac, sugar or both.
  • a syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor.
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and formulations.
  • the active compound may also be administered parenterally or intraperitoneally.
  • Solutions of the active compound as a free base or pharmacologically acceptable salt can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporanous preparation of sterile injectable solutions, dispersions, or liposomal or emulsion formulations.
  • the form must be sterile and should be fluid to enable administration by a syringe.
  • the form must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required additional ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and the freeze-drying technique, which yield a powder of the active ingredient plus any additional desired ingredient from previously sterile-filtered solutions thereof.
  • the new compounds of the invention may also be applied directly to tumors in the host whether internal or external, in topical compositions.
  • exemplary compositions include solutions of the new compounds in solvents, particularly aqueous solvents, most preferably water.
  • the present new compounds may be dispersed in the usual cream or salve formulations commonly used for this purpose (such as liposomes, ointments, gels, hydrogels, cremes and oils) or may be provided in the form of spray solutions or suspensions that may include a propellant usually. employed in aerosol preparations.
  • “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Any conventional media or agent that is compatible with the active ingredient can be used in the therapeutic compositions of the invention. Supplementary active ingredients can also be incorporated into the compositions.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated. Each unit contains a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specifications for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of cardiovascular diseases, diseases of the skin, cancers and other superficial disease conditions in living subjects.
  • the present invention provides a method of treating live cells, which includes, but is not limited to, animals such as humans and other mammals.
  • animals such as humans and other mammals.
  • the “mammals” also include farm animals, such as cows, hogs and sheep, as well as pet or sport animals, such as horses, dogs and cats.
  • the dosage of the pharmaceutical compositions of the invention is dependent on the method of administration, the patient's age, severity of the disease, and the like.
  • the compounds of the invention may be taken parentally or orally, generally being administered intravascularly, subcutaneously, or intramuscularly or interperitoneally.
  • the subject compounds may also be administered by inhalation, perivascular delivery, pericardial delivery (into perivascular sac), periadvential delivery (e.g., using a hydrogel wrap around the vessel), endovascular balloon catheters with micropores, channels, transmural injection ports, and the like.
  • an infusate can be placed and pressurized to facilitate intramural and transmural penetration into the target vessel.
  • Local delivery can also be enhanced by other mechanical and electrical means.
  • the depth of the penetration of the subject compounds by this local delivery method is a function of pressure in the perfused segment and the dwell time.
  • Delivery of the compounds of the invention may also be via antibody-drug conjugates, internalizing antibodies or antibody fragments conjugated to compounds into cells using endocytosis.
  • the subject compounds may also be impregnated into stent struts for local delivery.
  • the route of administration of these pharmaceutical preparations is not critical, but may be selected according to the dosage form, the patient's age, the severity of the disease to be treated and other factors.
  • the compounds of the invention may find use in conjunction with other interventions, diagnostics and therapies, where lower levels of other therapies having significant side effects may be used effectively to reduce the detrimental side effects.
  • Adjunctive interventions may include, but are not limited to: balloon angioplasty, invasive and non-invasive surgical procedures, stent deployment, cutting balloons, embolic protection devices, rotational and directional atherectomy, eximer lasers and the like.
  • Adjunctive therapies may include, but are not limited to radiation therapy, chemotherapy, anti-platelet agents, vasodilators, antihypertensives, anti-arrhythmics, hyperthermia, cryotherapy, magnetic force, viral and non-viral gene therapy, pharmacogenetic therapy, antibodies, vaccines, glycoprotein IIb/IIIa Inhibitors, growth factors, peptides, DNA delivery, nucleic acids, anticancer drugs, steroid hormones, anti-inflammatories, proteins, anti-apoptotic therapies, anti-sense agents, immunotoxins, immunomodulators, antibody-drug conjugates, anti-proliferative therapies, drug eluting stents containing pharmacologically active agents, transplant products and processes, prostaglandins and catheter based devices to detect vulnerable plaques, hormone products, chelating agents, diuretics, cardiac glycosides, bronchodilators, antibiotics, antivirals, antitioxins, cyclosporins, thrombolytic agents, interferon
  • Adjunctive diagnostics may include, but are not limited to: intravascular ultrasound imaging, angiography, quantitative vessel measurements and the use of radiological contrast agents, hormone products, chelating agents, diuretics, cardiac glycosides, bronchodilators, antibiotics, antivirals, antitoxins, cyclosporins, thrombolytic agents, interferons, blood products such as parental iron and hemin, anti-fungal agents, antianginals, anticoagulants, analgesics, narcotics, neuromuscular blockers, sedatives, bacterial vaccines, viral vaccines, DNA or RNA of natural or synthetic origin including recombinent RNA and DNA, cytokines and their antagonists/inhibitors, and chemokines and their antagonists/inhibitors.
  • alkyl refers to substituted or unsubstituted, straight or branched chain groups, preferably having one to twenty, more preferably having one to six, and most preferably having from one to four carbon atoms.
  • C 1 -C 20 alkyl represents a straight or branched alkyl chain having from one to twenty carbon atoms.
  • Exemplary C 1 -C 20 alkyl groups include methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, t-butyl, pentyl, neo-pentyl, hexyl, isohexyl, and the like.
  • C 1 -C 20 alkyl includes within its definition the term “C 1 -C 4 alkyl.”
  • Such alkyl groups may themselves be ethers or thioethers, or aminoethers or dendrimers.
  • cycloalkyl represents a substituted or unsubstituted, saturated or partially saturated, mono- or poly-carbocyclic ring, preferably having 5-14 ring carbon atoms.
  • exemplary cycloalkyls include monocyclic rings having from 3-7, preferably 3-6, carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
  • An exemplary cycloalkyl is a C 5 -C 7 cycloalkyl, which is a saturated hydrocarbon ring structure containing from five to seven carbon atoms.
  • aryl refers to an aromatic, monovalent monocyclic, bicyclic, or tricyclic radical containing 6, 10, 14, or 18 carbon ring atoms, which may be unsubstituted or substituted, and to which may be fused one or more cycloalkyl groups, heterocycloalkyl groups, or heteroaryl groups, which themselves may be unsubstituted or substituted by one or more suitable substituents.
  • aryl groups include, but are not limited to, phenyl, napthalenes, anthracenes, benzopyrenes, quinolines, benzoquinolines, benzoperylene, benzofluorenes, fluorenes, benzofurazans, benzodiphenylenes, benzofluoranthenes, benzanthracenes, benzacephenanthrylenes, bathophenanthrolines, indans, benzoquinolines, quinolines, pyrazines, quinolines, quinazoles, quinoxalines, imidazopyridines, indenes, indolines, thiazolines, benzopyrimidines, pyrimidines, benzimidazole, triazolopyrimidines, pyrazoles, tryptophans, phenanthrolines, benzooxadiazoles, benzoselenadiazole, benzocoumarins, chalcones, fluoranthenes, pyr
  • halogen represents chlorine, fluorine, bromine or iodine.
  • halocarbon or “haloalkyl” represents one or more halogens bonded to a one or more carbon bearing groups.
  • heterohaloalkyl represents for example halogenated alkylethers, halogenated alkyl amines, halogenated alkyl esters, halogenated alkyl amides, halogenated alkyl thioesters, halogenated alkyl thiols, where N, S, O, P atoms are present in the haloalkylated structure.
  • heteroalkyl represents for example ethers, alkylamines, alkylated thiols and alkylate phosphorus containing groups.
  • carrier represents a substituted or unsubstituted aromatic or a saturated or a partially saturated 5-14 membered monocyclic or polycyclic ring, such as a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring, wherein all the ring members are carbon atoms.
  • electronegative group is intended to mean a chemical group containing an electronegative element such as halogen, sulfur, nitrogen or oxygen.
  • a “heterocycloalkyl group” is intended to mean a non-aromatic, monovalent monocyclic, bicyclic, or tricyclic radical, which is saturated or unsaturated, containing 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 ring atoms, and which includes 1, 2, 3, 4, or 5 heteroatoms selected from nitrogen, oxygen and sulfur, wherein the radical is unsubstituted or substituted, and to which may be fused one or more cycloalkyl groups, aryl groups, or heteroaryl groups, which themselves may be unsubstituted or substituted.
  • heterocycloalkyl groups include, but are not limited to azetidinyl, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, tetrahydro-2H-1,4-thiazinyl, tetrahydrofuryl, dihydrofuryl, tetrahydropyranyl, dihydropyranyl, 1,3-dioxolanyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-oxathiolanyl, 1,3-oxathianyl, 1,3-dithianyl, azabicylo[3.2.1]octyl, azabicylo[3.3.1]nonyl, azabicylo[4.3.0]nonyl, oxabicylo[2.2.1]heptyl, 1,5,9-triazacyclododecyl, and the like.
  • heteroaryl group is intended to mean an aromatic monovalent monocyclic, bicyclic, or tricyclic radical containing 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 ring atoms, including 1, 2, 3, 4, or 5 heteroatoms selected from nitrogen, oxygen and sulfur, which may be unsubstituted or substituted, and to which may be fused one or more cycloalkyl groups, heterocycloalkyl groups, or aryl groups, which themselves may be unsubstituted or substituted.
  • heteroaryl groups include, but are not limited to, thienyl, pyrrolyl, imidazolyl, pyrazolyl, furyl, isothiazolyl, furazanyl, isoxazolyl, thiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, benzo[b]thienyl, naphtho[2,3-b]thianthrenyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathienyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, quinoxyalinyl, quinzolinyl, benzothiazolyl, benzimidazolyl, te
  • leaving group refers to any group that departs from a molecule in a substitution reaction by breakage of a bond.
  • Examples of leaving groups include, but are not limited to, halides, tosylates, arenesulfonates, alkylsulfonates, and triflates.
  • Suitable protecting groups are recognizable to those skilled in the art. Examples of suitable protecting groups can be found in T. Green & P. Wuts, Protective Groups in Organic Synthesis (2d ed. 1991), which is hereby incorporated by reference herein in its entirety.
  • Suitable salt anions include, but are not limited to, inorganics such as halogens, pseudohalogens, sulfates, hydrogen sulfates, nitrates, hydroxides, phosphates, hydrogen phosphates, dihydrogen phosphates, perchlorates, and related complex inorganic anions; and organics such as carboxylates, sulfonates, bicarbonates and carbonates.
  • inorganics such as halogens, pseudohalogens, sulfates, hydrogen sulfates, nitrates, hydroxides, phosphates, hydrogen phosphates, dihydrogen phosphates, perchlorates, and related complex inorganic anions
  • organics such as carboxylates, sulfonates, bicarbonates and carbonates.
  • substituents for alkyl and aryl groups include mercapto, thioether, nitro (NO 2 ), amino, aryloxyl, halogen, hydroxyl, alkoxyl, and acyl, as well as aryl, cycloalkyl and saturated and partially saturated heterocycles.
  • substituents for cycloalkyl groups include those listed above for alkyl and aryl, as well as alkyl.
  • Exemplary substituted aryls include a phenyl or naphthyl ring substituted with one or more substituents, preferably one to three substituents, independently selected from halo, hydroxy, morpholino(C 1 -C 20 )alkoxy carbonyl, pyridyl (C 1 -C 20 )alkoxycarbonyl, halo (C 1 -C 20 )alkyl, C 1 -C 20 alkyl, C 1 -C 20 alkoxy, carboxy, C 1 -C 20 alkocarbonyl, carbamoyl, N-(C 1 -C 20 )alkylcarbamoyl, amino, C 1 -C 20 alkylamino, di(C 1 -C 20 )alkylamino or a group of the formula —(CH 2 ) a —R 7 where a is 1, 2, 3, 4, 5; and R 7 is hydroxy, C 1 -C 20 alkoxy, carboxy, C 1
  • halo(C 1 -C 20 )alkyl which represents a straight or branched alkyl chain having at least one halogen atom attached to it.
  • exemplary halo(C 1 -C 20 )alkyl groups include chloromethyl, 2-bromoethyl, 1-chloroisopropyl, 3-fluoropropyl, 2,3-dibromobutyl, 3-chloroisobutyl, trifluoromethyl, trifluoroethyl, and the like.
  • hydroxy (C 1 -C 20 )alkyl which represents a straight or branched alkyl chain having from one to twenty carbon atoms with a hydroxy group attached to it.
  • exemplary hydroxy(C 1 -C 20 )alkyl groups include hydroxymethyl, 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxyisopropyl, 4-hydroxybutyl, and the like.
  • C 1 -C 20 alkylthio(C 1 -C 20 )alkyl is a straight or branched C 1 -C 20 alkyl group with a C 1 -C 20 alkylthio group attached to it.
  • Exemplary C 1 -C 20 alkylthio(C 1 -C 20 )alkyl groups include methylthiomethyl, ethylthiomethyl, propylthiopropyl, sec-butylthiomethyl, and the like.
  • heterocycle(C 1 -C 20 )alkyl is a straight or branched alkyl chain having from one to twenty carbon atoms with a heterocycle attached to it.
  • exemplary heterocycle(C 1 -C 20 )alkyls include pyrrolylmethyl, quinolinylmethyl, 1-indolylethyl, 2-furylethyl, 3-thien-2-ylpropyl, 1-imidazolylisopropyl, 4-thiazolylbutyl and the like.
  • aryl(C 1 -C 20 )alkyl which is a straight or branched alkyl chain having from one to twenty carbon atoms with an aryl group attached to it.
  • exemplary aryl(C 1 -C 20 )alkyl groups include phenylmethyl, 2-phenylethyl, 3-naphthyl-propyl, 1-naphthylisopropyl, 4-phenylbutyl and the like.
  • the heterocycloalkyls and the heteroaryls can, for example, be substituted with 1, 2 or 3 substituents independently selected from halo, halo(C 1 -C 20 )alkyl, C 1 -C 20 alkyl, C 1 -C 20 alkoxy, carboxy, C 1 -C 20 alkoxycarbonyl, carbamoyl, —(C 1 -C 20 )alkylcarbamoyl, amino, C 1 -C 20 alkylamino, di(C 1 -C 20 )alkylamino or a group having the structure —(CH 2 ) a —R 7 where a is 1, 2, 3, 4, 5 and R 7 is hydroxy, C 1 -C 20 alkoxy, carboxy, C 1 -C 20 alkoxycarbonyl, amino, carbamoyl, C 1 -C 20 alkylamino or di(C 1 -C 20 )alkylamino.
  • substituents independently selected from halo
  • substituted heterocycloalkyls include, but are not limited to, 3-N-t-butyl carboxamide decahydroisoquinolinyl and 6-N-t-butyl carboxamide octahydro-thieno[3,2-c]pyridinyl.
  • substituted heteroaryls include, but are not limited to, 3-methylimidazolyl, 3-methoxypyridyl, 4-chloroquinolinyl, 4-aminothiazolyl, 8-methylquinolinyl, 6-chloroquinoxalinyl, 3-ethylpyridyl, 6-methoxybenzimidazolyl, 4-hydroxyfuryl, 4-methylisoquinolinyl, 6,8-dibromoquinolinyl, 4,8-dimethylnaphthyl, 2-methyl-1,2,3,4-tetrahydroisoquinolinyl, N-methyl-quinolin-2-yl, 2-t-butoxycarbonyl-1,2,3,4-isoquinolin-7-yl and the like.
  • a “pharmaceutically acceptable solvate” is intended to mean a solvate that retains the biological effectiveness and properties of the biologically active components of the inventive compounds.
  • Examples of pharmaceutically acceptable solvates include, but are not limited to, compounds prepared using water, isopropanol, ethanol, DMSO, and other excipients generally reffered to as GRAS ingredients.
  • the compounds of the inventive methods may exist in different polymorph forms, such as stable and metastable crystalline forms and isotropic and amorphous forms, all of which are intended to be within the scope of the present invention.
  • a “pharmaceutically acceptable salt” is intended to mean those salts that retain the biological effectiveness and properties of the free acids and bases and that are not biologically or otherwise undesirable.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, citrates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates
  • the desired salt may be prepared by any suitable method known to the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, lactic acid, salicylic acid, pyranosidyl acids such as glucuronic acid and galacturonic acid, alpha-hydroxy acids such as citric acid and tartaric acid, amino acids such as aspartic acid and glutamic acid, aromatic acids such as benzoic acid and cinnamic acid, sulfonic acids such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.
  • an inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, ni
  • the desired salt may be prepared by any suitable method known to the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), or an alkali metal or alkaline earth metal hydroxide or the like.
  • suitable salts include organic salts derived from amino acids such as glycine and arginine; ammonia; primary, secondary and tertiary amines; cyclic amines such as piperidine, morpholine and piperazine; and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • shorter wavelength light may be equally important in other PDT applications that only require short wavelength excitation to effect a therapy.
  • Such applications may be, for example, in hollow organ disease (for example lung cancers, barrets esophagus), or in diseases of the skin (for example psoriasis, actinic keratosis, acne vulgaris).
  • the invention disclosed herein describes the synthesis of metallated photosensitizers having ring systems that have shown excellent efficacy in advanced animal model systems as well as preferred uptake in the target tissue, with excellent clearance characteristics and low toxicity.
  • the compounds of the invention are intended for use not only for effective photodynamic therapy treatment but also as MRI and radiodiagnostic diagostic agents. Such compounds may be used to diagnose, locate or treat cardiovascular disease and normal or abnormal conditions of the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system including bone, connective tissue, cartilage and skeletal muscle, pulmonary system, gastrointestinal system including the liver, reproductive system, skin, immune system, cardiovascular system, urinary system, ocular system, auditory or olfactory system.
  • photoactive derivatives of porphyrins and azaporphyrins are particularly advantageous where shorter wavelengths of light are necessary to effect a photodynamic response.
  • the azaporphyrin synthesized in example 13 was modified according to H. Fischer, E. Haarer and F. Stadler, Z. Physiol. Chem. 241, 209 (1936) by treatment with hydrazine hydrate (1.5 mL of an 80% water solution) in pyridine (20 mL) at room temperature. The solution was evaporated to dryness, dissolved/suspended in methanol (10 mL) and water (30 mL) was added. The methanol was removed by rotary evaporation and the precipitated porphyrin collected by filtration and dried. Yield of title compound: 250 g of a purple powder.
  • the azaporphyrin prepared in example 14 (100 mg) was metallated according to example 2. Yield of title compound 110 mg.
  • the dimethyl ester azaporphyrin of example 14 (1.0 g, 1.6 mmol) is dissolved in THF (200 ml) and a solution of KOH (100 mg)/MeOH (5 mL) is added. The solution is closely monitored until only a trace of starting material remains and the major product is the mono-hydrolyzed azaporphyrin. Acetic acid (107 mg) is added and the solution diluted with water (100 mL). The THF was removed by rotary evaporation and the precipitated azaporphyrins were collected and washed with methanol (20 mL) and dried.
  • the monoacid azaporphyrin prepared in example 30 (500 mg) is dissolved in dichloromethane (50 ml) and oxalyl chloride (5 ml) added. The solution is refluxed under dry conditions for 2 hrs and then evaporated to dryness, care being taken not to expose the crude material to moisture. The residue is dissolved in dichloromethane (50 mL, dry) and ethylene diamine (3 ml, dry) added all at once. The resulting solution is washed with a saturated sodium bicarbonate solution, followed by water and the organic layer is collected, dried over sodium sulfate, filtered and evaporated.
  • the crude reaction mixture is chromatographed on silica using 4% methanol/dichloromethane/0.5% triethylamine as eluent, and the major fraction collected.
  • the ligand that is produced by Example 22 (1.0 g, 0.74 mmol) is dissolved in THF (50 mL) and water (350 mL). Sodium hydroxide solution (10 mol) is added and it is stirred overnight at room temperature. After the ester groups have been completely saponified, the THF is removed by rotoevaporation. A pH of 4 is set with concentrated hydrochloric acid. It is evaporated to the dry state in a vacuum. The residue is chromatographed on RP 18 (eluant: H 2 O/tetrahydrofuran/gradient). Yield of title compound: 0.90 g of a reddish-brown powder.
  • the ligand produced by example 36 (200 mg, 0.13 mmol) is dissolved in water (100 mL), and gadolinium chloride (69 mg, 0.26 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium chloride is added, stirring is continued overnight at room temperature. The solvent is drawn off in a vacuum, and the residue is chromatographed on silica gel RP-18 (eluent:water/THF: 0-30%). Yield of title compound 247 mg.
  • This compound was prepared via the method of Neya, S., Hori, H., Imai, K., Konishi, Y. K., Suzuki, H., Shiro, Y., Lizuka, T., Funasaki, N., J. Biochem, 1997, 121, 654.
  • the compound produced by example 48 (200 mg, 0.148 mmol) is dissolved in water (100 mL), and gadolinium chloride (78.2 mg, 0.296 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium

Abstract

Substituted porphyrin and azaporphyrin deviations with various substitutents in the 12- and 17-positions of the prophyrin skeleton as pharmaceutical agents for use in photodynamic therapy, MRI diagnosis, and radiodiagnostics.

Description

    DESCRIPTION OF THE INVENTION
  • 1. Field of the Invention
  • This invention is directed to substituted porphyrin and azaporphyrin derivatives with various substituents in the 13- and 17-positions of the porphyrin skeleton suitable as pharmaceutical agents for use in photodynamic therapy, MRI diagnosis, and radiodiagnostics. The invention is also directed to pharmaceutical agents that contain these compounds, as well as a process for the production of these compounds and agents.
  • 2. Background of the Invention
  • Photodynamic therapy (“PDT”) is a new modality for the treatment of malignancies, diseased tissue, hyperproliferating tissues, pathogens or unwanted normal tissues. PDT involves a localized or systemic administration of a photosensitizing compound followed by exposure of target tissue to photoactivating light. The photoactivating light excites the photosensitizer which, in turn, interacts with singlet oxygen causing the production of cytotoxic oxygen species. The interaction of the cytotoxic oxygen species with tissues in which the photosensitizer is localized causes a modification of the tissue, resulting in a desired clinical effect. The tissue specificity of the resultant phototoxic damage is determined largely, although not entirely, by the relative concentrations of the photosensitizer in each tissue at the time of its exposure to the photoactivating light.
  • Following systemic administration, many photosensitizers accumulate to varying degrees within tissues depending on the pharmacokinetic and distribution profile of the photosensitizing compound and the cell types comprising the tissues. The chemical factors that enable certain photosensitizers to accumulate to a greater degree at a target site than other photosensitizers is not well understood. Indeed, the biological factors that result in the preferential uptake of some photosensitizers in certain tissue types compared to other tissue types are not well understood either. It is clear, however, that each photosensitizer has its own distribution and pharmacokinetic properties within different tissues and these properties determine the relative usefulness of the photosensitizer for the desired therapy. Currently, rigorous screening and biological evaluation in appropriate model systems is required to identify suitable photosensitizers that display the characteristics necessary to effect a therapy within the diseased or target tissues.
  • Porphyrins and azaporphyrins and their metallated derivatives belong to a family of substances that are suitable for PDT. These compounds accumulate in target tissues and absorb light in a range in which living tissue is still fairly permeable, namely between 380-680 nm. Moreover, porphyrins, azaporphyrins and their photoactive metallated derivatives exhibit high yields of the excited triplet state, a long lifetime in this state, and good energy transfer to oxygen with concomittent production of singlet oxygen. Of the porphyrins and their derivatives, several photosensitizers have been developed largely for use in oncological applications, but have also been examined in other disease areas in the PDT field in humans. (WO 92/06097; WO 97/20846; EP 0 811626; U.S. Pat. Nos. 5,633,275, 5,654,423, 5,675,001, 5,703,230, and 5,705,622). Such photosensitizers include Photofrin (U.S. Pat. No. 4,882,234), 5-aminolevulinic acid (protoporphyrin IX precursor), SnET2, Visudyne® (Benzoporphyrin derivative), Antrin®, Optrin® (Lutetium texaphyrin) and mono-aspartyl chlorin e6 (MACE). All of these compounds were designed specifically for the treatment of solid tumors. Specifically, many of these compounds were designed to have large absorptions in the 620-740 nm range so as to optimize the photoactivation of the drug with a wavelength that will penetrate to the greatest depths possible all tissue types. In particular, these drugs were designed to absorb outside of the blood absorption profile, thus ensuring efficient photoactivation in most tissue types.
  • The excitation light source (usually diode lasers or dye lasers) has historically been matched to the far-red absorption bandwidth of the photosensitizer in order to maximize light penetration through tissues. Indeed, the present inventor believes that all the tetrapyrrolic photosensitizers used have been designed for long wavelength absorption of light (>630 nm) to address this perceived issue. Surprisingly, it has been found that short wavelength photosensitizers (with activation absorptions <600 nm) are capable of delivering effective localized therapy to many disease indications where historically long wavelength photosensitizers (with activation absorptions >600 nm) have shown ineffective clinical outcomes. One such example is in coronary artery disease.
  • While several of the photosensitizers described above have been used to treat atheromatous plaques and some are able to display some inhibition of intimal hyperplasia in animal models, many if not all have characteristics that will limit the usefulness of these drugs in a clinical setting. One particular concern is the half-life of the photosensitizer. A photosensitizer delivered systemically with a long half-life (CASPc, Photofrin, SnET2) may have phototoxic side effects if exposed to direct light, within days of the procedure.
  • A second even more pressing concern that has to date escaped many of the investigators testing new photosensitizers in cardiovascular disease is photochemically induced damage to “normal” myocardial tissue surrounding the artery due to non-selective photosensitizer uptake and long depths of light penetration, which activates the photosensitizer in the myocardial tissue. Historically, it has been believed that attenuation of the photosensitizer excitation light by blood would inhibit the use of wavelengths of light shorter than 600 nm in the cardiovascular field. This may have been true several years ago when balloon catheter technology in PDT was not as advanced as it is today. New endovascular light ballon catheters, however, can remove most of the blood from the treatment area. This advance enables the use of short wavelengths of light that historically may have been attenuated by blood.
  • The use of wavelengths of light lower than 600 nm offers significant advantages in PDT because such wavelengths have penetration characteristics that deliver the PDT effect to the target sites (media and adventicia layers of the vessel) and not to myocardial tissue. Thus, effective therapy can be afforded at the target site, while deeper tissues are shielded from a PDT response by blood absorption within these tissues. Previously reported cardiovascular experiments performed to date on tetrapyrrolic molecules have been done at wavelengths >620 nm. Experiments that we have performed in pig arteries with new photosensitizer candidates at light activation >600 nm have resulted in unacceptable levels of damage to myocardial or cardiac muscle tissue surrounding the treatment area. This has major clinical implications to patients with existing ischemic myocardial or muscle tissue due to poor artery perfusion. Attempts to lower the light dosimetry in order to limit treatments to the target tissue (media/intima) leads to long treatment times and less efficacy. In addition, long treatment times in the artery exposes the patient to additional risks with inflation and deflation of the balloon devices. Importantly, we have demonstrated in pig arteries that effective treatment depths can be obtained with shorter wavelengths of light, with sparing of underlying tissue damage.
  • Thus, it is believed that, long wavelength absorbing molecules (>600 nm), unless highly selective to target myocardial and intimal tissues (which has not to date been reported with any photosensitizer in cardiovascular tissues), may cause unacceptable normal cardiac tissue damage. Therefore, it would appear that activation of lutetium texaphyrin, BPD-MA, MACE, CASPc, SnET2, and pheophorbide PH-II26 with red light may be of limited use in the treatment of cardiovascular disease as all of these compounds are “red” absorbers by design, in so much as all possess low energy absorbtion peaks at wavelengths >600 nm. It should be noted also that chlorins, phthalocyanines and texaphyrin type photosensitizers in general have little absorption in the 500-600 nm regions, and thus may be suboptimal with regard to light activation at green and yellow wavelengths in cardiovascular tissues. In addition, protoporphyrin IX and photofrin do not display absorption maximas at 532 nm, thus they may be inefficient at absorbing treatment light at this wavelength and have very low molar extinction coefficients at 575 nm (˜7000 cm−1/M−1). Furthermore, because long wavelength photosensitizers by design have red absorption peaks, operating room lighting in an emergency situation may cause serious photosensitivity in light exposed tissues. Attempts to use red light filters on operating room lights results in poor tissue contrast and sub-optimal lighting conditions, making surgical procedures under these conditions extremely difficult, if not impossible. Optical clarity is much better at shorter wavelengths (500-600 nm) where the depth of light peneration is limited to a few mm of tissue penetration during the surgical procedure.
  • Another significant drawback of the above long wavelength absorbing compounds mentioned is that they are only suitable for therapy; prior or simultaneous MRI-diagnostic monitoring of the success of the therapy is not possible with them, nor is radiodiagnostic imaging. For this purpose, it is necessary to administer another paramagnetic substance, which must have a biodistribution that is as close to that of the therapeutic agent as possible. This requirement often cannot be met.
  • There have been attempts by groups in the field to provide porphyrin linked MRI or radiodiagnostic compounds. Notable examples include: Hilgar, C. S., et al, U.S. Pat. No. 5,849,259; Niedballa, U., et. al., U.S. Pat. No. 5,275,801; Platzek, J., et. al., U.S. Pat. No. 6,136,841; Niedballa, U., et. al., EP 0355041 A2, A3, and B1; Sakata, I., et. al., U.S. Pat. No. 4,996,312; Sakata, I., et. al., U.S. Pat. No. 4,996,312; and Sakata, I., et. al., U.S. Pat. No. EP 0220686. It has been known for some time that porphyrin derivatives selectively accumulate in human and animal tumors (D. Kessel and T.-II. Chu, Cancer Res. 43, pp.1994-1999, 1983; P. Hambright, Bioinorg. Chem. 5, pp. 87-92, 1975; R. Lipson et al., Cancer 20, pp. 2250-2257, 1967; and D. Sanderson et al., Cancer 30, pp. 1368-1372, 1972). First attempts to use this class of compound as a diagnostic agent were also described in the literature (J. Winkelmann et al., Cancer Research 27, pp. 2060-2064, 1967; N. J. Patronas et al, Cancer Treatment Reports 70, pp. 391-395, 1986). However, the compounds so far described are far from being able to satisfactorily meet the desired requirements to be effective PDT, MRI and radiodiagnostic imaging agents.
  • Substituted hematoporphyrin complex compounds used in diagnosis and treatment are described in patent application EP 0 355 041. While these compounds show a good concentration behavior in various target organs, the described compounds used as NMR diagnostic agents are not satisfacatory because they require a dose necessary for optimal imaging that is too close to the lethal dose. Hematoporphyrin derivatives have the drawback that they can eliminate both pseudobenzylic OH groups in the hydroxyethyl side chains.
  • Derivatives of the deuteroporphyrin have been proposed (Sakata, et. al., U.S. Pat. No. 4,996,312 and EP 0220686) for tumor imaging with radioisotopes, containing as additional complexing groups polyaminopolycarboxylic acids bound to the porphyrin skeleton by ethylene glycol bridges (Photochemistry and Photobiology Vol. 46, pp. 783-788 (1987)). However, such porphyrin esters are not very suitable for parenteral use in patients, especially for NMR or radiodiagnostic diagnosis, since the injection solutions obtained from them can neither be heat-sterilized nor stored for a sufficiently long time.
  • Other derivatives of deuteroporphyrins have been proposed in Hilgar, C. S., et al. U.S. Pat. No. 5,849,259; Niedballa, U., et. al., U.S. Pat. No. 5,275,801; Platzek, J., et. al. U.S. Pat. No. 6,136,841; and Niedballa, U., et. al., EP 0355041 A2, A3, B1 with striking similarity to overcome certain deficiencies of Sakata's deuteroporphyrins by providing metalloporphyrin amide linkages. However, all of these approaches using deuteroporphyrins are suboptimal with respect to design of short wavelength PDT photosensitizers for use as MRI or radiodiagnostic agents for reasons detailed below.
  • Sakata's porphyrin-based PDT/MRI/radiodiagnostic compounds are based on a naturally occurring asymmetrical porphyrin ring system shown in FIG. 1.
    Figure US20050226810A1-20051013-P00001

    In his synthetic philosophy, Sakata has linked polyfunctional carboxyl groups that are capable of binding radioactive metals or MRI active metals to a) the R1 and R2 positions as shown via ether—alcohol linkages; and b) to positions R4 or R5 via ether linking units. This synthetic approach carries with it significant manufacturing problems. First, the linking of one metal chelating moiety to an asymmetrical porphyrin at R1 or R2, R3 or R4 (where R1 and R2 can be vinyl, ethyl, —CH(O-lower alkanoyl)CH3, —CH(OR)CH3 or —CH(O-loweralklene-OR)CH_,) generates at least two new chemical porphyrinic entities in the synthesis process if R1 and R2 (or R3 and R4) are the same linking moiety. This is outlined in scheme 1.
    Figure US20050226810A1-20051013-C00001
    Figure US20050226810A1-20051013-C00002
  • Naturally occurring porphyrins like hematoporphyrin or protoporphyrin cannot be chemically modified such that only one position, either R1 or R2, (or R3 or R4) is selectively modified to form a molecule with a single linking unit as the only product. In this instance, two compounds are always formed which must be separated to obtain a pure single molecule with which to link the metal chelating moiety. The separation of the two porphyrins is often difficult (if not impossible) and complicates both the manufacturing process and the cost of the final product. If one chooses not to separate the isomers, the isomeric components will each have their own toxicities, and pharmacokinetic and distribution profiles. If one of the isomers is not optimal therapeutically due to any one of these parameters, then the route to regulatory approval is often more complex, time consuming and costly than pursuing a single defined isomer. A second limiting factor that has been highlighted previously, is the instability of the various linking groups to aqueous hydrolysis, elimination at sterilization temperatures, or prolonged storage in solution. Additionally, the use of diastereotopic mixtures as occurs with —CH(OR)CH3 groups in porphyrins complicates the analysis of the molecules for development.
  • Niedballa and Platzek's approach also has the same synthetic manufacturing problems as explained for Sakata (except when R1═H), i.e., multiple compounds are produced when a single linking moiety is attached to the molecule. These molecules may offer enhanced stability over Sakata's due to the use of amine linkages. The limitation of R1═H symmetry does not, however, allow for modification of this molecule with other functionality that may enhance localization or uptake in tissues or target organelle, or changes in pharmacokinetic or elimination profiles for singly linked molecules. Compounds with high water solubility are often not taken up efficiently by tumors or cells. The ability to enhance the lipophilicity of the molecule is thus very important.
  • An additional problem, that has been overlooked by all of the prior workers (Sakata, Niedballa, and Platzek) in the development of short wavelength porphyrin photosensitizers, is the limited absortion profile of the porphyrin ring system metallated tetrapyrroles. In general, metallotetrapyrroles have green and yellow absorptions at about 532 and 575 nm with molar extinction coefficients of between 15,000-20,000 M−1cm−1. In the field of photodynamic therapy, the depth of light penetration into tissues is a function of the wavelength of the exciting light. The theoretical efficacy of a photosensitizer largely correlates to the molar extinction coefficient of the photosensitizer's absorption peak and its ability to absorb light. This is due primarily to the fact that the ability of a photosensitizer to absorb incidental light is a function of the cross sectional area of the molecule's absorption profile. Hence, photosensitizers with low molar extinction coefficients capture photons less efficiently than molecules with high molar extinction coefficients and are thus less efficient.
  • Therefore, there remains a need for novel photosensitizers that are easily manufactured, have excellent stability and solubility, and have more favorable wavelength absorption characteristics. There is a further need for photosensitizers that are capable of being modified to contain a wide range of substituents making biological targeting more possible and ultimately enabling control of the properties and uses of the compounds clinically for not only MRI and radiodiagnostic imaging, but also for treatment using photodynamic therapy.
  • The present inventor has found novel metal-free or metallated functionalized phototherapeutic agents that may be used for imaging (MRI or radiodiagnostic) before or after photodynamic therapy. These novel phototherapeutic agents are based on tetrapyrrolic ring systems such as the porphyrins and azaporphyrins that can be covalently linked by stable linkages to metal complexing agents. These new photosensitizers are useful in short wavelength applications in photodynamic therapy.
  • SUMMARY OF THE INVENTION
  • To achieve these and other advantages, and in accordance with the purpose of the invention as embodied and broadly described herein, the present invention in one aspect provides phototherapeutic compositions of metallo-tetrapyrrolic compounds of formula I which may be used as MRI, radiodiagnostic and PDT agents:
    Figure US20050226810A1-20051013-C00003

    In formula I, R1-R12 can be the same or different and can be selected from:
      • H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ethers, polyethers, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, CH(CH3)O-aryl;
      • (CH2)nO-alkoxy, or (CH2)nO-alkyl, where n is an integer from 0 to 8;
      • C(X)2C(X)3, where X is a halogen;
      • CO2R13, where R13 is selected from H, a physiologically acceptable counter ion, a C1-C20 straight or branched chain alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR14, where R14 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R15, (CHX)nCO2R15, or (CX2)nCO2R15, where X is a halogen and R15 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R16), CONHNH(R16), CO(R16), CON(R16)2, CON(R16)(R17), (CH2)nCONH(R16), (CH2)nCON(R16)2, (CH2)nCOR16, (CH2)nCON(R16)(R17), (CX2)nCONH(R16), (CX2)nCON(R16)2, (CX2)nCON(R16)(R17), (CX2)nCOR16, (CH2)nCONHNH(R16), (CX2)nCONHNH(R16), (CHX)nCONH(R16), (CHX)nCONHNH(R16), (CHX)nCO(R16), (CHX)nCON(R16)2, or (CHX)nCON(R16)(R17), where X is a halogen and R16 and R17 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R18), (CH2)nS(R18), (CH2)nNH(R18), (CH2)nNHNH(R18), (CH2)nN(R18)2, (CH2)nN(R18)(R19), or (CH2)nN(R18)(R19)(R20)+A, where R18, R19 and R20 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids (provided —NH(R18) is part of the amino acid), a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R18, R19 and R20 possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR21, or (CH2)nPO(OR21)2, (CH2)nPO2R21, (CH2)nPOR21 where R21 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR22, or (CH2)nNHNHCOR22, where R22 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R23, SO2NHR23, SO2N(R23)2, SO2NHNHR23, or SO2R23, where R23 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and NHR23 can also be an amino acid, an amino acid salt, an amino acid ester residue, an amino acid amide residue, and n is an integer between 0 and 4;
      • aryl or substituted aryl, which may optionally bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons; and
      • R1-R2, R4-R5, R7-R8, R10-R11, R2-R3, R5-R6, R8-R9, and R11-R12 may also possess the atoms necessary to form ring systems, which themselves may possess heteroatoms that may bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons;
      • with the proviso that at least one of the R1-R16 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link:
        Figure US20050226810A1-20051013-C00004
      • wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; and that when R1 and R4 are methyl, R2 and R5 cannot be methyl, a straight chain C1-C6 alkyl, a C7-C12 aralkyl, CH2O(C1-C3alkyl), CH2OH, CH(OH)CH3, CH2CH2OH, CH(NH(CH2)nNH2)CH3, CH2CH2NH(CH2)nNH2 (where n=2, 3, 4, 6), vinyl, ethyl, CH(O-lower alkanoyl)CH3, CH(O-lower alkylene-OR)CH3, or CH(OR)CH3 (where R═H, lower alkyl, a polyfunctional carbonyl compound excluding a hydrogen atom therefrom or a metal derivative of a polyfunctional carbonyl compound).
  • In formula I, M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+ either with or without a physiologically acceptable charge balancing counter ion.
  • In a preferred embodiment of the invention, provided are phototherapeutic compositions of metallo-tetrapyrrolic compounds of formula IA:
    Figure US20050226810A1-20051013-C00005

    In formula IA, R1-R7 can be the same or different and can be selected from:
      • H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ethers, polyethers, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, CH(CH3)O-aryl;
      • (CH2)nO-alkoxy, or (CH2)nO-alkyl; (where n is an integer from 0 to 8);
      • C(X)2C(X)3, where X is a halogen;
      • CO2R8, where R8 is selected from a physiologically acceptable counter ion, a straight or branched chain alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, ether or polyether, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR9, where R9 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R10, (CHX)nCO2R10, or (CX2)nCO2R10, where X is a halogen and R10 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R11), CO(R11), CON(R11)2, CON(R11)(R12), (CH2)nCONH(R11), (CH2)nCON(R11)2, (CH2)nCOR11, (CH2)nCON(R11)(R12), (CX2)nCONH(R11), (CX2)nCON(R11)2, (CX2)nCON(R11)(R12), (CX2)nCOR11, (CH2)nCONHNH(R11), (CX2)nCONHNH(R11), (CHX)nCONH(R11), (CHX)nCONHNH(R11), (CHX)nCON(R11)2, (CHX)nCON(R11)(R12), where X is a halogen and R11 and R12 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R13), (CH2)nS(R13), (CH2)nNH(R13), (CH2)nNH(R13), (CH2)nR13(CH2)nN(R13)(R14), or (CH2)nN(R13)(R14)(R15)+A, where R13, R14 and R15 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, an amino acid amide, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R13, R14 and R15 possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR16, or (CH2)nPO(OR16)2, (CH2)nPO2R16, (CH2)nPOR16 where R16 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR17, (CH2)nNHNHCOR17, where R17 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R18, SO2NHR18, SO2N(R18)2, SO2NHNHR18 or SO2R18, where R18 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, an amino acid residue, an amino acid salt, an amino acid ester residue, an amino acid amide residue, or a functional group of less than about 100,000 daltons;
      • aryl or substituted aryl, which may optionally bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons; and
      • R1-R2, and R3-R4 may also possess the atoms necessary to form ring systems, either aromatic or not, which themselves may possess heteroatoms that may be charged or neutral or bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons;
      • with the proviso that R1 and R4 are the same, R2 and R3 are the same, and that when R7 is H, R1-R4 cannot be methyl; and that at least one of the R1-R7 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, a ester, a ether or an amide link:
        Figure US20050226810A1-20051013-C00006

        wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides.
  • In formula IA, M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably scheduled from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+ either with or without a physiologically acceptable charge balancing counter ion.
  • In another preferred embodiment of the invention, provided are phototherapeutic compositions of metallo-tetrapyrrolic compounds of the formula IB:
    Figure US20050226810A1-20051013-C00007

    In formula IB, R1 and R2 can be the same or different and can be selected from H, NO2, CN, CHO, CO-alkyl, SO3H, SO3alkyl, SO3alkylether, SO3heteroalkyl, SO3Na, SO3K, SO2NHalkyl, SO2N(alkyl)2, SO2NHheteroalkyl, SO2N(heteroalkyl)2, SO2NHhaloalkyl, SO2N(haloalkyl)2, SO2NHhaloalkylether, SO2N(haloalkylether)2, SO2NHalkylether, SO2N(haloalkylether)2, CO-haloalkyl, haloalkyl, heteroalkyl, hydroxyhaloalkyl, haloalkyl ether, haloalkyl ester, a halogen, a alkylcarbonyloxy group;
    R3 and R4 can be the same or different and are selected from:
      • CO2R5, where R5 is selected from a physiologically acceptable counter ion, a straight or branched chain alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, ethers or polyethers, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR6, where R6 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R7, (CHX)nCO2R7, or (CX2)nCO2R7, where X is a halogen and R7 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R8), CO(R8), CON(R8)2, CON(R8)(R9), (CH2)nCONH(R8), (CH2)nCON(R8)2, (CH2)nCOR8, (CH2)nCON(R8)(R9), (CX2)nCONH(R8), (CX2)nCON(R8)2, (CX2)nCON(R8)(R9), (CX2)nCOR8, (CH2)nCONHNH(R8), (CX2)nCONHNH(R8), (CHX)nCONH(R8), (CHX)nCONHNH(R8), (CHX)nCON(R8)2, (CHX)nCON(R8)(R9), where X is a halogen and R8 and R9 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R10), (CH2)nS(R10), (CH2)nNH(R10), (CH2)nNH(R10), (CH2)nR10), (CH2)nN(R10)(R11), or (CH2)nN(R10)(R11)(R12)+A, where R10, R11 and R12 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, an amino acid amide, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R10, R11 and R12 possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR13, (CH2)nPO(OR13)2, (CH2)nPO2R13, (CH2)nPOR13 where R13 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR14, (CH2)nNHNHCOR14, where R14 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R15, SO2NHR15, SO2N(R15)2, SO2NHNHR15 or SO2R15, where R15 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, an amino acid residue, an amino acid salt, an amino acid ester residue, an amino acid amide residue, or a functional group of less than about 100,000 daltons;
      • aryl or substituted aryl, which may optionally bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons;
      • with the proviso that at least one of the R1-R4 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link:
        Figure US20050226810A1-20051013-C00008
      • wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides.
  • In formula IB, M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga3+; Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+ either with or without a physiologically acceptable charge balancing counter ion.
  • In another aspect of the invention, provided are phototherapeutic, MRI and radiodiagnostic compositions of metallo-tetrapyrrolic compounds of formula II that may be used as photosensitizers in photodynamic therapy:
    Figure US20050226810A1-20051013-C00009

    In formula II, R1-R11 can be the same or different and can be selected from:
      • H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ethers, polyethers, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, CH(CH3)O-aryl;
      • (CH2)nO-alkoxy, or (CH2)nO-alkyl, where n is an integer from 0 to 8;
      • C(X)2C(X)3, where X is a halogen;
      • CO2R12, where R12 is selected from a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR13, where R13 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R14, (CHX)nCO2R14, or (CX2)nCO2R14, where X is a halogen and R14 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R15), CONHNH(R15), CO(R15), CON(R15)2, CON(R15)(R16), (CH2)nCONH(R15), (CH2)nCONHNH(R15), (CH2)nCON(R15)2, (CH2)nCOR15, (CH2)nCON(R15)(R16), (CX2)nCONH(R15), (CX2)nCON(R15)2, (CX2)nCON(R15)(R16), (CX2)nCOR15, (CH2)nCONHNH(R15), (CX2)nCONHNH(R15), (CHX)nCONH(R15), (CHX)nCONHNH(R15), (CHX)nCON(R15)2, (CHX)nCON(R15)(R16), where X is a halogen and R15 and R16 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R17), (CH2)nS(R17), (CH2)nNH(R17), (CH2)nNH(R17), (CH2)nR17, (CH2)nN(R17)(R18), or (CH2)nN(R17)(R18)(R19)+A, where R17, R18 and R19 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, an amino acid amide, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R17, R18 and R19 possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR20, or (CH2)nPO(OR20)2, (CH2)nPO2R20, (CH2)nPOR20 where R20 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR21, or (CH2)nNHNHCOR21, where R21 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R22, SO2NHR22, SO2N(R22)2, SO2NHNHR22 or SO2R22, where R22 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, an amino acid residue, an amino acid salt, an amino acid ester residue, an amino acid amide residue, or a functional group of less than about 100,000 daltons;
      • aryl or substituted aryl, which may optionally bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons;
      • R1-R2, R3-R4, R6-R7, R9-R10, R4-R5, R5-R6, R7-R8, R8-R9, R10-R11, or R11-R1, may also possess the atoms necessary to form ring systems, either aromatic or not, which themselves may possess heteroatoms that may be charged or neutral or bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons; with the proviso that at least one of the R1-R11 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link:
        Figure US20050226810A1-20051013-C00010
      • wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides.
  • In formula II, M is 2H or a diamagnetic or paramagnetic metal ion that can be radioactive, photoactive metals being preferably selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+, either with or without a physiologically acceptable charge balancing counter ion.
  • In a preferred embodiment of the invention, provided are phototherapeutic compositions of metallo-tetrapyrrolic compounds of following formula IIA:
    Figure US20050226810A1-20051013-C00011

    In formula IIA, R1-R6 can be the same or different and can be selected from:
      • H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ethers, polyethers, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, or CH(CH3)O-aryl;
      • (CH2)nO-alkoxy, or (CH2)nO-alkyl, where n is an integer from 0 to 8;
      • C(X)2C(X)3, where X is a halogen;
      • CO2R7, where R7 is selected from a physiologically acceptable counter ion, a straight or branched chain alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR8, where R8 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R9, (CHX)nCO2R9, or (CX2)nCO2R9, where X is a halogen and R9 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R10), CONHNH(R10), CO(R10), CON(R10)2, CON(R10)(R11), (CH2)nCONH(R10), (CH2)nCONHNH(R10), (CH2)nCON(R10)2, (CH2)nCOR10, (CH2)nCON(R10)(R11), (CX2)nCONH(R10), (CX2)nCON(R10)2, (CX2)nCON(R10)(R11), (CX2)nCOR10, (CH2)nCONHNH(R10), (CX2)nCONHNH(R10), (CHX)nCONH(R10), (CHX)nCONHNH(R10), (CHX)nCON(R10)2, or (CHX)nCON(R10)(R11), where X is a halogen and R10 and R11 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R12), (CH2)nS(R12), (CH2)nNH(R12), (CH2)nNHNH(R12), (CH2)nN(R12)2, (CH2)nN(R12)(R13); or (CH2)nN(R12)(R13)(R14)+A, where R12, R13 and R14 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, or an amino acid amide provided —NH(R13) is part of the amino acid, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R12, R13 and R14 possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR15, or (CH2)nPO(OR15)2, (CH2)nPO2R15, (CH2)nPOR15where R15 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, amino acids and salts, esters, or amides thereof, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR16, or (CH2)nNHNHCOR16, where R16 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids and salts, esters, or amides thereof, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R17, SO2NHR17, SO2N(R17)2, SO2NHNHR17 or SO2R17, where R17 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, and NHR17 can be an amino acid residue, an amino acid salt, an amino acid ester residue, an amino acid amide residue;
      • aryl or substituted aryl, which may optionally bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons; and
      • R1-R2, and R3-R4 may also possess the atoms necessary to form ring systems, either aromatic or not, which themselves may possess heteroatoms that may be charged or neutral or bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons;
      • with the proviso that at least one of the R1-R4 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link:
        Figure US20050226810A1-20051013-C00012
      • wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides.
  • In formula IIA, M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga3+, Pt2+, Pd2+, S4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+, either with or without a physiologically acceptable charge balancing counter ion.
  • Additionally and in accordance with the present invention, provided are phototherapeutic compositions of metallo-tetrapyrrolic compounds of formula III that may be used as MRI, radiodiagnostic, or PDT agents:
    Figure US20050226810A1-20051013-C00013

    In formula III, R1-R10 can be the same or different and can be selected from:
      • H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ethers, polyethers, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group having a molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, or CH(CH3)O-aryl;
      • (CH2)nO-alkoxy, or (CH2)nO-alkyl, where n is an integer from 0 to 8;
      • C(X)2C(X)3, where X is a halogen;
      • CO2R11, where R11 is selected from a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR12, where R12 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R13, (CHX)nCO2R13, or (CX2)nCO2R13, where X is a halogen and R13 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R14), CONHNH(R14), CO(R14), CON(R14)2, CON(R14)(R15), (CH2)nCONH(R14), (CH2)nCONHNH(R14), (CH2)nCON(R14)2, (CH2)nCOR14, (CH2)nCON(R14)(R15), (CX2)nCONH(R14), (CX2)nCON(R14)2, (CX2)nCON(R14)(R15), (CX2)nCOR14, (CH2)nCONHNH(R14), (CX2)nCONHNH(R14), (CHX)nCONH(R14), (CHX)nCONHNH(R14), (CHX)nCON(R14)2, (CHX)nCON(R14)(R15), where X is a halogen and R14 and R15 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R16), (CH2)nS(R16), (CH2)nNH(R16), (CH2)nNHNH(R16), (CH2)nN(R16)2 (CH2)nN(R16)(R17), or (CH2)nN(R16)(R17)(R18)+A, where R16, R17 and R18 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, or an amino acid amide provided —NHR16 is part of the amino acid, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R16, R17 and R18 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR19, or (CH2)nPO(OR19)2, (CH2)nPO2R19, (CH2)nPOR19 where R19 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR20, (CH2)nNHNHCOR20, where R20 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R21, SO2NHR21, SO2N(R21)2, SO2NHNHR21 or SO2R21, where R21 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons; and NHR21 can be an amino acid residue, an amino acid salt, an amino acid ester residue, or an amino acid amide residue;
      • aryl or substituted aryl, which may optionally bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons; and
      • R1-R2, R3-R4, R6-R7, R8-R9, R4-R5, R5-R6, R9-R10 and R10-R1 may also possess the atoms necessary to form ring systems, either aromatic or not, which themselves may possess heteroatoms that may be charged or neutral or bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons;
      • with the proviso that at least one of the R1-R4 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link:
        Figure US20050226810A1-20051013-C00014
      • wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides.
  • In formula III, M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+ either with or without a physiologically acceptable charge balancing counter ion.
  • In a preferred embodiment of the invention, provided are phototherapeutic compositions of metal-tetrapyrolic compounds of formula IIIA:
    Figure US20050226810A1-20051013-C00015

    In formula IIIA, R1-R4 can be the same or different and are selected from:
      • a functional group of less than about 100,000 daltons;
      • CO2R5, where R5 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR6, where R6 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R7, (CHX)nCO2R7, or (CX2)nCO2R7, where X is a halogen and R7 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R8), (CONHNH(R8), CO(R8), CON(R8)2, CON(R8)(R9), (CH2)nCONH(R8), (CH2)nCONHNH(R8), (CH2)nCON(R8)2, (CH2)nCOR8, (CH2)nCON(R8)(R9), (CX2)nCONH(R8), (CX2)nCON(R8)2, (CX2)nCON(R8)(R9), (CX2)nCOR8, (CH2)nCONHNH(R8), (CX2)nCONHNH(R8), (CHX)nCONH(R8), (CHX)nCONHNH(R8), (CHX)nCON(R8)2, or (CHX)nCON(R8)(R9), where X is a halogen and R8 and R9 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R10), (CH2)nS(R10), (CH2)nNH(R10), (CH2)nNHNH(R10), (CH2)nN(R10)2, (CH2)nN(R10)(R11), or (CH2)nN(R10)(R11)(R12)+A, where R10, R11 and R12 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, or an amino acid amide provided —NHR10 is part of the amino acid, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R10, R11 and R12 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR13, or (CH2)nPO(OR13)2, (CH2)nPO2R13, (CH2)nPOR13 where R13 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR14, or (CH2)nNHNHCOR14, where R4 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R15, SO2NHR15, SO2N(R15)2, SO2NHNHR15 or SO2R15, where R15 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, halbheteroalkyl, aryl, heteroaryl, heterocycle; NHR15 can also be an amino acid residue, an amino acid salt, an amino acid ester residue, or an amino acid amide residue; a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons;
      • aryl or substituted aryl, which may optionally bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons;
      • with the proviso that at least one of the R1-R4 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link:
        Figure US20050226810A1-20051013-C00016
      • wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides.
  • In formula IIIA, M is 2H or a diamagnetic or paramagnetic metal ion that may be radioactive or not, photoactive metals being preferably selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+ either with or without a physiologically acceptable charge balancing counter ion.
  • In another aspect of the invention, provided are phototherapeutic compositions of metallo-tetrapyrrolic compounds of formula IV that may be used as MRI, radiodiagnostic, or PDT agents:
    Figure US20050226810A1-20051013-C00017

    In formula IV, R1-R8 can be the same or different and are selected from:
      • H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ethers, polyethers, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N(CH3)3 +A, CH═N(alkyl)2 +A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, or CH(CH3)O-aryl;
      • (CH2)nO-alkoxy, or (CH2)nO-alkyl; where n is an integer from 0 to 8;
      • C(X)2C(X)3, where X is a halogen;
      • CO2R9, where R9 is selected from a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
      • (CH2)nOH, or (CH2)nOR10, where R10 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nCO2R11, (CHX)nCO2R11, (CX2)nCO2R11, where X is a halogen and R11 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
      • CONH(R12), CONHNH(R12)CO(R12), CON(R12)2, CON(R12)(R13), (CH2)nCONH(R12), (CH2)nCONHNH(R12), (CH2)nCON(R12)2, (CH2)nCOR12, (CH2)nCON(R12)(R13), (CX2)nCONH(R12), (CX2)nCON(R12)2, (CX2)nCON(R12)(R13), (CX2)nCOR12, (CH2)nCONHNH(R12), (CX2)nCONHNH(R12), (CHX)nCONH(R12), (CHX)nCONHNH(R12), (CHX)nCON(R12)2, (CHX)nCON(R12)(R13), where X is a halogen and R12 and R13 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • S(R14), (CH2)nS(R14), (CH2)nNH(R14), (CH2)nNHNH(R14), (CH2)nN(R14)2, (CH2)nN(R14)(R15), or (CH2)nN(R14)(R15)(R16)+A, where R14, R15 and R16 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, or an amino acid amide provided —NH(R14) is part of the amino acid, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R14, R15 and R16 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
      • (CH2)nOPO2OR17, or (CH2)nPO(OR17)2, (CH2)nPO2R17, (CH2)nPOR17 where R17 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • (CH2)nNHCOR18, or (CH2)nNHNHCOR18, where R18 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
      • SO3R19, SO2NHR19, SO2N(R19)2, SO2NHNHR19 or SO2R19, where R19 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and NHR19 can also be an amino acid residue, an amino acid salt, an amino acid ester residue, or an amino acid amide residue; and
      • aryl or substituted aryl, which may optionally bear one or more substituents selected from hydroxy groups, alkyl groups, carboxyl groups and its esters and amides and sulfonic acid groups and their esters and amides, and substitiuents with a molecular weight of less than or equal to about 100,000 daltons;
      • with the proviso that at least one of the R1-R12 groups is linked to a complexing agent of general formula IIA, IIB, IIC, IID, IIE by way of an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link:
        Figure US20050226810A1-20051013-C00018
      • wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; and
      • A, B, C, and D can be the same or different and can be selected from N, CH, CR20 where R20 is selected from a halogen, aryl, subsitituted aryl, heteroaryl, alkyl, haloalkyl, heterohaloalkyl, heterocycle, hydroxyalky, hydroxyhaloalkyl, or a functional group of molecular weight of less than about 100,000 daltons.
  • In formula IV, M is selected from 2H or a diamagnetic or paramagnetic metal ion that can be radioactive, photoactive metals being preferably selected from Ga3+, Pt2+, Pd2+, S4+, In3+, Ge4+, Si4+, Al3+, Zn2+, Mg2+ the appropriate number of physiologically acceptable charge balancing counter ions.
  • In accordance with a preferred embodiment of the invention, the metallo-tetrapyrrolic compounds of the invention can be derived by various procedures from naturally occuring cyclic tetrapyrroles. The naturally occurring cyclic tetrapyrrolic molecules can have the basic ring structure of compounds I, II, III, and IV, whose substituents are outlined in Table 1, and are particularly preferred as starting materials for the synthesis of the compounds of structures I-IV. In particular, tetrapyrroles derived from naturally occuring ring systems that have one linking group are particularly preferred. These are shown in scheme 1.
  • In a second preferred aspect of this invention, the tetrapyrrole is derived by the coupling of suitably substituted dipyrromethane, dipyrromethenes, biladienes, builirubins, pyrroles and functionalized aldehydes, or functionalized maleonitriles. The cyclic tetrapyrroles that have the basic ring structure of compounds I-IV, whose substituents are outlined in Table 2, are particularly preferred as starting materials for the synthesis of compounds of structures I-IV.
  • It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention as claimed.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The terms “tetrapyrrole”, tetrapyrrolic molecule and “porphyrin” are used here to designate compounds of the cyclic structure where four pyrrolic ring systems are linked via either carbon or nitrogen bonds. Compounds within the scope of the invention include porphyrins, mono-, di-, tri- and tetraazaporphyrins, and porphyrin isomers such as porphycenes, isoporphycenes, hemiporphycenes, corroles, corrphycenes, and the like.
  • Included in the first class of metallated tetrapyrrolic compounds of the invention are those of the porphyrins. Scheme 1 outlines an example of the synthesis of porphyrins that are derived from plants. Particularly advantageous are the porphyrins based on chloroporphyrin e6 (9), chloroporphyrin e4 (10), phylloporphyrin (11), rhodoporphyrin (7), pyrroporphyrin (8), pheoporphyrin a5 (13) and phylloerythrin (12) and compounds having similar ring systems. Such compounds can be synthesized with single linking groups, which can be modified according to the invention to increase their biological activity and MRI and radiodiagnostic capacity. In particular, the propionic esters of (2), (3), (5) and (6) can be selectively hydrolized to form carboxylic acids, which can then be linked to the metal coordinating moiety. Alternatively, the carboxylic acids can be converted to amides with a free amine linking unit, which can then be linked to the metal co-ordinating moiety. Porphyrin amide derivatives like (4) (R2═NHR3) may be synthesized from phylloporphyrin such that an amine linking group is present. Examples include where R2═NHCH2CH2NH2, NHCH2CH2CH2NH2, NHCH2CH2NH2, NH(CH2)2O(CH2)2NH2 and similar compounds. These amine groups can then be linked to the metal co-ordinating moiety.
  • While plant derived porphyrins are preferred as starting materials in the invention due to their abundant availability, a very large number of synthetic porphyrins are generally applicable to the invention. Such porphyrins can be made by synthetic methods known to those skilled in the art, via coupling of pyrrolic precursors, dipyrromethanes, dipyrromethenes and biladienes to give the required porphyrins with widely ranging functionality at both the β and meso positions. The synthesis of porphyrins via the coupling of pyrrolic intermediates is outlined in detail in chapters 1, 2, 3 in “The Porphyrin Handbook” Editors, K. M. Kadish, K. M. Smith, R. Guilard, Volume 1, Academic press, 2000, p. 1-148, the disclosure of which is hereby incorporated by reference herein. Such functionality is explained in detail below. This functionality can be modified by further chemical reactions. Such compounds can then be modified according to the invention to produce metalloporphyrins that absorb at or about 400, 532 and 575 nm. Table 1 outlines some of the preferred porphyrins that may be used as starting materials in the development of these types of compounds.
    TABLE 1
    Figure US20050226810A1-20051013-C00019
    Tetrapyrrole R1 R2 R3 R4 R5 R6 R7 R8
    Hematoporphyrin IX Me EO Me EO Me PO PO Me
    Protoporphyrin IX Me V Me V Me PO PO Me
    Mesoporphyrin IX Me Et Me Et Me PO PO Me
    Deuteroporphyrin IX Me H Me H Me PO PO Me
    Hematoporphyrin dialkylethers Me EOE Me EOE Me PO PO Me
    Coproporphyrin I PO Me PO Me PO Me PO Me
    Coproporphyrin II Me PO PO Me Me PO PO Me
    Coproporphyrin III Me PO Me PO Me PO PO Me
    Uroporphyrin IX Me EO Me EO Me PO PO Me
    Pentacarboxyporphyrin I PO Me PO Me PO Me PO AO
    Pentacarboxyporphyrin III PO Me PO Me PO Me AO PO
    2, 4-dihalodeuteroporphyrin IX Me X Me X Me PO PO Me
    Hexacarboxyporphyrin I PO Me PO AO PO Me PO AO
    Hexacarboxyporphyrin III PO Me PO Me PO AO PO AO
    Heptacarboxyporphyrin I PO Me PO AO PO AO PO AO

    AO = —CH2CO2H; PO = —CH2CH2CO2H, EO =]—CH(OH)CH3, EOE = —CH(OR)CH3, Me = —CH3, Et = CH2CH3, V = —CH═CH2
  • Figure US20050226810A1-20051013-C00020
    Figure US20050226810A1-20051013-C00021
    Figure US20050226810A1-20051013-C00022
    Figure US20050226810A1-20051013-C00023
  • A second preferred class of compounds according to the invention are the mono-, di-, tri- and tetra-azaporphyrins. Schemes 3-7 outline the synthesis of mono-, di-, and tetra-azaporphyrins, examples of which are listed in Table 2.
    TABLE 2
    Figure US20050226810A1-20051013-C00024
    Tetrapyrrole A B C D R1 R2 R3 R4 R5 R6 R7 R8
    5-aza-coproporphyrin II N CH CH CH Me PO PO Me Me PO PO Me
    5-aza-protoporphyrin IX N CH CH CH Me V V Me Me PO PO Me
    5-aza-mesoporphyrin IX N CH CH CH Me Et Me Et Me PO PO Me
    5-aza-mesoporphyrin XIII N CH CH CH Me Et Et Me Me PO PO Me
    5-aza-uroporphyrin III N CH CH CH PO AO PO AO PO AO AO PO
    5-aza-isomesoporphyrin N CH CH CH Et Me Me Et Me PO PO Me
    5-aza-mesoporphyrin III N CH CH CH Me Et Me Et PO Me Me PO
    5,15-Diaza-coproporphyrin II N CH N CH Me PO PO Me Me PO PO Me
    5,15-diaza-mesoporphyrin III N CH N CH Me Et Me Et PO Me Me PO

    AO = —CH2CO2H; PO = —CH2CH2CO2H, EO = —CH(OH)CH3, EOE = —CH(OR)CH3, Me = —CH3, Et = CH2CH3, V = —CH═CH2
  • Schemes 3-7 outline synthetic routes to novel tetrapyrrolic molecules that possess a linking group with terminal amine groups. Such compounds can be linked to a metal complexing reagent (MCR) and subsequently modified to be phototherapeutic and diagnostic compounds.
    Figure US20050226810A1-20051013-C00025
    Figure US20050226810A1-20051013-C00026
  • Mono-azaporphyrins are synthesized efficiently via the coupling of dibromobiladienes with sodium azide or via the reaction of oxyporphyrins with ammonia. Copper and metal free diazaporphyrins can be obtained via the coupling of 5,5′-dibromopyrromethenes with sodium azide. Tetra-azaporphyrins are synthesized most efficiently via the treatment of substituted maleonitriles with magnesium powder or magnesium alcoxides. Such reactions are well known in the art and are outlined in detail by N. Kobayashi in “The Porphyrin Handbook,” K. M. Kadish, K. M. Smith, R. Guilard, Editors, Volume 2, Chapter 13, Academic Press, 2000, p. 301-360, the disclosure of which is hereby incorporated by reference herein.
  • The peripheral functionality of these compounds is important with respect to further derivatization to achieve the desired coupling to the metal complexing reagent (MCR) and the desired biological effect (both therapeutic and diagnostic). The types of peripheral functionality applicable to the invention are described in detail below. It is recognized that small changes in the peripheral functionality can have pronounced effects on the biological efficacy of the molecules as does metal co-ordination to the compounds. Schemes 4-7 outline synthetic routes to the novel tetrapyrrolic molecules of the invention.
  • The new compounds of the invention are based on the porphyrin, mono-, di-, tri- and tetra-azaporphyrin ring systems that bear peripheral functionality on the ring system. Such functionality includes esters, alcohols, amides, amines, ethers, and phosphates. Such derivatives may also have at least one hydroxylated residue present, or an amine group with which to couple the metal co-ordination compound. The new porphyrins themselves may be photodynamically active as metal free analogs and therefore useful as PDT agents. In addition, metallated derivatives of these compounds are also of particular interest for treatment and diagnosis of disorders of the cardiovascular system, normal or abnormal conditions of the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system including bone, connective tissue, cartilage and skeletal muscle, pulmonary system, gastrointestinal system including the liver, reproductive system, skin, immune system, cardiovascular system, urinary system, ocular system, auditory or olfactory system, where shorter wavelengths of light are necessary or advantageous to effect a desired therapy.
  • Scheme 4 outlines chemistry that has been undertaken to produce photosensitizing or diagnostic agents (based on non-naturally occuring porphyrin systems and azaporphyrins) that possess pendant terminal amine moieties and is exemplary only and is not intended to limit the invention. It should be noted that the functionality and position of the N and C meso atoms can be varied to produce analogs different from those shown. Additionally, the R groups in these schemes constitute functional groups that can be symmetrically substituted and can also, if desired, be modified by techniques known to those skilled in the art based on the chemistry described herein without departing from the spirit or scope of the invention.
    Figure US20050226810A1-20051013-C00027
  • In scheme 4, the ester functionality of porphyrins or azaporphyrins can be hydrolyzed to yield both mono- and di-acid compounds. It is preferred that the synthesis of mono-acid compounds via this method occurs on compounds that are symmetrical in their substitution pattern of R1-R4, such that isomers are not formed. The mono- or di-acid can then be converted to the desired amide via standard techniques, to produce pendant arm groups with terminal amine moieties. Alternatively, if R5 possesses a functional group that can be modified to produce a reactive linking moiety (for example C6H4SO3H, C6H4CO2H and the like), these may alternatively be chemically modified to produce compounds with pendant arm groups having terminal amine moieties. Such compounds can, if desired, be reacted with: metals to produce metallotetrapyrrolic complexes.
    Figure US20050226810A1-20051013-C00028
    Figure US20050226810A1-20051013-C00029
    Figure US20050226810A1-20051013-C00030
  • Scheme 5 outlines the synthesis of porphyrins and azaporphyrins possessing two pendant arm terminal amine moieties. In this instance, the ester functionality of porphyrins or azaporphyrins can be hydrolyzed to yield di-acid compounds. The mono-or di-acid can then be converted to its di-acid chloride, which subsequently can be reacted with sodium azide in acetone to yield the di-azide compound. The di-azide then can be rearranged in methanol to give the di-urethane derivative, which can then be hydrolysed in acid to give protonated amine compounds. Neutralization with base yields the free amine compound. Such compounds can, if desired, be reacted with metals to produce metallo-tetrapyrrolic diamine complexes.
  • Scheme 6 outlines the synthesis route of metal-free or metallated mono- or di-amine porphyrins based on rhodoporphyrin or pyrroporphyrin. In the case of rhodoporphyrin (R═CO2Me), it is possible to selectively hydrolyze the propionic acid ester with dilute HCl/water to give the mono acid derivative shown. Alternatively, both groups can be hydrolyzed with KOH/H2O to give the di-acid derivative. The acid groups can then be converted to the acid chloride derivatives and reacted with the appropriate amine to give compounds possessing one or two pendant arm terminal amine moieties. These compounds can, if desired, be reacted with metals to produce metallo-tetrapyrrolic mono or di-amine complexes.
  • Scheme 7 outlines the synthesis of metal-free or metallated mono or di-amine di-azaporphyrins. The di-azaporphyrins themselves can be synthesized via the coupling of appropriate brominated dipyrromethanes. Once synthesized, the peripheral functionality can be modified by similar chemistry as outlined in schemes 4 and 5. As before, it is preferred that if mono-amine functionalized compounds are to be made, R1-R4 should possess a symmetrical substitution pattern. This preference does not apply if di-amine substitution is desired.
  • The introduction of the desired metals (e.g., Zn, Ga, Al, Sn, In, Mg, Mn, Fe, etc) into the porphyrins or azaporphyrins can be carried out according to methods that are known in the literature (e.g., The Porphyrins, ed. D. Dolphin, Academic Press, New York 1980, Vol. V, p. 459; DE 4232925). In particular, metal substitution of pyrrolic NH's can be carried out by heating the metal-free ligand with the corresponding metal salt, preferably acetate or halide, optionally with the addition of acid-buffering agents, such as, for example, sodium acetate in a polar solvent. Alternatively, such substitutions can be carried out by metal exchange in which a metal that is already complexed by the porphyrin or azaporphyrin is displaced by the desired metal. An example of such a metal is cadmium. In this process, the preferred solvent is a polar solvent, such as, for example, methanol, glacial acetic acid, dimethylformamide, chloroform or water. In some instances where the metal is difficult to remove under acid conditions (Pt, Pd), it is more practical to generate the metalloporphyrin or metalloazaporphyrin compounds prior to modification to form the amine linking units. Additionally, the introduction of a diamagnetic or paramagnetic metal M into the porphyrin system can be carried out before or after linkage of the metal complexing agent radical (MCR). As a result, an especially flexible procedure for the synthesis of the compounds according to the invention is made possible.
  • The reaction of a metal-free or metallated porphyrin or azaporphyrin amine with a metal complexing agent (MCA) can be carried out according to methods that are known in the literature. Preferable MCA's include diethylenetriaminepentaacetic acid and 1,4,7,10-tetraazacyclododecane-1,4,7-triacetic acid, which can be bonded via a linker to the respective porphyrin or azaporphyrin derivatives. See, e.g., DE 4232925 for IIa and IId; DE 19507822, DE 19580858 and DE 19507819 for IIb; U.S. Pat. No. 5,053,503, WO 96/02669, WO 96/01655, EP 0430863, EP 255471, U.S. Pat. No. 5,277,895, EP 0232751, and U.S. Pat. No. 4,885,363 for IIc and IIe.
  • In accordance with the invention, an activated MCR can be reacted with the amine porphyrin or azaporphyrin derivatives such that a covalent link between the two compounds occurs. As can be seen in schemes 4-7, the nature of the linking amine moiety on the porphyrins or azaporphyrins (P) to the MCR compounds may be varied. Preferable examples include: P-(CH2)nNH2, where n is an integer from 1 to 10; P-(aryl)NH2, P-CONHNH2, or P-(CH2)nCONHNH2, where n is an integer from 1 to 10; P-CONH(CH2)nNH2, where n is an integer from 1 to 10; P-CONH(CH2)2O(CH2)2NH2, or P-CONH[(CH2)2]nOn/2[(CH2)2]nNH2, where n is an integer from 1 to 10; P-SO2NHNH2, or P-SO2NH(CH2)nNH2, where n is an integer from 1 to 10; P-SO2NH(CH2)2O(CH2)2NH2, or P-SO2NH[(CH2)2]nOn/2[(CH2)2]nNH2, where n is an integer from 1 to 10; P-(Aryl)-SO2NHNH2, or P-(Aryl)-SO2NH(CH2)nNH2, where n is an integer from 1 to 10; and P-(Aryl)-SO2NH(CH2)2O(CH2)2NH2, or P-(Aryl)-SO2NH[(CH2)2]nOn/2[(CH2)2]nNH2, where n is an integer from 1 to 10. It would be apparent to those skilled in the art that other suitable linking amine units could be used in accordance with the teachings of the specification.
  • Alternatively, in accordance with the invention, porphyrin and azaporphyrin units can be generated that possess alcohol terminal linking groups. Preferred examples of such groups include P-(CH2)nOH, where n is an integer from 1 to 10; P-(aryl)OH, P-CONHOH, or P-(CH2)nCONHOH, where n is an integer from 1 to 10; P-CONH(CH2)nOH, where n is an integer from 1 to 10; P-CONH(CH2)2O(CH2)2OH, or P-CONH[(CH2)2]nOn/2[(CH2)2]nOH, where n is an integer from 1 to 10; P-SO2NHOH, or P-SO2NH(CH2)nOH, where n is an integer from 1 to 10; P-SO2NH(CH2)2O(CH2)2OH, or P-SO2NH[(CH2)2]nOn/2[(CH2)2]nOH, where n is an integer from 1 to 10; P-(Aryl)-SO2NHOH, or P-(Aryl)-SO2NH(CH2)nOH, where n is an integer from 1 to 10; P-(Aryl)-SO2NH(CH2)2O(CH2)2OH, or P-(Aryl)-SO2NH[(CH2)2]nOn/2[(CH2)2]nOH, where n is an integer from 1 to 10; P-CO2(CH2)nOH, where n is an integer from 1 to 10; P-CO2(CH2)2O(CH2)2OH, or P-CO2[(CH2)2]nOn/2[(CH2)2]nOH, where n is an integer from 1 to 10; P-SO3(CH2)nOH, where n is an integer from 1 to 10; P-SO3(CH2)2O(CH2)2OH, or P-SO3[(CH2)2]nOn/2[(CH2)2]nOH, where n is an integer from 1 to 10; P-(Aryl)-SO3(CH2)nOH, where n is an integer from 1 to 10; and P-(Aryl)-SO3(CH2)2O(CH2)2OH, or P-(Aryl)-SO3[(CH2)2]nOn/2[(CH2)2]nOH, where n is an integer from 1 to 10. Such compounds can then be linked to the MCR group. It would be apparent to those skilled in the art that other suitable linking alcohol units or other reactive moieties can be used in accordance with the teachings of the specification.
  • The MCR linking group Q is an organic group that when linked to the amine or alcohol porphyrin or azaporphyrin results in a product that is an ester, an amide, an amine, an ether, or a thiolate. Preferable reactive MCR's are described in, for example, U.S. Pat. No. 4,885,363, U.S. Pat. No. 5,730,956, U.S. Pat. No. 6,136,841, and U.S. Pat. No. 5,275,801 the disclosures which are hereby incorporated herein by reference. Examples are illustrated in FIG. 2.
    Figure US20050226810A1-20051013-P00002
  • Examples of linker groups Q include —CO—, —CS—, —COCH2NH—, —CO(CH2)2NH, —CO(CH2)2—, —COCH2OC6H4CO, —COC6H4NH—, —COCH2OCH2NH—, —COC6H4—, —COCH2NHCOCH2CH(CH2COOH)C6H4NH—, phenyleneoxy, a C1-C12 alkylene or a C7-C12 aralkylene group by one or more oxygen atoms. Obviously a large variety of linker groups are possible and it would be apparent to those skilled in the art that other suitable groups can be used in accordance with the teachings of the specification without deviating from the spirit of the invention.
  • In accordance with the invention, the porphyrin or azaporphyrin linked MCR compounds can then be modified to produce PDT/MRI radiodiagnostic compounds. If the compounds are to be used for NMR diagnosis, paramagnetic metal ions must be present in the complex. These are preferably divalent or trivalent ions of the elements of atomic numbers 21-29, 42-44 and 57-71. In this instance, the paramagnetic metal may be coordinated to either the inner pyrrolic core of the porphyrin or azaporphyrin, or in the MCR pendant arm, or in both. Suitable ions include, for example, chromium, gadolinium, dysprosium, manganese, iron, cobalt, cobalt, nickel, copper, praseodymium, neodymium, samarium, terbium, holmium, erbium and ytterbium ions. Because of their high magnetic moment, the gadolinium, dysprosium, manganese, terbium, holmium, erbium and iron ions are especially preferred.
  • For the use of the agents according to the invention for photodynamic therapy, the porphyrin or azaporphyrin compound should be metal free, i.e, M=2H, or should have coordinated photoactive metals, preferred examples of which include zinc, indium, gallium, tin, germanium, palladium, platinum, aluminum, silicon, ruthenium, yttrium, ytterbium, magnesium, lutetium, and cadmium.
  • For the use of the agents according to the invention in nuclear medicine, the metal ions must be radioactive. Examples that are suitable for the invention include radioisotopes of the elements copper, cobalt, gallium, zinc, germanium, yttrium, strontium, technetium, indium, ytterbium, gadolinium, samarium, thallium, and iridium. In this instance, the radioactive isotope may be coordinated to the porphyrin or azaporphyrin ligand or within the MCR, or both. Alternatively, such compounds can be modified such that they become active or excited using sonotherapy (e.g., M=gallium) or photothermally.
  • Metal chelation to the MCR group can be carried out by techniques known in the literature (see, e.g., DE3401052) by the metal oxide or metal salt (e.g., nitrate, acetate, carbonate, chloride or sulfate) of the metal that is desired. In each case the metal oxide or salt can be suspended or dissolved in polar solvents such as water or aqueous alcohols and then reacted with the corresponding amount of the complexing ligand. If desired, acidic hydrogen atoms or acid groups that are present can be substituted by cations of inorganic and/or organic bases or amino acids.
  • In accordance with the invention, neutralization can be carried out with the aid of inorganic bases, such as, e.g., alkali or alkaline-earth hydroxides, carbonates or bicarbonates and/or organic bases such as, for example, primary, secondary and tertiary amines, such as, e.g., ethanolamine, morpholine, glucamine, N-methyl- and N,N-dimethylglucamine, as well as basic amino acids, such as, e.g., lysine, arginine and ornithine or amides of originally neutral or acidic amino acids.
  • For the production of neutral complex compounds, a sufficient amount of the desired bases can be added to, for example, the acidic complex salts in aqueous solution or suspension to ensure that the neutral point is reached. The solution that is obtained can then be evaporated to the dry state in a vacuum. It is may be advantageous to precipitate the neutral salts that are formed by adding water-miscible solvents, such as, for example, lower alcohols (e.g., methanol, ethanol, isopropanol, acetonitrile), lower ketones (e.g., acetone), polar ethers (e.g., tetrahydrofuran, dioxane, 1,2-dimethoxyethane) and thus to obtain easily isolated and readily purified crystallizates. It has proven especially advantageous to add the desired base as early as during the complexing of the reaction mixture and thus eliminate a process step.
  • If the acidic complex compounds contain several free acid groups, it is often advantageous to produce neutral mixed salts that contain both inorganic and organic cations as counter-ions. This can be achieved, for example, by reacting the complexing ligands in aqueous suspension or solution with the oxide or salt of the element that yields the central ion and half of the amount of an organic base that is required for neutralization. The complex salt that is formed can then be isolated, optionally purified, and then mixed for complete neutralization with the required amount of inorganic base. The sequence in which the base is added can also be reversed. Another way of obtaining neutral complex compounds consists of converting the remaining acid groups in the complex completely or partially into esters. This can be achieved by subsequent reaction on the finished complex, e.g., by exhaustive reaction of free carboxy groups with dimethylsulfate.
  • Pharmaceutical agents of the invention can be produced by adding to the complex compounds of the invention certain additives that are commonly used in the pharmaceutical industry to suspend or dissolve the compounds in an aqueous medium, and then the suspension or solution can be sterilized by techniques known in the art. Suitable additives include, for example, physiologically harmless buffers (such as, e.g., trimethamine), small additions of complexing agents (such as, e.g., diethylenetriaminepentaacetic acid) or, if necessary, electrolytes such as, e.g., sodium chloride or antioxidants such as, e.g., ascorbic acid, butylate hydroxy toluene, or tocopherol.
  • If suspensions or solutions of the agents according to the invention in water or in physiological salt solution are desired for enteral administration or other purposes, they can be mixed with one or more adjuvants that are commonly used in galenicals (e.g., methylcellulose, lactose, mannitol) and/or surfactant(s) (e.g., lecithins, Tween, and/or flavoring substances for taste correction (e.g., ethereal oils).
  • In principle, it is possible to produce the pharmaceutical agents according to the invention even without isolating the complex salt. In any case, special care must be taken to perform the chelation such that the salts and salt solutions according to the invention are virtually free of noncomplexed metal ions that may have a toxic effect. This can be ensured, for example, by the use of color indicators such as xylenol orange by control titrations during the production process. As a final precaution, there remains purification of the isolated complex salt.
  • To avoid undesirable photoreactions of porphyrins and azaporphyrins, the compounds and agents according to the invention should be stored and handled as much as possible in a light-free environment.
  • The pharmaceutical agents according to the invention preferably contain from about 20 μmol/L to about 200 mmol/L of the complex salt and are generally dosed in amounts of 0.01 μmol to 2 mmol/kg of body weight, both for their use in PDT and for therapy monitoring using MRI diagnosis. They are intended for enteral and parenteral administration or are administered with the methods of interventional radiology.
  • The agents according to the invention are especially suitable for PDT and as MRI contrast media. After administration, they can enhance the informational value of the image that is obtained from a nuclear spin tomograph by increasing the signal intensity. They are effective without burdening the body with large amounts of foreign substances.
  • The high water-solubility of the agents according to the invention allows the production of highly concentrated solutions, so as to keep the volume burden of the circulation within acceptable limits and to compensate for dilution by bodily fluid. In addition, the agents according to the invention show not only a high stability in vitro but also a surprisingly high stability in vivo, so that a release or an exchange of the ions, which are inherently toxic and not covalently bonded in the complexes, will not be harmful within the time that it takes for the contrast media to be completely excreted.
  • Similar reactions can be undertaken on tetrapyrrolic molecules in which more than two carboxylic acid functionalities are present, for example, those compounds shown in Tables 1 and 2. Such reactions on mono-, di- and tetra-azaporphyrin compounds are particularly preferred because metallo-derivatives of such compounds have larger molar extinction co-efficents than the porphyrins in the green and yellow region, and thus theoretically may be more efficient photosensitizers as a larger cross-sectional area of light may be absorbed. While the specification describes several chemical modifications to the tetrapyrrolic compounds, those skilled in the art would know that additional modifications can be made to the tetrapyrrolic ring systems in accordance with the teachings of the specification.
  • The scope of the present invention is not limited to the specific disclosure provided herein. As shown by the above disclosure, any porphyrinic molecule may be modified according to the invention to form the desired photoactive compounds with widely differing functionality as described in the literature (for example see “Porphyrins and Metalloporphyrins” Ed. K. Smith, Elsevier, 1975, N.Y., “The Porphyrins”, Ed. D. Dolphin, Vol I-V, Academic Press, 1978, and “The Porphyrin Handbook”, Eds. K. Kadish, K. M. Smith, R. Guilard, Academic Press, 2000). These compounds contain various and ranging substituents on the β-pyrrole positions or meso-positions of the porphyrin ring, either symmetrically or asymmetrically substituted on the ring.
  • Examples of such functionality include functional groups having a molecular weight less than about 100,000 daltons and can be a biologically active group or organic. Examples include, but are not limited to: (1) hydrogen; (2) halogen, such as fluoro, chloro, iodo and bromo (3) lower alkyl, such as methyl, ethyl, n-propyl, butyl, hexyl, heptyl, octyl, isopropyl, t-butyl, n-pentyl and the like groups; (4) lower alkoxy, such as methoxy, ethoxy, isopropoxy, n-butoxy, t-pentoxy and the like; (5) hydroxy; (6) carboxylic acid or acid salts, such as —CH2COOH, —CH2COONa, —CH2CH2COOH, —CH2CH2COONa, —CH2CH2CH(Br)COOH, —CH2CH2CH(CH3)COOH, —CH2CH(Br)COOH, —CH2CH(CH3)COOH, —CH(Cl)CH2CH(CH3)COOH, —CH2CH2C(CH3)2COOH, —CH2CH2C(CH3)2COOK, —CH2CH2CH2CH2COOH, C(CH3)2COOH, CH(Cl)2COOH and the like; (7) carboxylic acid esters, such as —CH2CH2COOCH3, —CH2CH2COOCH2CH3, —CH2CH(CH3)COOCH2CH3, —CH2CH2COOCH2CH2CH3, —CH2CH2CH2COOCH2CH2CH3, —CH2CH(CH3)COOCH2CH3, —CH2CH2COOCH2CH2OH, —CH2CH2COOCH2CH2N(CH3)2 and the like, particularly halogenated alkyl esters; (8) sulfonic acid or acid salts, for example, group I and group II salts, ammonium salts, and organic cation salts such as alkyl and quaternary ammonium salts; (9) sulfonylamides such as —SO2NH(alkyl), —SO2N(alkyl)2, —SO2NH(alkyl-OH), —SO2N(alkyl-OH)2, —SO2NH(alkyl)-N(alkyl)2, —SO2N(alkyl-N(alkyl)2)2, SO2(NH(alkyl)-N(alkyl)3 +Z) and the like, wherein Zis a counterion, —SO2NHCH2CO2H, substituted and unsubstituted benzene sulfonamides and sulfonylamides of aminoacids and the like; (10) sulfonic acid esters, such as SO3(alkyl), SO3(alkyl-OH), SO3(alkyl-N(alkyl)2), SO3(alkyl-N(alkyl)3 +Z) and the like, wherein Zis a counterion, SO3CH2CO2H, and the like; (11) amino, such as unsubstituted or substituted primary amino, methylamino, ethylamino, n-propylamino, isopropylamino, butylamino, sec-butylamino, dimethylamino, trimethylamino, diethylamino, triethylamino, di-n-propylamino, methylethylamino, dimethyl-sec-butylamino, 2-aminoethoxy, ethylenediamino, cyclohexylamino, benzylamino, phenylethylamino, anilino, N-methylanilino, N,N-dimethylanilino, N-methyl-N-ethylanilino, 3,5-dibromo-4-anilino, p-toluidino, diphenylamino, 4,4′-dinitrodiphenylamino and the like; (12) cyano; (13) nitro; (14) a biologically active group; (15) amides, such as —CH2CH2CONHCH3, —CH2CH2CONHCH2CH3, —CH2CH2CON(CH3)2, —CH2CH2CON(CH2CH3)2, —CH2CONHCH3, —CH2CONHCH2CH3, —CH2CON(CH3)2, —CH2CON(CH2CH3)2, —CH2CH2CONHSO2CH3; (16) iminium salts, for example —CH═N(CH3)2 +Zand the like, wherein Zis a counterion); (17) boron containing complexes; (18) carbon cage complexes (e.g., C20 and the like); (19) polyfunctional carboxylic acid groups and their metal cluster complexes, for example metal complexes of polyfunctional carboxylic acid moieties such as of EDTA, DTPA, EGTA, crown ethers, cyclams, cyclens, and the like; (20) other porphyrin, chlorin, bacteriochlorin, isobacteriochlorin, azaporphyrin, tetraazaporphyrin, phthalocyanine, naphthalocyanine, texaphyrins, tetrapyrrolic macrocycles or dye molecules and the like; (21) alkynyl, including alkyl, aryl, acid and heteroatom substituted alkynes; (22) leaving or protecting groups; (23) aromatic ring systems (aryl), such as substituted phenyls, napthalenes, anthracenes, benzopyrenes, quinolines, benzoquinolines, benzoperylene, benzofluorenes, fluorenes, benzofurazans, benzodiphenylenes, benzofluoranthenes, benzanthracenes, benzacephenanthrylenes, bathophenanthrolines, indans, benzoquinolines, quinolines, pyrazines, quinolines, quinazoles, quinoxalines, imidazopyridines, indenes, indolines, thiazolines, bezopyrimidines, pyrimidines, benzimidazole, triazolopyrimidines, pyrazoles, tryptophans, phenanthrolines, benzooxadiazoles, benzoselenadiazole, benzocoumarins, chalcones, fluoranthenes, pyridoindoles, pentacenes, perylenes, phenatholines, phenazines, phenoxazines, phenoxathiins, phenothiazines, pyrroles, thiophenes, or heteroaromatics containing 5, 6, 7, 8, membered ring systems; 24) —NHCS groups or any other substituent that increases the hydrophilic, amphiphilic or lipophilic nature or stability of the compounds. It is recognized that such groups can affect the biological activity of the compounds in vivo.
  • The term “biologically active group” can be any group that selectively promotes the accumulation, elimination, binding rate, or tightness of binding in a particular biological environment. For example, one category of biologically active groups is the substituents derived from sugars, specifically: (1) aldoses such as glyceraldehyde, erythrose, threose, ribose, arabinose, xylose, lyxose, allose, altrose, glucose, mannose, gulose, idose, galactose, and talose; (2) ketoses such as hydroxyacetone, erythrulose, rebulose, xylulose, psicose, fructose, sorbose, and tagatose; (3) pyranoses such as glucopyranose; (4) furanoses such as fructo-furanose; (5) O-acyl derivatives such as penta-O-acetyl-α-glucose; (6) O-methyl derivatives such as methyl α-glucoside, methyl β-glucoside, methyl α-glucopyranoside, and methyl-2,3,4,6-tetra-O-methyl-glucopyranoside; (7) phenylosazones such as glucose phenylosazone; (8) sugar alcohols such as sorbitol, mannitol, glycerol, and myo-inositol; (9) sugar acids such as gluconic acid, glucaric acid and glucuronic acid, δ-gluconolactone, δ-glucuronolactone, ascorbic acid, and dehydroascorbic acid; (10) phosphoric acid esters such as α-glucose 1-phosphoric acid, α-glucose 6-phosphoric acid, α-fructose 1,6-diphosphoric acid, and α-fructose 6-phosphoric acid; (11) deoxy sugars such as 2-deoxy-ribose, rhammose (deoxy-mannose), and fructose (6-deoxy-galactose); (12) amino sugars such as glucosamine and galactosamine; muramic acid and neurarninic acid; (13) disaccharides such as maltose, sucrose and trehalose; (14) trisaccharides such as raffinose (fructose, glucose, galactose) and melezitose (glucose, fructose, glucose); (15) polysaccharides (glycans) such as glucans and mannans; and (16) storage polysaccharides such as α-amylose, amylopectin, dextrins, and dextrans.
  • Amino acid derivatives are also useful biologically active substituents, such as those derived from valine, leucine, isoleucine, threonine, methionine, phenylalanine, tryptophan, alanine, arginine, aspartic acid, cystine, cysteine, glutamic acid, glycine, histidine, proline, serine, tyrosine, asparagine and glutamine. Also useful are peptides, particularly those known to have affinity for specific receptors, for example, oxytocin, vasopressin, bradykinin, LHRH, thrombin and the like.
  • Another useful group of biologically active substituents are those derived from nucleosides, for example, ribonucleosides such as adenosine, guanosine, cytidine, and uridine; and 2′-deoxyribonucleosides, such as 2′-deoxyadenosine, 2′-deoxyguanosine, 2′-deoxycytidine, and 2′-deoxythymidine.
  • Another category of biologically active groups that is particularly useful is any ligand that is specific for a particular biological receptor. The term “ligand specific for a biological receptor” refers to a moiety that binds a receptor at cell surfaces, and thus contains contours and charge patterns that are complementary to those of the biological receptor. The ligand is not the receptor itself, but a substance complementary to it. It is well understood that a wide variety of cell types have specific receptors designed to bind hormones, growth factors, or neurotransmitters. However, while these embodiments of ligands specific for receptors are known and understood, the phrase “ligand specific for a biological receptor”, as used herein, refers to any substance, natural or synthetic, that binds specifically to a receptor.
  • Examples of such ligands include: (1) the steroid hormones, such as progesterone, estrogens, androgens, and the adrenal cortical hormones; (2) growth factors, such as epidermal growth factor, nerve growth factor, fibroblast growth factor, and the like; (3) other protein hormones, such as human growth hormone, parathyroid hormone, and the like; (4) neurotransmitters, such as acetylcholine, serotonin, dopamine, and the like; and (5) antibodies. Any analog of these substances that also succeeds in binding to a biological receptor is also included within the invention.
  • Particularly useful examples of substituents tending to increase the amphiphilic nature of the compounds include, but are not limited to: (1) short or long chain alcohols, such as, for example, —C12H24—OH; (2) fatty acids and their salts, such as, for example, the sodium salt of the long-chain fatty acid oleic acid; (3) phosphoglycerides, such as, for example, phosphatidic acid, phosphatidyl ethanolamine, phosphatidyl choline, phosphatidyl serine, phosphatidyl inositol, phosphatidyl glycerol, phosphatidyl 3′-O-alanyl glycerol, cardiolipin, or phosphatidyl choline; (4) sphingolipids, such as, for example, sphingomyelin; and (5) glycolipids, such as, for example, glycosyldiacylglycerols, cerebrosides, sulfate esters of cerebrosides or gangliosides. It would be known to those skilled in the art what other substituents, or combinations of the subsituents described, would be suitable for use in the invention.
  • The compounds of the present invention, or their pharmaceutically acceptable salts, solvates, prodrugs, or metabolites, can be administered to the host in a variety of forms adapted to the chosen route of administration, e.g., orally, intravenously, topically, intramuscularly or subcutaneously.
  • The active compound may be orally administered, for example with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets, or it may be incorporated directly with food. For oral therapeutic administration, the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least about 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may, for example, conveniently be between about 2 to about 60% of the weight of the administered product. The amount of active compound in such therapeutically useful compositions is can be selected so that a suitable dosage will be obtained. Preferred compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains between about 50 and 300 mg of active compound.
  • The tablets, troches, pills, capsules and the like may also contain the following: a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; a sweetening agent such as sucrose, lactose or saccharin; or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and formulations.
  • The active compound may also be administered parenterally or intraperitoneally. Solutions of the active compound as a free base or pharmacologically acceptable salt can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporanous preparation of sterile injectable solutions, dispersions, or liposomal or emulsion formulations. In all cases the form must be sterile and should be fluid to enable administration by a syringe. The form must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required additional ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze-drying technique, which yield a powder of the active ingredient plus any additional desired ingredient from previously sterile-filtered solutions thereof.
  • The new compounds of the invention may also be applied directly to tumors in the host whether internal or external, in topical compositions. Exemplary compositions include solutions of the new compounds in solvents, particularly aqueous solvents, most preferably water. Alternatively, for topical application particularly to skin tumors or psoriasis, the present new compounds may be dispersed in the usual cream or salve formulations commonly used for this purpose (such as liposomes, ointments, gels, hydrogels, cremes and oils) or may be provided in the form of spray solutions or suspensions that may include a propellant usually. employed in aerosol preparations.
  • As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Any conventional media or agent that is compatible with the active ingredient can be used in the therapeutic compositions of the invention. Supplementary active ingredients can also be incorporated into the compositions.
  • It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated. Each unit contains a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specifications for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of cardiovascular diseases, diseases of the skin, cancers and other superficial disease conditions in living subjects.
  • The present invention provides a method of treating live cells, which includes, but is not limited to, animals such as humans and other mammals. The “mammals” also include farm animals, such as cows, hogs and sheep, as well as pet or sport animals, such as horses, dogs and cats. The dosage of the pharmaceutical compositions of the invention is dependent on the method of administration, the patient's age, severity of the disease, and the like.
  • The compounds of the invention may be taken parentally or orally, generally being administered intravascularly, subcutaneously, or intramuscularly or interperitoneally. The subject compounds may also be administered by inhalation, perivascular delivery, pericardial delivery (into perivascular sac), periadvential delivery (e.g., using a hydrogel wrap around the vessel), endovascular balloon catheters with micropores, channels, transmural injection ports, and the like.
  • For local catheter-based delivery of the compounds of the invention, an infusate can be placed and pressurized to facilitate intramural and transmural penetration into the target vessel. Local delivery can also be enhanced by other mechanical and electrical means. The depth of the penetration of the subject compounds by this local delivery method is a function of pressure in the perfused segment and the dwell time. Although little attention has been paid to the quantitative characteristics of the compounds of the invention in this setting, deposition of the substance should obey the principles governing transmural convection and diffusion.
  • Delivery of the compounds of the invention may also be via antibody-drug conjugates, internalizing antibodies or antibody fragments conjugated to compounds into cells using endocytosis. The subject compounds may also be impregnated into stent struts for local delivery. The route of administration of these pharmaceutical preparations is not critical, but may be selected according to the dosage form, the patient's age, the severity of the disease to be treated and other factors.
  • The compounds of the invention may find use in conjunction with other interventions, diagnostics and therapies, where lower levels of other therapies having significant side effects may be used effectively to reduce the detrimental side effects. Adjunctive interventions may include, but are not limited to: balloon angioplasty, invasive and non-invasive surgical procedures, stent deployment, cutting balloons, embolic protection devices, rotational and directional atherectomy, eximer lasers and the like.
  • Adjunctive therapies may include, but are not limited to radiation therapy, chemotherapy, anti-platelet agents, vasodilators, antihypertensives, anti-arrhythmics, hyperthermia, cryotherapy, magnetic force, viral and non-viral gene therapy, pharmacogenetic therapy, antibodies, vaccines, glycoprotein IIb/IIIa Inhibitors, growth factors, peptides, DNA delivery, nucleic acids, anticancer drugs, steroid hormones, anti-inflammatories, proteins, anti-apoptotic therapies, anti-sense agents, immunotoxins, immunomodulators, antibody-drug conjugates, anti-proliferative therapies, drug eluting stents containing pharmacologically active agents, transplant products and processes, prostaglandins and catheter based devices to detect vulnerable plaques, hormone products, chelating agents, diuretics, cardiac glycosides, bronchodilators, antibiotics, antivirals, antitioxins, cyclosporins, thrombolytic agents, interferons, blood products such as parental iron and hemin, anti-fungal agents, antianginals, anticoagulants, analgesics, narcotics, neuromuscular blockers, sedatives, bacterial vaccines, viral vaccines, DNA or RNA of natural or synthetic origin including recombinent RNA and DNA, cytokines and their antagonists/inhibitors, chemokines and their antagonists/inhibitors,
  • Adjunctive diagnostics may include, but are not limited to: intravascular ultrasound imaging, angiography, quantitative vessel measurements and the use of radiological contrast agents, hormone products, chelating agents, diuretics, cardiac glycosides, bronchodilators, antibiotics, antivirals, antitoxins, cyclosporins, thrombolytic agents, interferons, blood products such as parental iron and hemin, anti-fungal agents, antianginals, anticoagulants, analgesics, narcotics, neuromuscular blockers, sedatives, bacterial vaccines, viral vaccines, DNA or RNA of natural or synthetic origin including recombinent RNA and DNA, cytokines and their antagonists/inhibitors, and chemokines and their antagonists/inhibitors.
  • Definitions
  • As used in the present application, the following definitions apply:
  • The term “alkyl” as used herein refers to substituted or unsubstituted, straight or branched chain groups, preferably having one to twenty, more preferably having one to six, and most preferably having from one to four carbon atoms. The term “C1-C20 alkyl” represents a straight or branched alkyl chain having from one to twenty carbon atoms. Exemplary C1-C20 alkyl groups include methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, t-butyl, pentyl, neo-pentyl, hexyl, isohexyl, and the like. The term “C1-C20 alkyl” includes within its definition the term “C1-C4 alkyl.” Such alkyl groups may themselves be ethers or thioethers, or aminoethers or dendrimers.
  • The term “cycloalkyl” represents a substituted or unsubstituted, saturated or partially saturated, mono- or poly-carbocyclic ring, preferably having 5-14 ring carbon atoms. Exemplary cycloalkyls include monocyclic rings having from 3-7, preferably 3-6, carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. An exemplary cycloalkyl is a C5-C7 cycloalkyl, which is a saturated hydrocarbon ring structure containing from five to seven carbon atoms.
  • The term “aryl” as used herein refers to an aromatic, monovalent monocyclic, bicyclic, or tricyclic radical containing 6, 10, 14, or 18 carbon ring atoms, which may be unsubstituted or substituted, and to which may be fused one or more cycloalkyl groups, heterocycloalkyl groups, or heteroaryl groups, which themselves may be unsubstituted or substituted by one or more suitable substituents. Illustrative examples of aryl groups include, but are not limited to, phenyl, napthalenes, anthracenes, benzopyrenes, quinolines, benzoquinolines, benzoperylene, benzofluorenes, fluorenes, benzofurazans, benzodiphenylenes, benzofluoranthenes, benzanthracenes, benzacephenanthrylenes, bathophenanthrolines, indans, benzoquinolines, quinolines, pyrazines, quinolines, quinazoles, quinoxalines, imidazopyridines, indenes, indolines, thiazolines, benzopyrimidines, pyrimidines, benzimidazole, triazolopyrimidines, pyrazoles, tryptophans, phenanthrolines, benzooxadiazoles, benzoselenadiazole, benzocoumarins, chalcones, fluoranthenes, pyridoindoles, pentacenes, perylenes, phenatholines, phenazines, phenoxazines, phenoxathiins, phenothiazines ad the like.
  • The term “halogen” represents chlorine, fluorine, bromine or iodine. The term “halocarbon” or “haloalkyl” represents one or more halogens bonded to a one or more carbon bearing groups. The term “heterohaloalkyl” represents for example halogenated alkylethers, halogenated alkyl amines, halogenated alkyl esters, halogenated alkyl amides, halogenated alkyl thioesters, halogenated alkyl thiols, where N, S, O, P atoms are present in the haloalkylated structure. The term. heteroalkyl represents for example ethers, alkylamines, alkylated thiols and alkylate phosphorus containing groups.
  • The term “carbocycle” represents a substituted or unsubstituted aromatic or a saturated or a partially saturated 5-14 membered monocyclic or polycyclic ring, such as a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring, wherein all the ring members are carbon atoms.
  • The term “electron withdrawing group” is intended to mean a chemical group containing an electronegative element such as halogen, sulfur, nitrogen or oxygen.
  • A “heterocycloalkyl group” is intended to mean a non-aromatic, monovalent monocyclic, bicyclic, or tricyclic radical, which is saturated or unsaturated, containing 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 ring atoms, and which includes 1, 2, 3, 4, or 5 heteroatoms selected from nitrogen, oxygen and sulfur, wherein the radical is unsubstituted or substituted, and to which may be fused one or more cycloalkyl groups, aryl groups, or heteroaryl groups, which themselves may be unsubstituted or substituted. Illustrative examples of heterocycloalkyl groups include, but are not limited to azetidinyl, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, tetrahydro-2H-1,4-thiazinyl, tetrahydrofuryl, dihydrofuryl, tetrahydropyranyl, dihydropyranyl, 1,3-dioxolanyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-oxathiolanyl, 1,3-oxathianyl, 1,3-dithianyl, azabicylo[3.2.1]octyl, azabicylo[3.3.1]nonyl, azabicylo[4.3.0]nonyl, oxabicylo[2.2.1]heptyl, 1,5,9-triazacyclododecyl, and the like.
  • A “heteroaryl group” is intended to mean an aromatic monovalent monocyclic, bicyclic, or tricyclic radical containing 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 ring atoms, including 1, 2, 3, 4, or 5 heteroatoms selected from nitrogen, oxygen and sulfur, which may be unsubstituted or substituted, and to which may be fused one or more cycloalkyl groups, heterocycloalkyl groups, or aryl groups, which themselves may be unsubstituted or substituted. Illustrative examples of heteroaryl groups include, but are not limited to, thienyl, pyrrolyl, imidazolyl, pyrazolyl, furyl, isothiazolyl, furazanyl, isoxazolyl, thiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, benzo[b]thienyl, naphtho[2,3-b]thianthrenyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathienyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, quinoxyalinyl, quinzolinyl, benzothiazolyl, benzimidazolyl, tetrahydroquinolinyl, cinnolinyl, pteridinyl, carbazolyl, beta-carbolinyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, phenazinyl, isothiazolyl, phenothiazinyl, and phenoxazinyl and the like.
  • The term “leaving group” as used herein refers to any group that departs from a molecule in a substitution reaction by breakage of a bond. Examples of leaving groups include, but are not limited to, halides, tosylates, arenesulfonates, alkylsulfonates, and triflates.
  • Suitable protecting groups are recognizable to those skilled in the art. Examples of suitable protecting groups can be found in T. Green & P. Wuts, Protective Groups in Organic Synthesis (2d ed. 1991), which is hereby incorporated by reference herein in its entirety.
  • Suitable salt anions include, but are not limited to, inorganics such as halogens, pseudohalogens, sulfates, hydrogen sulfates, nitrates, hydroxides, phosphates, hydrogen phosphates, dihydrogen phosphates, perchlorates, and related complex inorganic anions; and organics such as carboxylates, sulfonates, bicarbonates and carbonates.
  • Examples of substituents for alkyl and aryl groups include mercapto, thioether, nitro (NO2), amino, aryloxyl, halogen, hydroxyl, alkoxyl, and acyl, as well as aryl, cycloalkyl and saturated and partially saturated heterocycles. Examples of substituents for cycloalkyl groups include those listed above for alkyl and aryl, as well as alkyl.
  • Exemplary substituted aryls include a phenyl or naphthyl ring substituted with one or more substituents, preferably one to three substituents, independently selected from halo, hydroxy, morpholino(C1-C20)alkoxy carbonyl, pyridyl (C1-C20)alkoxycarbonyl, halo (C1-C20)alkyl, C1-C20 alkyl, C1-C20 alkoxy, carboxy, C1-C20 alkocarbonyl, carbamoyl, N-(C1-C20)alkylcarbamoyl, amino, C1-C20alkylamino, di(C1-C20)alkylamino or a group of the formula —(CH2)a—R7 where a is 1, 2, 3, 4, 5; and R7 is hydroxy, C1-C20 alkoxy, carboxy, C1-C20 alkoxycarbonyl, amino, carbamoyl, C1-C20 alkylamino or di(C1-C20)alkylamino, sulfonic acids, sulfonic esters, sulfonic amides, amides, esters and the like.
  • Another substituted alkyl is halo(C1-C20)alkyl, which represents a straight or branched alkyl chain having at least one halogen atom attached to it. Exemplary halo(C1-C20)alkyl groups include chloromethyl, 2-bromoethyl, 1-chloroisopropyl, 3-fluoropropyl, 2,3-dibromobutyl, 3-chloroisobutyl, trifluoromethyl, trifluoroethyl, and the like.
  • Another substituted alkyl is hydroxy (C1-C20)alkyl, which represents a straight or branched alkyl chain having from one to twenty carbon atoms with a hydroxy group attached to it. Exemplary hydroxy(C1-C20)alkyl groups include hydroxymethyl, 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxyisopropyl, 4-hydroxybutyl, and the like.
  • Yet another substituted alkyl is C1-C20 alkylthio(C1-C20)alkyl, which is a straight or branched C1-C20 alkyl group with a C1-C20 alkylthio group attached to it. Exemplary C1-C20 alkylthio(C1-C20)alkyl groups include methylthiomethyl, ethylthiomethyl, propylthiopropyl, sec-butylthiomethyl, and the like.
  • Yet another exemplary substituted alkyl is heterocycle(C1-C20)alkyl, which is a straight or branched alkyl chain having from one to twenty carbon atoms with a heterocycle attached to it. Exemplary heterocycle(C1-C20)alkyls include pyrrolylmethyl, quinolinylmethyl, 1-indolylethyl, 2-furylethyl, 3-thien-2-ylpropyl, 1-imidazolylisopropyl, 4-thiazolylbutyl and the like.
  • Yet another substituted alkyl is aryl(C1-C20)alkyl, which is a straight or branched alkyl chain having from one to twenty carbon atoms with an aryl group attached to it. Exemplary aryl(C1-C20)alkyl groups include phenylmethyl, 2-phenylethyl, 3-naphthyl-propyl, 1-naphthylisopropyl, 4-phenylbutyl and the like.
  • The heterocycloalkyls and the heteroaryls can, for example, be substituted with 1, 2 or 3 substituents independently selected from halo, halo(C1-C20)alkyl, C1-C20 alkyl, C1-C20 alkoxy, carboxy, C1-C20 alkoxycarbonyl, carbamoyl, —(C1-C20)alkylcarbamoyl, amino, C1-C20alkylamino, di(C1-C20)alkylamino or a group having the structure —(CH2)a—R7 where a is 1, 2, 3, 4, 5 and R7 is hydroxy, C1-C20 alkoxy, carboxy, C1-C20 alkoxycarbonyl, amino, carbamoyl, C1-C20alkylamino or di(C1-C20)alkylamino.
  • Examples of substituted heterocycloalkyls include, but are not limited to, 3-N-t-butyl carboxamide decahydroisoquinolinyl and 6-N-t-butyl carboxamide octahydro-thieno[3,2-c]pyridinyl. Examples of substituted heteroaryls include, but are not limited to, 3-methylimidazolyl, 3-methoxypyridyl, 4-chloroquinolinyl, 4-aminothiazolyl, 8-methylquinolinyl, 6-chloroquinoxalinyl, 3-ethylpyridyl, 6-methoxybenzimidazolyl, 4-hydroxyfuryl, 4-methylisoquinolinyl, 6,8-dibromoquinolinyl, 4,8-dimethylnaphthyl, 2-methyl-1,2,3,4-tetrahydroisoquinolinyl, N-methyl-quinolin-2-yl, 2-t-butoxycarbonyl-1,2,3,4-isoquinolin-7-yl and the like.
  • A “pharmaceutically acceptable solvate” is intended to mean a solvate that retains the biological effectiveness and properties of the biologically active components of the inventive compounds.
  • Examples of pharmaceutically acceptable solvates include, but are not limited to, compounds prepared using water, isopropanol, ethanol, DMSO, and other excipients generally reffered to as GRAS ingredients.
  • In the case of solid formulations, it is understood that the compounds of the inventive methods may exist in different polymorph forms, such as stable and metastable crystalline forms and isotropic and amorphous forms, all of which are intended to be within the scope of the present invention.
  • A “pharmaceutically acceptable salt” is intended to mean those salts that retain the biological effectiveness and properties of the free acids and bases and that are not biologically or otherwise undesirable.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, citrates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, hydroxybutyrates, glycolates, tartrates, methanesulfoantes, propanesulfonates, naphthalene-1-sulfonates, naphthalene-2-sulfonates, and mandelates.
  • If a compound of the present invention is a base, the desired salt may be prepared by any suitable method known to the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, lactic acid, salicylic acid, pyranosidyl acids such as glucuronic acid and galacturonic acid, alpha-hydroxy acids such as citric acid and tartaric acid, amino acids such as aspartic acid and glutamic acid, aromatic acids such as benzoic acid and cinnamic acid, sulfonic acids such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.
  • If a compound of the present invention is an acid, the desired salt may be prepared by any suitable method known to the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), or an alkali metal or alkaline earth metal hydroxide or the like. Illustrative examples of suitable salts include organic salts derived from amino acids such as glycine and arginine; ammonia; primary, secondary and tertiary amines; cyclic amines such as piperidine, morpholine and piperazine; and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • In summary, there is a real need for better “shorter wavelength” absorbing photodynamic agents that do not display red absorptions, that are cleared rapidly from normal tissues, especially skin, and agents that may be used as MRI diagnostics or radiodiagnostic agents in addition to therapeutics. Additionally, as more disease indications are realized, shorter wavelength light may be equally important in other PDT applications that only require short wavelength excitation to effect a therapy. Such applications may be, for example, in hollow organ disease (for example lung cancers, barrets esophagus), or in diseases of the skin (for example psoriasis, actinic keratosis, acne vulgaris). The invention disclosed herein describes the synthesis of metallated photosensitizers having ring systems that have shown excellent efficacy in advanced animal model systems as well as preferred uptake in the target tissue, with excellent clearance characteristics and low toxicity. (See co-pending application filed on May 31, 2001 entitled “Metallotetrapyrrolic Photosensitizing Agents For Use In Photodynamic Therapy,” inventors Byron C. Robinson, Ian M. Leitch, Stephanie Greene, and Steve Rychnovsky, Attorney Docket No. 07328-0015.)
  • The compounds of the invention are intended for use not only for effective photodynamic therapy treatment but also as MRI and radiodiagnostic diagostic agents. Such compounds may be used to diagnose, locate or treat cardiovascular disease and normal or abnormal conditions of the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system including bone, connective tissue, cartilage and skeletal muscle, pulmonary system, gastrointestinal system including the liver, reproductive system, skin, immune system, cardiovascular system, urinary system, ocular system, auditory or olfactory system. In particular, photoactive derivatives of porphyrins and azaporphyrins are particularly advantageous where shorter wavelengths of light are necessary to effect a photodynamic response.
  • EXAMPLES
  • Preparation of compounds according to the invention is illustrated by reference to the following non-limiting examples. It will be appreciated by persons skilled in the art with the teachings of the examples and the rest of the specification (i) how the chemistry may be applied to other peripheral groups on tetrapyrrolic ring structures that fall within the scope of this invention and (ii) that other synthetic routes may be suitable for preparation of the desired compounds.
  • Example 1 8,12-Diethyl-3,7,13,17-tetramethylporphyrin-2,18-dipropionylhydrazide
  • 1.3 g of 8,12-diethyl-3,7,13,17-tetramethylporphyrin-2,18-diyl-di(methoxycarbonylpropionic acid) was modified according to H. Fischer, E. Haarer and F. Stadler, Z. Physiol. Chem. 241, 209 (1936) by treatment with hydrazine hydrate (7 mL of an 80% water solution) in pyridine (30 mL) at room temperature overnight. The solvent was removed by rotoevaporation and the solid suspended/dissolved in methanol (10 mL). Water (30 mL) was added and the methanol removed by rotary evaporation. The precipitated porphyrin was collected by filtration and dried to give the title compound. Yield: 1.25 g of a reddish-brown powder.
  • Example 2 Zinc 8,12-Diethyl-3,7,13,17-tetramethylporphyrin-2,18-dipropionylhydrazide
  • The compound produced in example 1 (100 mg) was dissolved in chloroform/methanol (80:20) and zinc acetate (200 mg) was added. The solution was refluxed until complete by UV. The solution was evaporated to dryness and redissolved in dichloromethane (50 mL). Water (100 mL) was added and the dichloromethane removed by rotary evaporation. The precipitated solid was collected by filtration, washed with water (50 mL) and dried. Yield of the title compound=110 mg.
  • Example 3 Platinum 8,12-Diethyl-3,7,13,17-tetramethylporphyrin-2,18-dipropionylhydrazide
  • Platinum 8,12-Diethyl-3,7,13,17-tetramethylporphyrin-2,18-dipropionic acid methyl ester (1.2 g) was modified according to H. Fischer, E. Haarer and F. Stadler, Z. Physiol. Chem. 241, 209 (1936) by treatment with hydrazine hydrate (7 mL of an 80% water solution) in pyridine (30 mL) at room temperature. The solution was evaporated to dryness, dissolved/suspended in methanol (10 mL) and water (30 mL) was added. The methanol was removed by rotary evaporation and the precipitated porphyrin collected by filtration and dried. Yield: 1.25 g of an orange-red powder.
  • Example 4 8,12-Diethyl-3,7,13,17-tetramethylporphyrin-2-{N(6-tert-butoxycarbonylaminohexyl)propionylamide}-18-propionic Acid & 8,12-Diethyl-3,7,13,17-tetramethylporphyrin-2,18-{N,N′-bis-(6-tert-butoxycarbonylaminohexyl)}dipropionylamide
  • 8,12-Diethyl-3,7,13,17-tetramethylporphyrin-2,18-dipropionic acid (1.8 g), 1-hydroxybenzotriazole (810 mg), N-BOC-1,6-diaminohexane hydrochloride (1.52 g) and triethylamine (608 mg) is dissolved in dimethylformamide (300 mL). The solution is stirred under argon and cooled to −10° C. Dicyclohexylcarbodiimide (1.24 g) is added and the solution is stirred for 1 hr at −10° C., then left to warm to room temperature. After three hours the solution is evaporated to dryness and the residue dissolved in dichloromethane (50 mL). The solution is washed with a saturated sodium bicarbonate solution. The dichloromethane layer was dried over sodium sulfate and filtered and evaporated. The crude residue is chromatographed on silica using dichloromethane/methanol (0-10%) as eluent and the two fractions collected. The diamide is eluted first (800 mg), followed sluggishly by the mono amide title compound (1500 mg).
  • Example 5 8,12-Diethyl-3,7,13,17-Tetramethylporphyrin-2-{N-(6-Aminohexyl)-Propionylamide}-18-Propionic Acid
  • The mono amide produced in example 4 (500 mg) is dissolved in 2N hydrochloric acid in glacial acetic acid (10 mL). When no more starting material could be detected, the solution is evaporated to dryness and dissolved in distilled water. A saturated sodium bicarbonate solution is added dropwise until the porphyrin precipitated. The porphyrin is collected by filtration and dried. Yield of title compound=400 mg.
  • Example 6 8,12-Diethyl-3,7,13,17-Tetramethylporphyrin-2,18-{N,N′-Bis-(6-Aminohexyl}Propionylamide
  • The di-amide produced in example 4 (500 mg) is dissolved in 2N hydrochloric acid in glacial acetic acid (10 mL) and the solution is stirred overnight. When no more starting material could be detected, the solution is evaporated to dryness and dissolved in distilled water. A saturated sodium bicarbonate solution is added dropwise until the porphyrin precipitated. The porphyrin was collected by filtration and dried. Yield of title compound=400 mg.
  • Example 7 Zinc 8,12-Diethyl-3,7,13,17-Tetramethylporphyrin-2-{N-(6-Aminohexyl}Propionylamide-18-Propionic Acid
  • The porphyrin prepared in Example 5 (100 mg) is metallated as described in example 2. Yield of title compound=(100 mg).
  • Example 8 Zinc 8,12-Diethyl-3,7,13,17-Tetramethylporphyrin-2,18-(N,N′-Bis-(6-Aminohexyl)Propionylamide
  • The porphyrin prepared in Example 6 (100 mg) is metallated as described in example 2. Yield of title compound=(100 mg).
  • Example 9 Platinum 8,12-diethyl-3,7,13,17-tetramethylporphyrin-2,18-{N,N′-bis-(6-tert-butoxycarbonylaminohexyl}dipropionylamide
  • The porphyrin prepared in Example 2 (100 mg) is hydrolyzed in THF (50 mL) containing KOH/methanol (0.4 g; 5 mL). The solution is neutralized using acetic acid and evaporated to 10 mL. Water (50 mL) is added and the precipitated porphyrin collected by filtration and dried under vacuum. The diacid porphyrin is converted to the protected amide according to example 4, except that the solution is reacted over night. The diamide protected compound is chromatographed on silica using 5% methanol/dichloromethane as eluent. Yield of title compound=85 mg.
  • Example 10 Platinum 8,12-Diethyl-3,7,13,17-Tetramethylporphyrin-2,18-{N,N′-Bis-(6-Aminohexyl)}Propionylamide
  • The porphyrin of example 9 (85 mg) is dissolved in dichloromethane (20 mL) and trifluoroacetic acid (10 mL) is added. The solution is stirred for 4 hrs, after which the solvent is removed by rotary evaporation. The compound is partially dissolved in methanol and triethylamine (0.4 mL) added. The precipitated porphyrin is collected and washed with water and dried. Yield of title compound=72 mg.
  • Example 11 8,12-Diethyl-3,7,13,17-Tetramethylporphyrin-2,18-{N,N′-Bis-(2-Aminoethyl}Propionylamide
  • 8,12-diethyl-3,7,13,17-tetramethylporphyrin-2,18-propionic acid (250 mg) is suspended in dichloromethane (50 mL) and oxalyl chloride (5 mL) is added. The solution is refluxed for 2 hrs under a dry inert atmosphere and the solvent removed by rotary evaporation. The solid is redissolved in dichloromethane (dry, 20 mL) and ethylenediamine (4 mL) is added all at once. The resulting solution is stirred at room temperature for 2 hrs and the solvent removed by rotary evaporation. The solid is suspended in methanol and triethylamine (5 drops) added. The flocculated porphyrin is collected by filtration, dissolved in dichloromethane (30 mL) and chromatographed on silica using 10-15% methanol/dichloromethane (with 1% triethylamine) and the major red band collected. The solvent is removed and the porphyrin dissolved in dichloromethane and precipitated from methanol by slow evaporation of dichloromethane. The solid is collected and washed with methanol. Yield of title compound=210 mg.
  • Example 12 Gallium Hydroxide 8,12-Diethyl-3,7,13,17-Tetramethylporphyrin-2,18-{N,N′-Bis-(2-Aminoethyl)}Propionylamide
  • The porphyrin prepared in Example 11 (100 mg) is metallated by refluxing a solution of the porphyrin and gallium acetylacetonate (100 mg) in acetic acid for 1.5 hrs. The solvent is removed and the solid dissolved in dichloromethane (50 mL) and washed with a saturated sodium bicarbonate solution. The organic layer is dried over sodium sulfate, filtered, evaporated to 10 ml, and hexane (10 mL) is added. The dichloromethane is slowly removed by rotary evaporation and the precipitated porphyrin collected by filtration. Yield of title compound=(100 mg).
  • Example 13 Indium Hydroxide 8,12-diethyl-3,7,13,17-tetramethylporphyrin-2,18-{(N,N′-bis-(2-aminoethyl)}propionylamide
  • The porphyrin prepared in Example 11 (100 mg) is metallated by refluxing a solution of the porphyrin, indium chloride (100 mg) and sodium acetate (100 mg) in acetic acid for 2 hrs. The solvent is removed and the solid dissolved in dichloromethane (50 mL) and washed with a saturated sodium bicarbonate solution. The organic layer is dried over sodium sulfate, filtered, evaporated to 10 ml, and hexane (10 mL) is added. The dichloromethane is slowly removed by rotary evaporation and the precipitated porphyrin collected by filtration. Yield of title compound=(100 mg).
  • Example 14 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-di(propionic Acid Methyl Ester)
  • To a solution of 5,5′-dicarboxy-3,3′-di(2-methoxycarbonylethyl)-4,4′-dimethylpyrromethane (5 g) in methanol (70 mL) was added ammonium hydroxide (2.6 ml) and the solution stirred until the dipyrromethane had dissolved. 2-bromo-5-formyl-3,4-diethylpyrrole (5.3 g) and HBr (33%, 25 mL) were added. The solution was stirred at room temperature for 2 hrs after which time the solid 1,19-dibromobiladiene was filtered and dried to yield=7.2 g. A smaller amount of 1,19-dibromobiladiene (3 g) was refluxed in methanol containing sodium azide (4 g) for 4 hrs. The solvent was removed and the residue dissolved in dichloromethane and chromatographed on silica using dichloromethane as eluent. The major purple band was collected and evaporated to dryness. The compound was dissolved in dichloromethane (50 mL) and methanol (50 mL) added. The dichloromethane was removed by rotary evaporation and the precipitated azaporphyrin collected by filtration. Yield of title compound=1.7 g.
  • Example 15 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-di(propanyl Hydrazide)
  • The azaporphyrin synthesized in example 13 (250 mg) was modified according to H. Fischer, E. Haarer and F. Stadler, Z. Physiol. Chem. 241, 209 (1936) by treatment with hydrazine hydrate (1.5 mL of an 80% water solution) in pyridine (20 mL) at room temperature. The solution was evaporated to dryness, dissolved/suspended in methanol (10 mL) and water (30 mL) was added. The methanol was removed by rotary evaporation and the precipitated porphyrin collected by filtration and dried. Yield of title compound: 250 g of a purple powder.
  • Example 16 Zinc 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-di(propanyl Hydrazide)
  • The azaporphyrin prepared in example 14 (100 mg) was metallated according to example 2. Yield of title compound 110 mg.
  • Example 17 Gallium 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-di(propanyl Hydrazide)
  • The azaporphyrin prepared in example 14 (100 mg) was metallated according to example 12. Yield of title compound=110 mg.
  • Example 18 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-(N,N′-bis-(2-aminoethyl)propionylamide
  • 8,12-diethyl-3,7,13,17-tetramethylporphyrin-2,18-propionic acid potassium salt (250 mg)(prepared by hydrolysis of the azaporphyrin prepared in example 14 with KOH/methanol) was converted to the title compound via the procedure described in example 11. Yield=200 mg.
  • Example 19 Zinc 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-{N,N′-bis-(2-aminoethyl)}propionylamide
  • The porphyrin prepared in example 18 (100 mg) was metallated by the procedure of example 14. Yield of title compound=100 mg.
  • Example 20 Gallium Hydroxyl 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-(N,N′-bis-(2-aminoethyl)propionylamide
  • The porphyrin prepared in example 18 (100 mg) was metallated by the procedure of example 12. Yield of title compound=100 mg.
  • Example 21 Indium Hydroxyl 7,8,12,13-tetraethyl-12,17-dimethyl-10-azaporphyrin-2,18-(N,N′-bis-(2-aminoethyl)propionylamide
  • The porphyrin prepared in example 18 (100 mg) was metallated by the procedure of example 13. Yield of title compound=100 mg.
  • Example 22 8,12-Diethyl-3,8,13,17-tetramethyl-2,18-bis{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-17-oxa-4,5,8,11,14-pentaazanonadec-1-yl}porphyrin
  • 806.8 mg (2 mmol) of 3-ethoxy-carbonylmethyl-6-[2-(2,6-dioxomorpholino)ethyl]-3,6-diazaoctanedioic acid (DTPA-monoethylester-monoanhydride) is suspended in 250 ml of absolute dimethylformamide. It is then covered with a layer of nitrogen, 1.0 g (10 mmol) of triethylamine and 593 mg (1 mmol) of the title compound synthesized via Example I are added, and the resulting reaction mixture is stirred for 3 days at room temperature. After the reaction is complete, it is filtered, the solvent is drawn off in a vacuum, and the remaining oil is pulverized with 500 ml of diethyl ether. The precipitated solid is filtered off and washed with diethyl ether and n-hexane. For purification, it is chromatographed on silica gel RP-18 (eluant: H2O/tetrahydrofuran: 0-30%). Yield of title compound: 1.30 g of a reddish-brown powder
  • Example 23 [8,12-Diethyl-3,8,13,17-tetramethyl-2,18-bis{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-17-oxa-4,5,8,11,14-pentaazanonadec-1-yl}porphyrinato(2-)]-zinc
  • The zinc porphyrin (658 mg) of example 2 is reacted as described in example 22. Yield of title compound=1.0 g
  • Example 24 [8,12-Diethyl-3,8,13,17-tetramethyl-2,18-bis{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-17-oxa-4,5,8,11,14-pentaazanonadec-1-yl}porphyrinato(2-)]platinum
  • The platinum porphyrin of example 3 (786 mg) is reacted as described in example 22. Yield of title compound=1.5 g
  • Example 25 [8,12-Diethyl-3,8,13,17-tetramethyl-2,-propionicacid-18-{3,12,22-trioxo-14,17,20-tris(carboxymethyl)-23-oxa-4,11,14,17,20-pentaazapentacosan-1-yl}porphyrin
  • The mono-hexylamine porphyrin (663 mg) of example 5 is reacted analogously as described in example 22, except using DTPA-monoethylester-monoanhydride (404 mg) and triethylamine (0.5 g). Yield of title compound=900 mg.
  • Example 26 [8,12-Diethyl-3,8,13,17-tetramethyl-2,18-{3,12,22-trioxo-14,17,20-tris(carboxymethyl)-23-oxa-4,11,14,17,20-pentaazapentacosan-1-yl}porphyrin
  • The dihexylamine porphyrin (663 mg) of example 5 is reacted as described in example 22, using DTPA-monoethylester-monoanhydride (808 mg) and triethylamine (1.0 g). Yield of title compound=1.3 g.
  • Example 27 [7,8,12,13-Tetraethyl-3,17-dimethyl-2,18-{3,8,18-trioxo-10,13,16-tris(carboxymethyl)-19-oxa-4,7,10,13,16-pentaazaheneicosan-1-yl}-10-azaporphyrin
  • The diamine azaporphyrin (663 mg) of example 18 is reacted as described in example 24, using DTPA-monoethylester-monoanhydride (808 mg) and triethylamine (1.0 g). Yield of title compound=1.32 g.
  • Example 28 [7,8,12,13-Tetraethyl-3,17-dimethyl-2,18-{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-17-oxa-4,5,8,11,14-pentaazanonadec-1-yl}-10-azaporphyrinato(2-)]zinc
  • The zinc dihydrazide azaporphyrin (686 mg) of example 16 is reacted as described in example 22, using DTPA-monoethylester-monoanhydride (808 mg) and triethylamine (1.0 g). Yield of title compound=1.22 g.
  • Example 29 [7,8,12,13-Tetraethyl-3,17-dimethyl-2,18-{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-17-oxa-4,5,8,11,14-pentaazanonadec-1-yl}-10-azaporphyrinato(2-)]galliumhydroxide
  • The gallium dihydrazide azaporphyrin (707 mg) of example 17 is reacted as described in example 22, using DTPA-monoethylester-monoanhydride (808 mg) and triethylamine (1.0 g). Yield of title compound=1.31 g.
  • Example 30 [7,8,12,13-Tetraethyl-3,17-dimethyl-2,18-{3,8,18-trioxo-10,13,16-tris(carboxymethyl)-19-oxa-4,7,10,13,16-pentaazaheneicosan-1-yl}-10-azaporphyrinato(2-)]zinc
  • The zinc azaporphyrin of example 19 (742 mg) is reacted as described in example 22, using DTPA-monoethylester-monoanhydride (808 mg) and triethylamine (1.0 g). Yield of title compound=1.35 g.
  • Example 31 7,8,12,13-tetraethyl-12,17-dimethyl-2-(3-carboxypropyl)-18-(3-methoxycarbonylpropyl)-10-azaporphyrin
  • The dimethyl ester azaporphyrin of example 14 (1.0 g, 1.6 mmol) is dissolved in THF (200 ml) and a solution of KOH (100 mg)/MeOH (5 mL) is added. The solution is closely monitored until only a trace of starting material remains and the major product is the mono-hydrolyzed azaporphyrin. Acetic acid (107 mg) is added and the solution diluted with water (100 mL). The THF was removed by rotary evaporation and the precipitated azaporphyrins were collected and washed with methanol (20 mL) and dried. The solid was dissolved in dichloromethane containing 2% methanol and the solution is chromatographed on silica using 2% methanol/dichloromethane as eluent. The second major fraction is collected and evaporated to dryness. Yield of title compound=0.62 g.
  • Example 32 7,8,12,13-tetraethyl-12,17-dimethyl-2-{N-(2-aminoethyl)propionylamide}-18-(3-methoxycarbonylpropyl)-10-azaporphyrin
  • The monoacid azaporphyrin prepared in example 30 (500 mg) is dissolved in dichloromethane (50 ml) and oxalyl chloride (5 ml) added. The solution is refluxed under dry conditions for 2 hrs and then evaporated to dryness, care being taken not to expose the crude material to moisture. The residue is dissolved in dichloromethane (50 mL, dry) and ethylene diamine (3 ml, dry) added all at once. The resulting solution is washed with a saturated sodium bicarbonate solution, followed by water and the organic layer is collected, dried over sodium sulfate, filtered and evaporated. The crude reaction mixture is chromatographed on silica using 4% methanol/dichloromethane/0.5% triethylamine as eluent, and the major fraction collected. The dichloromethane is removed by rotary evaporation and the precipitated porphyrin collected by filtration and dried. Yield of title compound=0.51 g.
  • Example 33 [7,8,12,13-Tetraethyl-3,17-dimethyl-2-(3-methoxycarbonylpropyl)-18-{3,8,18-trioxo-10,13,16-tris(carboxymethyl)-19-oxa-4,7,10,13,16-pentaazaheneicosan-1-yl}-10-azaporphyrin
  • The azaporphyrin of example 32 (0.5 g, 0.76 mmol) is reacted as described in example 22, using DTPA-monoethylester-monoanhydride (404 mg) and triethylamine (0.5 g). Yield of title compound=0.8 g.
  • Example 34 8,12-Diethyl-3,8,13,17-tetramethyl-2,18-bis{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-4,5,8,11,14-pentaazahexadecanoato-1-yl}porphyrin
  • The ligand that is produced by Example 22 (1.0 g, 0.74 mmol) is dissolved in THF (50 mL) and water (350 mL). Sodium hydroxide solution (10 mol) is added and it is stirred overnight at room temperature. After the ester groups have been completely saponified, the THF is removed by rotoevaporation. A pH of 4 is set with concentrated hydrochloric acid. It is evaporated to the dry state in a vacuum. The residue is chromatographed on RP 18 (eluant: H2O/tetrahydrofuran/gradient). Yield of title compound: 0.90 g of a reddish-brown powder.
  • Example 35 [8,12-Diethyl-3,8,13,17-tetramethyl-2,18-bis{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-4,5,8,11,14-pentaazahexadecanoato-1-yl}porphyrinato(2-)]zinc
  • The esters on the ligand produced by Example 23 (1.3 g, 0.885 mmol) are hydrolyzed and acidified as described in example 34. Yield of title compound=1.12 g.
  • Example 36 [8,12-Diethyl-3,8,13,17-tetramethyl-2,18-bis{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-4,5,8,11,14-pentaazahexadecanoato-1-yl}porphyrinato(2-)]platinum
  • The esters on the ligand produced by Example 24 (1.0 g, 0.62 mmol) are hydrolyzed and acidified as described in example 34. Yield of title compound=1.12 g.
  • Example 37 [7,8,12,13-Tetraethyl-3,17-dimethyl-2,18-{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-4,5,8,11,14-pentaazahexadecanoato-1-yl}-10-azaporphyrinato(2-)]zinc
  • The esters on the ligand produced in Example 28 (1.0 g, 0.69 mmol) are hydrolyzed and acidified as described in example 34. Yield of title compound=0.85 g.
  • Example 38 [7,8,12,13-Tetraethyl-3,17-dimethyl-2-propionic acid-18-{3,8,18-trioxo-10,13,16-tris(carboxymethyl)-4,7,10,13,16-pentaazaheneicosan-1-yl}-10-azaporphyrin
  • The esters on the ligand produced in Example 33 (1.0 g, 1 mmol) are hydrolyzed and acidified as described in example 34. Yield of title compound=0.85 g.
  • Example 39 [7,8,12,13-Tetraethyl-3,17-dimethyl-2-18-{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-4,5,8,11,14-pentaazahexadecanoato-1-yl}-10-azaporphyrinato(2-)]gallium Chloride
  • The esters on the ligand produced in Example 29 (1.0 g, 0.68 mmol) are hydrolyzed and acidified as described in example 34. Yield of title compound=0.85 g.
  • Example 40 7,8,12,13-Tetraethyl-3,17-dimethyl-2-18-{3,8,18-trioxo-10,13,16-tris(carboxymethyl)-4,7,10,13,16-pentaazaheneicosan-1-yl}-10-azaporphyrin
  • The esters on the ligand produced in Example 27 (1.0 g, 0.7 mmol) are hydrolyzed and acidified as described in example 34. Yield of title compound=0.82 g.
  • Example 41 Mu-[(16,16′-[zinc(II)-8,12-Diethyl-3,8,13,17-tetramethylporphyrin-2,18-diyl]-bis[3,6,9-tris(carboxymethyl)-11,14-dioxo-3,6,9,12,13-pentaazahexadecanoato]}(8-)]}digadolinato(2-), Disodium
  • The ligand produced by example 35 (200 mg, 0.14 mmol) is dissolved in water (100 mL), and gadolinium chloride (74 mg, 0.28 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium chloride is added, stirring is continued overnight at room temperature. The solvent is drawn off in a vacuum, and the residue is chromatographed on silica gel RP-18 (eluent:water/THF: 0-30%). Yield of title compound=260 mg.
  • Example 42 Mu-[(16,16′-[Platinum(II)-8,12-Diethyl-3,8,13,17-tetramethylporphyrin-2,18-diyl]-bis[3,6,9-tris(carboxymethyl)-11,14-dioxo-3,6,9,12,13-pentaazahexadecanoato]}(8-)]}digadolinato(2-), Disodium
  • The ligand produced by example 36 (200 mg, 0.13 mmol) is dissolved in water (100 mL), and gadolinium chloride (69 mg, 0.26 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium chloride is added, stirring is continued overnight at room temperature. The solvent is drawn off in a vacuum, and the residue is chromatographed on silica gel RP-18 (eluent:water/THF: 0-30%). Yield of title compound 247 mg.
  • Example 43 {Mu-[(16,16′-[8,12-Diethyl-3,8,13,17-tetramethylporphyrin-2,18-diyl]-bis[3,6,9-tris(carboxymethyl)-11,14-dioxo-3,6,9,12,13-pentaazahexadecanoato]}(8-)]}digadolinato(2-), Disodium
  • The ligand produced by example 34 (200 mg, 0.15 mmol) is dissolved in water (10 mL), and gadolinium chloride (79 mg, 0.30 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium chloride is added, stirring is continued overnight at room temperature. The solvent is drawn off in a vacuum, and the residue is chromatographed on silica gel RP-18 (eluent:water/THF: 0-30%). Yield of title compound=258 mg.
  • Example 44 {Mu-[(16,16′-[Zinc(II)[7,8,11,12-tetraethyl-13,17-dimethyl-10-azaporphyrin-2,18-diyl]-bis[3,6,9-tris(carboxymethyl)-11,14-dioxo-3,6,9,12,13-pentaazahexadecanoato]}(8-)]}digadolinato(2-), Disodium
  • The ligand produced by example 37 (200 mg, 0.15 mmol) is dissolved in water (100 mL), and gadolinium chloride (79 mg, 0.30 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium chloride is added, stirring is continued overnight at room temperature. The solvent is drawn off in a vacuum, and the residue is chromatographed on silica gel RP-18 (eluent:water/THF: 0-30%). Yield of title compound=260 mg.
  • Example 45 {Mu-[18′-[Zinc(II)[7,8,11,12-tetraethyl-13,17-dimethyl-10-azaporphyrin-2-(3-carboxypropanyl)-18-yl]-[{3,6,9-tris(carboxymethyl)-11,16-dioxo-3,6,9,12,15-pentaazaheneicosanato}](8-)]}digadolinato(2-), Disodium
  • The ligand produced by example 38 (200 mg, 0.21 mmol) is dissolved in water (100 mL), and gadolinium chloride (110 mg, 0.42 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium chloride is added, stirring is continued overnight at room temperature. The solvent is drawn off in a vacuum, and the residue is chromatographed on silica gel RP-18 (eluent:water/THF: 0-30%). Yield of title compound=260 mg.
  • Example 46 8,12-Diethyl-3,7,13,17-tetramethyl-2,18-(3-methoxycarboxylpropyl)-5,15-diazaporphyrin
  • This compound was prepared via the method of Neya, S., Hori, H., Imai, K., Konishi, Y. K., Suzuki, H., Shiro, Y., Lizuka, T., Funasaki, N., J. Biochem, 1997, 121, 654. The free bases of 5,5′-dibromo-3,3′-diethyldipyrromethene (2.00 g, 5.18 mmol) and 5,5′-dibromo-3,3′-di(methoxycarbonylethyl)-4,4′-dimethyldipyrromethene (2.60 g, 5.18 mmol) and sodium azide (10 g, 154 mmol) were placed in a round bottom flask containing methanol (1 L). the mixture was gently refluxed for 72 hrs. After solvent evaporation the residue was chromatographed on silica using chloroform as eluent. The title compound was eluted as the middle fraction of the three possible diazaporphyrins. The middle fraction was collected and recrystallized from dichloromethane/methanol. Yield=240 mg.
  • Example 47 8,12-Diethyl-3,7,13,17-tetramethyl-2,18-dipropionylhydrazide-5,15-diazaporphyrin
  • The compound produced by example 46 (1.0 g) was modified according to H. Fischer, E. Haarer and F. Stadler, Z. Physiol. Chem. 241, 209 (1936) by treatment with hydrazine hydrate (7 mL of an 80% water solution) in pyridine (30 mL) at room temperature overnight. The solvent was removed by rotoevaporation and the solid suspended/dissolved in methanol (10 mL). Water (30 mL) was added and the methanol removed by rotary evaporation. The precipitated porphyrin was collected by filtration and dried to give the title compound. Yield: 1.25 g of purple powder.
  • Example 48 8,12-Diethyl-3,7,13,17-tetramethyl-2,18-{3,6,16-trioxo-8,11,14-tris(carboxymethyl)-17-oxa-4,5,8,11,14-pentaazanonadec-1-yl}-5,15-diazaporphyrin
  • The compound produced by example 47 (500 mg, 0.83 mmol) is modified according to example 22 except using DTPA-monoethylester-monoanhydride (674 mg, 1.67 mmol) and triethylamine (1.0 g) Yield of title compound=990 mg.
  • Example 49 {Mu-[(16,16′-[8,12-Diethyl-3,7,13,17-tetramethyl-5,15-diazaporphyrin-2,18-diyl]-bis[3,6,9-tris(carboxymethyl)-11,14-dioxo-3,6,9,12,13-pentaazahexadecanoato]}(8-)]}digadolinato(2-), Disodium
  • The compound produced by example 48 (200 mg, 0.148 mmol) is dissolved in water (100 mL), and gadolinium chloride (78.2 mg, 0.296 mmol) and 2N aqueous sodium hydroxide solution are added alternately in portions, such that the pH of the reaction mixture remains between 6.8 and 7.2. After all of the gadolinium

Claims (120)

1. A compound of formula I:
Figure US20050226810A1-20051013-C00031
wherein R1-R12 can be the same or different and are selected from:
H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ether, polyether, alkoxy group, aryloxy group, haloalkoxy, group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)-alkoxy, CH(CH3)O-aryl;
(CH2)nO-alkoxy, or (CH2)nO-alkyl, where n is an integer from 0 to 8;
C(X)2C(X)3, where X is a halogen;
CO2R13, where R13 is selected from H, a physiologically acceptable counter ion, a C1-C20 straight or branched chain alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
(CH2)nOH, or (CH2)nOR14, where R14 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nCO2R15, (CHX)nCO2R15, or (CX2)nCO2R15, where X is a halogen and R15 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
CONH(R16), CONHNH(R16), CO(R16), CON(R16)2, CON(R16)(R17), (CH2)nCONH(R16), (CH2)nCON(R16)2, (CH2)nCOR16, (CH2)nCON(R16)(R17), (CX2)nCONH(R16), (CX2)nCON(R16)2, (CX2)nCON(R16)(R17), (CX2)nCOR16, (CH2)nCONHNH(R16), (CX2)nCONHNH(R16), (CHX)nCONH(R16), (CHX)nCONHNH(R16), (CHX)nCO(R16), (CHX)nCON(R16)2, or (CHX)nCON(R16)(R17), where X is a halogen and R16 and R17 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
S(R18), (CH2)nS(R18), (CH2)nNH(R18), (CH2)nNHNH(R18), (CH2)nN(R18)2, (CH2)nN(R18)(R19), or (CH2)nN(R18)(R19)(R20)+A, where R18, R19 and R20 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids (provided —NH(R18) is part of the amino acid), an amino acid ester, an amino acid amide, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R18, R19 and R20 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
(CH2)nOPO2OR21, (CH2)nPO(OR21)2, (CH2)nPO2R21, or (CH2)nPOR21 where R21 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nNHCOR22, or (CH2)nNHNHCOR22, where R22 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
SO3R23, SO2NHR23, SO2N(R23)2, SO2NHNHR23, or SO2R23, where R23 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and NHR23 can also be an amino acid, an amino acid salt, an amino acid ester residue, an amino acid amide residue[, and n is an integer between 0 and 4];
Aryl or substituted aryl, which may bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons; and
R1-R2, R4-R5, R7-R8, R10-R11, R2-R3, R5-R6, R8-R9, and R11-R12 may also possess the atoms necessary to form ring systems, which themselves may possess heteroatoms that may bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons;
with the proviso that at least one of the R1-R23 groups is linked via an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link, to a complexing agent of general formula 11a, 11b, 11c, 11d, 11e;
Figure US20050226810A1-20051013-C00032
wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; with the proviso that when R1 and R4 are methyl, R2 and R5 cannot be methyl, a straight chain C1-C6 alkyl, a C7-C12 aralkyl, CH2O(C1-C3 alkyl), CH2OH, CH(OH)CH3, CH2CH2OH, CH(NH(CH2)nNH2)CH3, CH2CH2NH(CH2)nNH2 (where n=2, 3, 4, 6), vinyl, ethyl, CH2OR25 (where R25 is a hydrogen or a C1-C3 alkyl), CH(O-lower alkanoyl)CH3, CH(O-lower alkylene-OR26)CH3, or CH(OR26)CH3 (where R26═H, lower alkyl, a polyfunctional carbonyl compound excluding a hydrogen atom therefrom or a metal derivative of a polyfunctional carbonyl compound); and wherein
M is 2H or a diamagnetic or paramagnetic photoactive metal ion selected from Ga3+,
Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, and Mg2+.
2. A method of using the compound of claim 1 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging.
3. A method of using the compound of claim 1 comprising administering the compound to a patient and, after a period of time, irradiating targeted tissue of the patient with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
4. A method of using the compound of claim 1 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging then, after a second period of time, irradiating the targeted tissue with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
5. A method for the detection or treatment of tissue comprising administering to a patient a therapeutic amount of the compound of claim 1 locally, systemically, intramuscularly or interperitoneally and irradiating said compound with energy at a wavelength able to excite the molecule, such that a desired therapeutic effect is observed, whereby said tissue belongs to the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system, skin, pulmonary system, gastrointestinal system, reproductive system, immune system, cardiovascular system, urinary system, auditory or olfactory system.
6. The method of claim 2, wherein said method is for diagnosing disorders in a vessel wall, tissue adjoining the vessel wall, or material attached to the vessel wall of a coronary, carotid or peripheral vasculature.
7. The method of claim 6 wherein said vessel is an artery or a vein.
8. The method of claim 2 wherein the tissue is atherosclerosis, restenosis or graft disease.
9. The method of claim 3 wherein the tissue is atherosclerosis, restenosis or graft disease.
10. The method of claim 4 wherein the tissue is atherosclerosis, restenosis or graft disease.
11. The method of claim 5 wherein the tissue is atherosclerosis, restenosis or graft disease.
12. The method of claim 4 wherein the therapy is selected from ablation, reduction and stabilization of vessel wall plaque.
13. The method of claim 4 wherein said energy source is selected from light, ultrasound, magnetic force, and electromagnetic radiation in the UV/visible electromagnetic spectrum or near infrared.
14. The method of claim 4 wherein said administration of the compound is prior to, concomitant with, or subsequent to adjunctive interventions, diagnostics or therapies.
15. The method of claim 4 wherein said administration is a single bolus or plurality of doses administered to the patient.
16. A compound of formula IA:
Figure US20050226810A1-20051013-C00033
wherein R1-R7 can be the same or different and are selected from:
H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ether, polyether, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, CH(CH3)O-aryl; (CH2)nO-alkoxy, or (CH2)nO-alkyl, where n is an integer from 0 to 8;
C(X)2C(X)3, where X is a halogen;
CO2R8, where R8 is selected from a physiologically acceptable counter ion, a C1-C20 straight or branched chain alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-,. or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, ether or polyether, or a functional group of less than about 100,000 daltons;
(CH2)nOH, or (CH2)nOR9, where R9 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nCO2R10, (CHX)nCO2R10, or (CX2)nCO2R10, where X is a halogen and R10 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
CONH(R11), CO(R11), CON(R11)2, CON(R11)(R12), (CH2)nCONH(R11), (CH2)nCON(R11)2, (CH2)nCOR11, (CH2)nCON(R11)(R12), (CX2)nCONH(R11), (CX2)nCON(R11)2, (CX2)nCON(R11)(R12), (CX2)nCOR11, (CH2)nCONHNH(R11), (CX2)nCONHNH(R11), (CHX)nCONH(R11), (CHX)nCONHNH(R11), (CHX)nCON(R11)2, (CHX)nCON(R11)(R12), where X is a halogen and R11 and R12 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
S(R13), (CH2)nS(R13), (CH2)nNH(R13), (CH2)nNHNH(R13), (CH2) n NR13(CH2)nN(R13)(R14), or (CH2)nN(R13)(R14)(R15)+A, where R13, R14 and R15 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids (provided —NH(R13 is part of the amino acid), an amino acid ester, an amino acid amide, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R13, R14 and R15 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
(CH2)nOPO2OR16, (CH2)nPO(OR16)2, (CH2)nPO2R16, or (CH2)nPOR16 where R16 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nNHCOR17, (CH2)nNHNHCOR17, where R17 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
SO3R18, SO2NHR18, SO2N(R18)2, SO2NHNHR18 or SO2R18, where R18 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, and NHR18 can also be an amino acid residue, an amino acid salt, an amino acid ester residue, an amino acid amide residue, or a functional group of less than about 100,000 daltons;
Aryl or substituted aryl, which may bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons; and
R1-R2, and R3-R4 may also possess the atoms necessary to form ring systems, which themselves may possess heteroatoms that may bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons;
with the proviso that R1 and R4 are the same, R2 and R3 are the same, and that when R7 is H, R1-R4 cannot be methyl; and that at least one of the R1-R7 groups is linked via an organic group that has as part or all of its structure a group Q, which is an amine, a ester, a ether or an amide link, to a complexing agent of general formula 11a, 11b, 11c, 11d, 11e:
Figure US20050226810A1-20051013-C00034
wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; and wherein
M is 2H or a diamagnetic or paramagnetic photoactive metal ion selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, and Mg2+.
17. A method of using the compound of claim 16 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging.
18. A method of using the compound of claim 16 comprising administering the compound to a patient and, after a period of time, irradiating targeted tissue of the patient with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
19. A method of using the compound of claim 16 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging then, after a second period of time, irradiating the targeted tissue with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
20. A method for the detection or treatment of tissue comprising administering to a patient a therapeutic amount of the compound of claim 16 locally, systemically, intramuscularly or interperitoneally and irradiating said compound with energy at a wavelength able to excite the molecule, such that a desired therapeutic effect is observed, whereby said tissue belongs to the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system, skin, pulmonary system, gastrointestinal, reproductive system, immune system, cardiovascular system, urinary system, auditory or olfactory system.
21. The method of claim 17, wherein said method is for diagnosing disorders in a vessel wall, tissue adjoining the vessel wall, or material attached to the vessel wall of a coronary, carotid or peripheral vasculature.
22. The method of claim 21 wherein said vessel is an artery or a vein.
23. The method of claim 17 wherein the tissue is atherosclerosis, restenosis or graft disease.
24. The method of claim 18 wherein the tissue is atherosclerosis, restenosis or graft disease.
25. The method of claim 19 wherein the tissue is atherosclerosis, restenosis or graft disease.
26. The method of claim 20 wherein the tissue is atherosclerosis, restenosis or graft disease.
27. The method of claim 19 wherein the therapy is selected from ablation, reduction and stabilization of vessel wall plaque.
28. The method of claim 19 wherein said energy source is selected from light, ultrasound, magnetic force, and electromagnetic radiation in the UV/visible electromagnetic spectrum or near infrared.
29. The method of claim 19 wherein said administration of the compound is prior to, concomitant with, or subsequent to adjunctive interventions, diagnostics or therapies.
30. The method of claim 19 wherein said administration is a single bolus or plurality of doses administered to the patient.
31. A compound of formula IB:
Figure US20050226810A1-20051013-C00035
wherein R1 and R2 can be the same or different and are selected from H, NO2, CN, CHO, CO-alkyl, SO3H, SO3alkyl, SO3alkylether, SO3heteroalkyl, SO3Na, SO3K, SO2NHalkyl, SO2N(alkyl)2, SO2NHheteroalkyl, SO2N(heteroalkyl)2, SO2NHhaloalkyl, SO2N(haloalkyl)2, SO2NHhaloalkylether, SO2N(haloalkylether)2, SO2NHalkylether, SO2N(haloalkylether)2, CO-haloalkyl, haloalkyl, heteroalkyl, hydroxyhaloalkyl, haloalkyl ether, haloalkyl ester, a halogen, and a alkylcarbonyloxy group;
R3 and R4 can be the same or different and are selected from:
CO2R5, where R5 is selected from a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, ethers or polyethers, or a functional group of less than about 100,000 daltons;
(CH2)nOH, or (CH2)nOR6, where R6 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nCO2R7, (CHX)nCO2R7, or (CX2)nCO2R7, where X is a halogen and R7 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
CONH(R8), CO(R8), CON(R8)2, CON(R8)(R9), (CH2)nCONH(R8), (CH2)nCON(R8)2, (CH2)nCOR8, (CH2)nCON(R8)(R9), (CX2)nCONH(R8), (CX2)nCON(R8)2, (CX2)nCON(R8)(R9), (CX2)nCOR8, (CH2)nCONHNH(R8), (CX2)nCONHNH(R8), (CHX)nCONH(R8), (CHX)nCONHNH(R8), (CHX)nCON(R8)2, (CHX)nCON(R8)(R9), where X is a halogen and R8 and R9 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
S(R10), (CH2)nS(R10), (CH2)nNH(R10), (CH2)nNHNH(R10), (CH2) n NR10(CH2)nN(R10)(R11), or (CH2)nN(R10)(R11)(R12)+A, where R10, R11 and R12 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids (provided —NH(R10 is part of the amino acid), an amino acid ester, an amino acid amide, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R10, R11 and R12 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
(CH2)nOPO2OR13, (CH2)nPO(OR13)2, (CH2)nPO2R13, or (CH2)nPOR13 where R13 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nNHCOR14, (CH2)nNHNHCOR14, where R14 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
SO3R15, SO2NHR15, SO2N(R15)2, SO2NHNHR15 or SO2R15, where R15 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl or heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, and NHR15 can also be an amino acid residue, an amino acid salt, an amino acid ester residue, an amino acid amide residue, or a functional group of less than about 100,000 daltons;
Aryl or substituted aryl, which may bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons;
with the proviso that at least one of the R1-R4 groups is linked via an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link, to a complexing agent of general formula 11a, 11b, 11c, 11d, 11e:
Figure US20050226810A1-20051013-C00036
wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; and wherein
M is 2H or a diamagnetic or paramagnetic photoactive metal ion selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, and Mg2+.
32. A method of using the compound of claim 31 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging.
33. A method of using the compound of claim 31 comprising administering the compound to a patient and, after a period of time, irradiating targeted tissue of the patient with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
34. A method of using the compound of claim 31 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging then, after a second period of time, irradiating the targeted tissue with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
35. A method for the detection or treatment of tissue comprising administering to a patient a therapeutic amount of the compound of claim 31 locally, systemically, intramuscularly or interperitoneally and irradiating said compound with energy at a wavelength able to excite the molecule, such that a desired therapeutic effect is observed, whereby said tissue belongs to the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system, skin, pulmonary system, gastrointestinal, reproductive system, immune system, cardiovascular system, urinary system, auditory or olfactory system.
36. The method of claim 32, wherein said method is for diagnosing disorders in a vessel wall, tissue adjoining the vessel wall, or material attached to the vessel wall of a coronary, carotid or peripheral vasculature.
37. The method of claim 36 wherein said vessel is an artery or a vein.
38. The method of claim 32 wherein the tissue is atherosclerosis, restenosis or graft disease.
39. The method of claim 33 wherein the tissue is atherosclerosis, restenosis or graft disease.
40. The method of claim 34 wherein the tissue is atherosclerosis, restenosis or graft disease.
41. The method of claim 35 wherein the tissue is atherosclerosis, restenosis or graft disease.
42. The method of claim 34 wherein the therapy is selected from ablation, reduction and stabilization of vessel wall plaque.
43. The method of claim 34 wherein said energy source is selected from light, ultrasound, magnetic force, and electromagnetic radiation in the UV/visible electromagnetic spectrum or near infrared.
44. The method of claim 34 wherein said administration of the compound is prior to, concomitant with, or subsequent to adjunctive interventions, diagnostics or therapies.
45. The method of claim 34 wherein said administration is a single bolus or plurality of doses administered to the patient.
46. (canceled)
47. (canceled)
48. (canceled)
49. (canceled)
50. (canceled)
51. (canceled)
52. (canceled)
53. (canceled)
54. (canceled)
55. (canceled)
56. (canceled)
57. (canceled)
58. (canceled)
59. (canceled)
60. (canceled)
61. (canceled)
62. (canceled)
63. (canceled)
64. (canceled)
65. (canceled)
66. (canceled)
67. (canceled)
68. (canceled)
69. (canceled)
70. (canceled)
71. (canceled)
72. (canceled)
73. (canceled)
74. (canceled)
75. (canceled)
76. A compound of formula III:
Figure US20050226810A1-20051013-C00037
wherein R1-R10 can be the same or different and are selected from:
H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ether, polyether, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group having a molecular weight of less than about 100,000 daltons; CH═CHCH2N+(CH3)3A, CH═N(alkyl)2A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, or CH(CH3)O-aryl;
(CH2)nO-alkoxy, or (CH2)nO-alkyl, where n is an integer from 0 to 8;
C(X)2C(X)3, where X is a halogen;
CO2R11, where R11 is selected from a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
(CH2)nOH, or (CH2)nOR12, where R12 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nCO2R13, (CHX)nCO2R13, or (CX2)nCO2R13, where X is a halogen and R13 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
CONH(R14), CONHNH(R14), CO(R14), CON(R14)2, CON(R14)(R15), (CH2)nCONH(R14), (CH2)nCONHNH(R14), (CH2)nCON(R14)2, (CH2)nCOR14, (CH2)nCON(R14)(R15), (CX2)nCONH(R14), (CX2)nCON(R14)2, (CX2)nCON(R14)(R15), (CX2)nCOR14, (CH2)nCONHNH(R14), (CX2)nCONHNH(R14), (CHX)nCONH(R14), (CHX)nCONHNH(R14), (CHX)nCON(R14)2, (CHX)nCON(R14)(R15), where X is a halogen and R14 and R15 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
S(R16), (CH2)nS(R16), (CH2)nNH(R16), (CH2)nNHNH(R16), (CH2)nN(R16)2 (C2)nN(R16)(R17), or (CH2)nN(R16)(R17)(R18)+A, where R16, R17 and R18 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, or an amino acid amide provided —NHR16 is part of the amino acid, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R16, R17 and R18 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
(CH2)nOPO2OR19, (CH2)nPO(OR19)2, (CH2)nPO2R19, or (CH2)nPOR19 where R19 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nNHCOR20, (CH2)nNHNHCOR20, where R20 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
SO3R21, SO2NHR21, SO2N(R21)2, SO2NHNHR21 or SO2R21, where R21 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons; and NHR21 can be an amino acid residue, an amino acid salt, an amino acid ester residue, or an amino acid amide residue;
Aryl or substituted aryl, which may bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons; and
R1-R2, R3-R4, R6-R7; R8-R9, R4-R5, R5-R6, R9-R10 and R10-R1 may also possess the atoms necessary to form ring systems, which themselves may possess heteroatoms that may neutral or bear one or more functional groups of molecular weight equal to or less than about 100,000 daltons;
with the proviso that at least one of the R1-R10 groups is linked via an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link, to a complexing agent of general formula 11a, 11b, 11c, 11d, 11e:
Figure US20050226810A1-20051013-C00038
wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; and wherein
M is 2H or a diamagnetic or paramagnetic photoactive metal ion selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, and Mg2+.
77. A method of using the compound of claim 76 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging.
78. A method of using the compound of claim 76 comprising administering the compound to a patient and, after a period of time, irradiating targeted tissue of the patient with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
79. A method of using the compound of claim 76 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging then, after a second period of time, irradiating the targeted tissue with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
80. A method for the detection or treatment of tissue comprising administering to a patient a therapeutic amount of the compound of claim 76 locally, systemically, intramuscularly or interperitoneally and irradiating said compound with energy at a wavelength able to excite the molecule, such that a desired therapeutic effect is observed, whereby said tissue belongs to the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system, skin, pulmonary system, gastrointestinal, reproductive system, immune system, cardiovascular system, urinary system, auditory or olfactory system.
81. The method of claim 77, wherein said method is for diagnosing disorders in a vessel wall, tissue adjoining the vessel wall, or material attached to the vessel wall of a coronary, carotid or peripheral vasculature.
82. The method of claim 81 wherein said vessel is an artery or a vein.
83. The method of claim 77 wherein the tissue is atherosclerosis, restenosis or graft disease.
84. The method of claim 78 wherein the tissue is atherosclerosis, restenosis or graft disease.
85. The method of claim 79 wherein the tissue is atherosclerosis, restenosis or graft disease.
86. The method of claim 80 wherein the tissue is atherosclerosis, restenosis or graft disease.
87. The method of claim 79 wherein the therapy is selected from ablation, reduction and stabilization of vessel wall plaque.
88. The method of claim 79 wherein said energy source is selected from light, ultrasound, magnetic force, and electromagnetic radiation in the UV/visible electromagnetic spectrum or near infrared.
89. The method of claim 79 wherein said administration of the compound is prior to, concomitant with, or subsequent to adjunctive interventions, diagnostics or therapies.
90. The method of claim 79 wherein said administration is a single bolus or plurality of doses administered to the patient.
91. A compound of formula IIIA:
Figure US20050226810A1-20051013-C00039
wherein R1-R4 can be the same or different and are selected from a functional group of less than about 100,000 daltons;
CO2R5, where R5 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
(CH2)nOH, or (CH2)nOR6, where R6 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, heterocycle, aryl, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nCO2R7, (CHX)nCO2R7, or (CX2)nCO2R7, where X is a halogen and R7 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, heterocycle, aryl, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
CONH(R8), (CONHNH(R8), CO(R8), CON(R8)2, CON(R8)(R9), (CH2)nCONH(R8), (CH2)nCONHNH(R8), (CH2)nCON(R8)2, (CH2)nCOR8, (CH2)nCON(R8)(R9), (CX2)nCONH(R8), (CX2)nCON(R8)2, (CX2)nCON(R8)(R9), (CX2)nCOR8, (CH2)nCONHNH(R8), (CX2)nCONHNH(R8), (CHX)nCONH(R8), (CHX)nCONHNH(R8), (CHX)nCON(R8)2, or (CHX)nCON(R8)(R9), where X is a halogen and R8 and R9 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
S(R10), (CH2)nS(R10), (CH2)nNH(R10), (CH2)nNHNH(R10), (CH2)nN(R10)2, (CH2)nN(R10)(R11), or (CH2)nN(R10)(R11)(R12)+A, where R10, R11 and R12 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, or an amino acid amide provided —NHR10 is part of the amino acid, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R10, R11 and R12 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
(CH2)nOPO2OR13, (CH2)nPO(OR13)2, (CH2)nPO2R13, or (CH2)nPOR13 where R13 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nNHCOR14, or (CH2)nNHNHCOR14, where R14 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
SO3R15, SO2NHR15, SO2N(R15)2, SO2NHNHR15 or SO2R15, where R15 is selected from H, a physiologically acceptable counter ion, a, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle; NHR15 can also be an amino acid residue, an amino acid salt, an amino acid ester residue, or an amino acid amide residue; a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-,
or polyetheraryl residue, or a functional group of less than about 100,000 daltons;
Aryl or substituted aryl, which may bear one or more substituents with a molecular weight of less than or equal to about 100,000 daltons;
with the proviso that at least one of the R1-R4 groups is linked via an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link, to a complexing agent of general formula 11a, 11b, 11c, 11d, 11e:
Figure US20050226810A1-20051013-C00040
wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; and wherein
M is 2H or a diamagnetic or paramagnetic photoactive metal ion selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, and Mg2+.
92. A method of using the compound of claim 91 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging.
93. A method of using the compound of claim 91 comprising administering the compound to a patient and, after a period of time, irradiating targeted tissue of the patient with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
94. A method of using the compound of claim 91 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging then, after a second period of time, irradiating the targeted tissue with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
95. A method for the detection or treatment of tissue comprising administering to a patient a therapeutic amount of the compound of claim 91 locally, systemically, intramuscularly or interperitoneally and irradiating said compound with energy at a wavelength able to excite the molecule, such that a desired therapeutic effect is observed, whereby said tissue belongs to the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system, skin, pulmonary system, gastrointestinal, reproductive system, immune system, cardiovascular system, urinary system, auditory or olfactory system.
96. The method of claim 92, wherein said method is for diagnosing disorders in a vessel wall, tissue adjoining the vessel wall, or material attached to the vessel wall of a coronary, carotid or peripheral vasculature.
97. The method of claim 96 wherein said vessel is an artery or a vein.
98. The method of claim 92 wherein the tissue is atherosclerosis, restenosis or graft disease.
99. The method of claim 93 wherein the tissue is atherosclerosis, restenosis or graft disease.
100. The method of claim 94 wherein the tissue is atherosclerosis, restenosis or graft disease.
101. The method of claim 95 wherein the tissue is atherosclerosis, restenosis or graft disease.
102. The method of claim 94 wherein the therapy is selected from ablation, reduction and stabilization of vessel wall plaque.
103. The method of claim 94 wherein said energy source is selected from light, ultrasound, magnetic force, and electromagnetic radiation in the UV/visible electromagnetic spectrum or near infrared.
104. The method of claim 94 wherein said administration of the compound is prior to, concomitant with, or subsequent to adjunctive interventions, diagnostics or therapies.
105. The method of claim 94 wherein said administration is a single bolus or plurality of doses administered to the patient.
106. A compound of formula IV:
Figure US20050226810A1-20051013-C00041
wherein R1-R8 can be the same or different and are selected from:
H, halide, substituted or unsubstituted alkyl, heteroalkyl, haloalkyl, heterohaloalkyl, cyclic alkyl, aryl, substituted aryl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amide, ester, ether, polyether, alkoxy group, aryloxy group, haloalkoxy group, amino group, alkylcarbonyloxy group, alkoxycarbonyl group, aryloxycarbonyl group, azo group, arylcarbonyloxy group, alkoxycarbonyloxy group, aryloxycarbonyloxy group, sulfinyl group, sulfonyl group, silil group, carbamoyl group, heterocyclic group, nitro group, nitroso group, formyloxy group, isocyano group, cyanate group, isocyanate group, thiocyanate group, isothiocyanate group, N(alkyl)2, N(aryl)2, CH═CH(aryl), CH═CHCH2N(CH3)2, or a functional group of molecular weight of less than about 100,000 daltons; CH═CHCH2N(CH3)3 +A, CH═N(alkyl)2 +A, or N(alkyl)3 +A, where A is a charge balancing ion; CN, OH, CHO, COCH3, CO(alkyl), CO2H, CO2Na, CO2K, CH(CH3)OH, CH(CH3)O-alkyl, CH(CH3)O-alkoxy, or CH(CH3)O-aryl;
(CH2)nO-alkoxy, or (CH2)nO-alkyl; where n is an integer from 0 to 8;
C(X)2C(X)3, where X is a halogen;
CO2R9, where R9 is selected from a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons;
(CH2)nOH, or (CH2)nOR10, where R10 is selected from alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a protecting group, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nCO2R11, (CHX)nCO2R11, (CX2)nCO2R11 where X is a halogen and R11 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heterocycle, heteroaryl, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 1 and 4;
CONH(R12), CONHNH(R12)CO(R12), CON(R12)2, CON(R12)(R13), (CH2)nCONH(R12), (CH2)nCONHNH(R12), (CH2)nCON(R12)2, (CH2)nCOR12 (CH2)nCON(R12)(R13), (CX2)nCONH(R12), (CX2)nCON(R12)2, (CX2)nCON(R12)(R13), (CX2)nCOR12, (CH2)nCONHNH(R12), (CX2)nCONHNH(R12), (CHX)nCONH(R12), (CHX)nCONHNH(R12), (CHX)nCON(R12)2, (CHX)nCON(R12)(R13), where X is a halogen and R12 and R13 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, haloheteroalkyl, heteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, an amino acid, an amino acid salt, an amino acid ester, an amino acid amide, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
S(R14), (CH2)nS(R14), (CH2)nNH(R14), (CH2)nNHNH(R14), (CH2)nN(R14)2, (CH2)nN(R14)(R15), or (CH2)nN(R14)(R15)(R16)+A, where R14, R15 and R16 can be the same or different and are selected from H, NH2, straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, an amino acid ester, or an amino acid amide provided —NH(R14) is part of the amino acid, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, a functional group of less than about 100,000 daltons, or where R14, R15 and R16 together possess the atoms necessary to constitute an aromatic ring system, n is an integer between 0 and 4, and A is a physiologically acceptable counter ion;
(CH2)nOPO2OR17, (CH2)nPO(OR17)2, (CH2)nPO2R17, or (CH2)nPOR17 where R17 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, amino acids, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
(CH2)nNHCOR18, or (CH2)nNHNHCOR18, where R18 is selected from a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, or a functional group of less than about 100,000 daltons, and n is an integer between 0 and 4;
SO3R19, SO2NHR19, SO2N(R19)2, SO2NHNHR19 or SO2R19, where R19 is selected from H, a physiologically acceptable counter ion, a straight or branched chain C1-C20 alkyl, haloalkyl, heteroalkyl, haloheteroalkyl, aryl, heteroaryl, heterocycle, a mono-, di-, or polyhydroxyalkyl residue, a mono-, di-, or polyhydroxyaryl residue, a mono-, di-, or polyetheralkyl residue, a mono-, di-, or polyetheraryl residue, or a functional group of less than about 100,000 daltons, and NHR19 can also be an amino acid residue, an amino acid salt, an amino acid ester residue, or an amino acid amide residue; and
Aryl or substituted aryl, which may bear one or more substituents selected from hydroxy groups, alkyl groups, carboxyl groups and its esters and amides and sulfonic acid groups and their esters and amides, and substitiuents with a molecular weight of less than or equal to about 100,000 daltons;
with the proviso that at least one of the R1-R12 groups is linked via an organic group that has as part or all of its structure a group Q, which is an amine, an ester, an ether or an amide link, to a complexing agent of general formula 11a, 11b, 11c, 11d, 11e:
Figure US20050226810A1-20051013-C00042
wherein R24 is selected from a hydrogen, a straight or branched chain C1-C7 alkyl group, a phenyl or benzyl group; L1, L2, L3, L4, independently of one another, are selected from a hydrogen atom or a metal ion equivalent of an element of the atomic numbers 20-32, 37-39, 42-51, or 57-83, which may be radioactive, provided that at least two of L1, L2, L3 and L4 are metal ion equivalents, that other anions are present to compensate for optionally present charges on the porphyrin, and free carboxylic acid groups that are not required for complexing are optionally present as salts with physiologically compatible inorganic cations, or organic cations, or as esters or amides; and
A, B, C, and D can be the same or different and are selected from N, CH, and CR20 where R20 is selected from a halogen, aryl, subsitituted aryl, heteroaryl, alkyl, haloalkyl, heterohaloalkyl, heterocycle, hydroxyalky, hydroxyhaloalkyl, or a functional group of molecular weight of less than about 100,000 daltons; and wherein
M is selected from 2H or a diamagnetic or paramagnetic photoactive metal ion selected from Ga3+, Pt2+, Pd2+, Sn4+, In3+, Ge4+, Si4+, Al3+, Zn2+, and Mg2+.
107. A method of using the compound of claim 106 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging.
108. A method of using the compound of claim 106 comprising administering the compound to a patient and, after a period of time, irradiating targeted tissue of the patient with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
109. A method of using the compound of claim 106 comprising administering the compound to a patient and, after a period of time, imaging targeted tissue of the patient through MRI or radio-diagnostic imaging then, after a second period of time, irradiating the targeted tissue with an energy source that excites the compound thereby producing a desired therapeutic response in the target tissue.
110. A method for the detection or treatment of tissue comprising administering to a patient a therapeutic amount of the compound of claim 106 locally, systemically, intramuscularly or interperitoneally and irradiating said compound with energy at a wavelength able to excite the molecule, such that a desired therapeutic effect is observed, whereby said tissue belongs to the hematological system, lymphatic reticuloendothelial system, nervous system, endocrine and exocrine system, skeletomuscular system, skin, pulmonary system, gastrointestinal, reproductive system, immune system, cardiovascular system, urinary system, auditory or olfactory system.
111. The method of claim 107, wherein said method is for diagnosing disorders in a vessel wall, tissue adjoining the vessel wall, or material attached to the vessel wall of a coronary, carotid or peripheral vasculature.
112. The method of claim 111 wherein said vessel is an artery or a vein.
113. The method of claim 107 wherein the tissue is atherosclerosis, restenosis or graft disease.
114. The method of claim 108 wherein the tissue is atherosclerosis, restenosis or graft disease.
115. The method of claim 109 wherein the tissue is atherosclerosis, restenosis or graft disease.
116. The method of claim 1 10 wherein the tissue is atherosclerosis, restenosis or graft disease.
117. The method of claim 109 wherein the therapy is selected from ablation, reduction and stabilization of vessel wall plaque.
118. The method of claim 109 wherein said energy source is selected from light, ultrasound, magnetic force, and electromagnetic radiation in the UV/visible electromagnetic spectrum or near infrared.
119. The method of claim 109 wherein said administration of the compound is prior to, concomitant with, or subsequent to adjunctive interventions, diagnostics or therapies.
120. The method of claim 109 wherein said administration is a single bolus or plurality of doses administered to the patient.
US11/059,557 2001-05-31 2005-02-17 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis Abandoned US20050226810A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/059,557 US20050226810A1 (en) 2001-05-31 2005-02-17 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US29534301P 2001-05-31 2001-05-31
US10/159,580 US6906050B2 (en) 2001-05-31 2002-05-31 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis
US11/059,557 US20050226810A1 (en) 2001-05-31 2005-02-17 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/159,580 Division US6906050B2 (en) 2001-05-31 2002-05-31 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis

Publications (1)

Publication Number Publication Date
US20050226810A1 true US20050226810A1 (en) 2005-10-13

Family

ID=23137288

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/159,580 Expired - Fee Related US6906050B2 (en) 2001-05-31 2002-05-31 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis
US11/059,557 Abandoned US20050226810A1 (en) 2001-05-31 2005-02-17 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/159,580 Expired - Fee Related US6906050B2 (en) 2001-05-31 2002-05-31 Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis

Country Status (4)

Country Link
US (2) US6906050B2 (en)
EP (1) EP1401430A4 (en)
CA (1) CA2448570A1 (en)
WO (1) WO2002096417A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090256141A1 (en) * 2007-10-12 2009-10-15 University Of Southern California Organic photosensitive optoelectronic devices containing tetra-azaporphyrins
US11896852B2 (en) 2020-12-21 2024-02-13 Xerox Corporation Closed-loop non-invasive transcranial stimulation and neural activity recording system and method

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE60029944T2 (en) * 1999-04-30 2007-04-05 Cellgate, Inc., Redwood City POLYAMINE AND ITS THERAPEUTIC USE
US7097826B2 (en) * 1999-12-23 2006-08-29 Health Research, Inc. Chlorin and bacteriochlorin-based difunctional aminophenyl DTPA and N2S2 conjugates for MR contrast media and radiopharmaceuticals
CA2448562A1 (en) * 2001-05-31 2002-12-05 Miravant Pharmaceuticals, Inc. Metallotetrapyrrolic photosensitizing agents for use in photodynamic therapy
DE60219627T2 (en) * 2001-06-04 2008-02-07 The General Hospital Corp., Boston IDENTIFICATION AND THERAPY OF SENSITIVE PLAQUE WITH PHOTODYNAMIC COMPOUNDS
US7375216B2 (en) * 2002-06-04 2008-05-20 Infacare Pharmaceutical Corporation Preparation of metal mesoporphyrin compounds
ES2197818B1 (en) * 2002-06-11 2005-02-01 Institut Quimic De Sarria Cets 2,7,12,17 ALQUENIL, ARIL AND HETEROARIL DERIVED FROM 3,6,13,16-TETRAAZAPORFICENO, AND PROCEDURE, INTERMEDIATE COMPOUND AND CORRESPONDING UTILIZATIONS.
WO2004002991A1 (en) * 2002-06-26 2004-01-08 Cellgate, Inc. Porphyrin-polyamine conjugates for cancer therapy
US20040192665A1 (en) * 2002-08-02 2004-09-30 Slil Biomedical Corporation Conjugates of porphyrin compounds with chemotherapeutic agents
US20040097481A1 (en) * 2002-11-20 2004-05-20 Benjamin Levinson Water-soluble mesoporphyrin compounds and methods of preparation
JP4614281B2 (en) * 2002-12-16 2011-01-19 ジーイー・ヘルスケア・アクスイェ・セルスカプ Magnetic resonance imaging method and compound used in the method
GB2397067B (en) * 2002-12-23 2005-05-11 Destiny Pharma Ltd Porphin & azaporphin derivatives with at least one cationic-nitrogen-containing meso-substituent for use in photodynamic therapy & in vitro sterilisation
US20050209193A1 (en) * 2003-12-05 2005-09-22 Keller Gregory S Method for enhanced photodynamic therapy
FR2867473B1 (en) * 2004-03-12 2006-06-23 Guerbet Sa PORPHYRINE COMPOUND AND HIGH FIELD USE IN MRI
EP1765163A1 (en) * 2004-05-20 2007-03-28 Pharmacyclics, Inc. A method of enhancing visualization of atherosclerotic plaque
GB2415372A (en) 2004-06-23 2005-12-28 Destiny Pharma Ltd Non photodynamical or sonodynamical antimicrobial use of porphyrins and azaporphyrins containing at least one cationic-nitrogen-containing substituent
US20060222668A1 (en) * 2005-04-01 2006-10-05 Wellspring Pharmaceutical Corporation Stannsoporfin compositions, drug products and methods of manufacture
ES2275416B1 (en) * 2005-07-15 2008-06-01 Institut Quimic De Sarria Cets HALOGENATED DERIVATIVES OF 4H, 5H-TIENO (3,2-B: 4,5-B ') DIPIRROLES AND PROCEDURE AND CORRESPONDING UTILIZATIONS.
US20080085293A1 (en) * 2006-08-22 2008-04-10 Jenchen Yang Drug eluting stent and therapeutic methods using c-Jun N-terminal kinase inhibitor
EP2076273A2 (en) * 2006-10-04 2009-07-08 InfaCare Pharmaceutical Corporation Treatment of infant hyperbilirubinemia using low dosages of stannsoporfin
EP2992886A1 (en) 2006-10-04 2016-03-09 InfaCare Pharmaceutical Corporation High-purity large-scale preparation of stannsoporfin
US8287839B2 (en) * 2006-12-04 2012-10-16 Brookhaven Science Associates, Llc Carboranylporphyrins and uses thereof
US8444953B2 (en) * 2007-03-22 2013-05-21 Brookhaven Science Associates, Llc Symmetric and asymmetric halogen-containing metallocarboranylporphyrins and uses thereof
US20080279781A1 (en) * 2007-05-10 2008-11-13 Brookhaven Science Associates, Llc Glycosylated Carboranylporphyrins and Uses Thereof
US7952719B2 (en) * 2007-06-08 2011-05-31 Prescient Medical, Inc. Optical catheter configurations combining raman spectroscopy with optical fiber-based low coherence reflectometry
US20100113906A1 (en) * 2008-11-06 2010-05-06 Prescient Medical, Inc. Hybrid basket catheters
PT2691398T (en) 2011-03-30 2017-01-02 Infacare Pharmaceutical Corp Methods for synthesizing metal mesoporphyrins
CN102363619A (en) * 2011-11-18 2012-02-29 中国医学科学院生物医学工程研究所 Water soluble protoporphyrin compound and preparation method and use thereof
EP2796101B1 (en) * 2013-04-23 2016-04-20 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Kit for producing a cross-linked gel for encapsulating kidney stones and/or kidney stone fragments
PL2796100T3 (en) 2013-04-23 2016-08-31 Fraunhofer Ges Forschung Gelling system for the removal of kidney stone fragments
US20160235866A1 (en) * 2013-10-04 2016-08-18 The Governing Council Of The University Of Toronto Mri contrast agents for cell labeling
WO2019060770A1 (en) * 2017-09-21 2019-03-28 Health Research, Inc. Tetrapyrrolic conjugates and uses thereof for imaging
CZ307580B6 (en) * 2017-10-16 2018-12-19 Fyziologický ústav AV ČR, v. v. i. A liposomal dosage form with light-converting nanoparticles, its preparation and use
CN116003820B (en) * 2023-02-15 2023-12-19 山东大学 Palladium porphyrin and triptycene-based porous organic polymer, and preparation method and application thereof

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4647447A (en) * 1981-07-24 1987-03-03 Schering Aktiengesellschaft Diagnostic media
US4882234A (en) * 1986-11-12 1989-11-21 Healux, Inc. Storage-stable porphin compositions and a method for their manufacture
US4885363A (en) * 1987-04-24 1989-12-05 E. R. Squibb & Sons, Inc. 1-substituted-1,4,7-triscarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US4996312A (en) * 1985-10-23 1991-02-26 Nihon Medi-Physics Co., Ltd. Porphyrin derivatives, and their production and use
US5053503A (en) * 1989-02-17 1991-10-01 Centocor Chelating agents
US5275801A (en) * 1988-08-13 1994-01-04 Schering Aktiengesellschaft 13,17-propionic acid and propionic acid derivative substituted porphyrin complex compounds, process for their production and pharmaceutical agents containing them
US5277895A (en) * 1990-11-08 1994-01-11 Schering Aktiengesellschaft Mono-n-substituted 1,4,7,10-tetraazacyclododecane derivatives, process for their production and pharmaceutical agents containing these derivatives
US5284647A (en) * 1988-03-18 1994-02-08 Schering Aktiengesellschaft Mesotetraphenylporphyrin complex compounds, process for their production and pharmaceutical agents containing them
US5364614A (en) * 1989-11-21 1994-11-15 Schering Aktiengesellschaft Cascade polymer bound chelating compounds, their chelates and conjugates, processes for their production, and pharmaceutical agents containing them
US5633275A (en) * 1995-09-06 1997-05-27 Meiji Seika Kaisha, Ltd. Photochemotherapeutical obstruction of newly-formed blood vessels
US5654423A (en) * 1990-11-21 1997-08-05 Regents Of The University Of California Boronated metalloporphyrine and therapeutic methods
US5675001A (en) * 1995-03-14 1997-10-07 Hoffman/Barrett, L.L.C. Heteroatom-functionalized porphyrazines and multimetallic complexes and polymers derived therefrom
US5676923A (en) * 1995-02-21 1997-10-14 Schering Aktiengesellschaft Substituted DTPA monoamides of the central carboxylic acid group and their metal complexes
US5703230A (en) * 1994-12-02 1997-12-30 University Of British Columbia Meso-monoiodo-substituted tetramacrocyclic compounds and methods for making and using the same
US5705622A (en) * 1988-06-08 1998-01-06 London Diagnostics, Inc. Sensitizer conjugates containing porphyrins
US5730956A (en) * 1995-02-21 1998-03-24 Schering Aktiengesellschaft DTPA di-alkyl monoamides for x-ray and MRI
US5849259A (en) * 1992-09-28 1998-12-15 Institut Fur Diagnostikforschung Gmbh 3-,8-substituted deuteroporphyrin derivatives, pharmaceutical agents containing the latter and process for their production
US6136841A (en) * 1998-06-02 2000-10-24 Schering Aktiengesellschaft 3-, 8-substituted deuteroporphyrin derivatives, pharmaceutical agents that contain the latter, process for their production and their use in photodynamic therapy and MRI diagnosis
US6251367B1 (en) * 1998-07-24 2001-06-26 Schering Aktiengesellschaft Paramagnetic 3-,8-substituted deuteroporphyrin derivatives, pharmaceutical agents that contain the latter, process for their production, and their use for MR imaging of necrosis and infarction

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0232751B1 (en) 1986-01-23 1991-09-11 E.R. Squibb &amp; Sons, Inc. 1-substituted-4,7,10-triscarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
AU655942B2 (en) 1990-10-05 1995-01-19 Queen's University At Kingston Porphyrin derivatives
DE4425857A1 (en) 1994-07-07 1996-01-11 Schering Ag Cascade polymer complexes, processes for their preparation and pharmaceutical compositions containing them
IL114235A0 (en) 1994-07-14 1995-10-31 Schering Ag Oligonucleotide conjugates and processes for noninvasive diagnosis utilizing the same
GB9524028D0 (en) 1995-11-23 1996-01-24 Secr Defence Substituted porphyrins
EP0811626B1 (en) 1996-06-04 2002-08-14 Roche Diagnostics GmbH Use of metallo-porphyrin conjugates for the detection of biological substances
DE19825512A1 (en) 1998-06-02 1999-12-09 Schering Ag 3-, 8-substituted deuteroporphyrin derivatives, pharmaceutical compositions containing them, processes for their preparation and their use in photodynamic therapy and MRI diagnostics
DE19835082A1 (en) 1998-07-24 2000-02-03 Schering Ag Paramagnetic 3-, 8-substituted deuteroporphyrin derivatives, pharmaceutical compositions containing them, processes for their preparation and their use for necrosis and infarct MR imaging

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4647447A (en) * 1981-07-24 1987-03-03 Schering Aktiengesellschaft Diagnostic media
US4996312A (en) * 1985-10-23 1991-02-26 Nihon Medi-Physics Co., Ltd. Porphyrin derivatives, and their production and use
US4882234A (en) * 1986-11-12 1989-11-21 Healux, Inc. Storage-stable porphin compositions and a method for their manufacture
US4885363A (en) * 1987-04-24 1989-12-05 E. R. Squibb & Sons, Inc. 1-substituted-1,4,7-triscarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US5284647A (en) * 1988-03-18 1994-02-08 Schering Aktiengesellschaft Mesotetraphenylporphyrin complex compounds, process for their production and pharmaceutical agents containing them
US5705622A (en) * 1988-06-08 1998-01-06 London Diagnostics, Inc. Sensitizer conjugates containing porphyrins
US5275801A (en) * 1988-08-13 1994-01-04 Schering Aktiengesellschaft 13,17-propionic acid and propionic acid derivative substituted porphyrin complex compounds, process for their production and pharmaceutical agents containing them
US5053503A (en) * 1989-02-17 1991-10-01 Centocor Chelating agents
US5364614A (en) * 1989-11-21 1994-11-15 Schering Aktiengesellschaft Cascade polymer bound chelating compounds, their chelates and conjugates, processes for their production, and pharmaceutical agents containing them
US5277895A (en) * 1990-11-08 1994-01-11 Schering Aktiengesellschaft Mono-n-substituted 1,4,7,10-tetraazacyclododecane derivatives, process for their production and pharmaceutical agents containing these derivatives
US5654423A (en) * 1990-11-21 1997-08-05 Regents Of The University Of California Boronated metalloporphyrine and therapeutic methods
US5849259A (en) * 1992-09-28 1998-12-15 Institut Fur Diagnostikforschung Gmbh 3-,8-substituted deuteroporphyrin derivatives, pharmaceutical agents containing the latter and process for their production
US5703230A (en) * 1994-12-02 1997-12-30 University Of British Columbia Meso-monoiodo-substituted tetramacrocyclic compounds and methods for making and using the same
US5676923A (en) * 1995-02-21 1997-10-14 Schering Aktiengesellschaft Substituted DTPA monoamides of the central carboxylic acid group and their metal complexes
US5730956A (en) * 1995-02-21 1998-03-24 Schering Aktiengesellschaft DTPA di-alkyl monoamides for x-ray and MRI
US5675001A (en) * 1995-03-14 1997-10-07 Hoffman/Barrett, L.L.C. Heteroatom-functionalized porphyrazines and multimetallic complexes and polymers derived therefrom
US5633275A (en) * 1995-09-06 1997-05-27 Meiji Seika Kaisha, Ltd. Photochemotherapeutical obstruction of newly-formed blood vessels
US6136841A (en) * 1998-06-02 2000-10-24 Schering Aktiengesellschaft 3-, 8-substituted deuteroporphyrin derivatives, pharmaceutical agents that contain the latter, process for their production and their use in photodynamic therapy and MRI diagnosis
US6251367B1 (en) * 1998-07-24 2001-06-26 Schering Aktiengesellschaft Paramagnetic 3-,8-substituted deuteroporphyrin derivatives, pharmaceutical agents that contain the latter, process for their production, and their use for MR imaging of necrosis and infarction

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090256141A1 (en) * 2007-10-12 2009-10-15 University Of Southern California Organic photosensitive optoelectronic devices containing tetra-azaporphyrins
US8158972B2 (en) * 2007-10-12 2012-04-17 The University Of Southern California Organic photosensitive optoelectronic devices containing tetra-azaporphyrins
US11896852B2 (en) 2020-12-21 2024-02-13 Xerox Corporation Closed-loop non-invasive transcranial stimulation and neural activity recording system and method

Also Published As

Publication number Publication date
EP1401430A1 (en) 2004-03-31
WO2002096417A1 (en) 2002-12-05
US20030100752A1 (en) 2003-05-29
CA2448570A1 (en) 2002-12-05
EP1401430A4 (en) 2005-10-19
US6906050B2 (en) 2005-06-14

Similar Documents

Publication Publication Date Title
US6906050B2 (en) Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis
US6827926B2 (en) Metallotetrapyrrolic photosensitizing agents for use in photodynamic therapy
JP3530529B2 (en) Metal complexes of water-soluble texaphyrins
AU2002344234A1 (en) Metallotetrapyrrolic photosensitizing agents for use in photodynamic therapy
US6114321A (en) Porphyrin derivatives, pharmaceutical agents that contain the latter, and their use in photodynamic therapy and MRI diagnosis
JPH10500659A (en) Texaphyrin metal complexes with improved functionality
JP5372371B2 (en) Cationic bacteriochlorophyll derivatives and uses thereof
US8133473B2 (en) Chlorin and bacteriochlorin-based difunctional aminophenyl DTPA and N2S2 conjugates for MR contrast media and radiopharmaceuticals
JPH08504399A (en) 3-, 8-Substituted deuteroporphyrin derivative, drug containing the same, and process for producing the same
US6136841A (en) 3-, 8-substituted deuteroporphyrin derivatives, pharmaceutical agents that contain the latter, process for their production and their use in photodynamic therapy and MRI diagnosis
JP4733126B2 (en) Addendum of fluorescent dye and tumor affinity tetrapyrrole
US20050020559A1 (en) Chlorin photosensitizing agents for use in photodynamic therapy
US20080275232A1 (en) Chlorins possessing fused ring systems useful as photoselective compounds for photodynamic therapy
AU2002314857A1 (en) Substituted porphyrin and azaporphyrin derivatives and their use in photodynamic therapy, radioimaging and MRI diagnosis
AU2008200847A1 (en) Metallotetrapyrrolic photosensitizing agents for use in photodynamic therapy
Boateng Design, synthesis and biological evaluation of folate-targeted photodynamic therapy agents

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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