WO2014022742A1 - Paa nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded - Google Patents

Paa nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded Download PDF

Info

Publication number
WO2014022742A1
WO2014022742A1 PCT/US2013/053350 US2013053350W WO2014022742A1 WO 2014022742 A1 WO2014022742 A1 WO 2014022742A1 US 2013053350 W US2013053350 W US 2013053350W WO 2014022742 A1 WO2014022742 A1 WO 2014022742A1
Authority
WO
WIPO (PCT)
Prior art keywords
paa
photosensitizer
alkyl
substituted
imaging
Prior art date
Application number
PCT/US2013/053350
Other languages
French (fr)
Inventor
Ravindra K. Pandey
Raoul Kopelman
Anurag Gupta
Munawwar Sajjad
Original Assignee
Health Research, Inc.
The Research Foundation Of State University Of New York
Regents Of The University Of Michigan
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
Priority claimed from US13/566,444 external-priority patent/US8906343B2/en
Application filed by Health Research, Inc., The Research Foundation Of State University Of New York, Regents Of The University Of Michigan filed Critical Health Research, Inc.
Publication of WO2014022742A1 publication Critical patent/WO2014022742A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09BORGANIC DYES OR CLOSELY-RELATED COMPOUNDS FOR PRODUCING DYES, e.g. PIGMENTS; MORDANTS; LAKES
    • C09B47/00Porphines; Azaporphines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • Nanoscience is being developed in conjunction with advanced medical science for further precision in diagnosis and treatment.
  • Nanoplatforms and nanovectors that deliver a therapeutic or imaging agent for biomedical applications show promise for cancer diagnosis and therapy.
  • Therapeutic examples include nanoparticle containing PDT agents, folate receptor-targeted, boron containing dendrimers for neutron capture and nanoparticle-directed thermal therapy.
  • Nanoparticles have had disadvantages when considered for use in photodynamic therapy (PDT).
  • PDT photodynamic therapy
  • certain nanoparticles have no relatively large knowledge base on cancer imaging, PDT, chemical sensing, stability and biodegradation.
  • (2) have in in- vivo toxicity.
  • (3) Have short plasma circulation time without surface modification and unstable or uncontrollable biodegradation and bioelimination rates (4)
  • (5) have additional limitations including relative difficulty in incorporating hydrophobic compounds, leaching of small hydrophilic components unless they are "anchored", and unknown limitation on bulk tumor permeability because of hydrogel swelling.
  • Photodynamic therapy is a clinically effective and still evolving locally selective therapy for cancers.
  • the utility of PDT has been demonstrated with various photosensitizers for multiple types of disease. It is FDA approved for early and late stage lung cancer, obstructive esophageal cancer, high-grade dysplasia associated with Barrett's esophagus, age-related macular degeneration and actinic keratoses.
  • PDT employs tumor localizing photosensitizers that produce reactive singlet oxygen upon absorption of light which is believed to be responsible for the destruction of the tumor.
  • PDT Photodynamic therapy
  • Fluorescence the mission of absorbed light at a longer wavelength, can be highly sensitive: a typical cyanine dye with a lifetime of 0.6 nsec can emit up to 1032 photons/second/mole. A sensitive optical detector can image ⁇ 103 photons/second. Thus even with low excitation power, low levels of fluorescent molecular beacons can be detected. A challenge is to deliver the dyes selectively and in high enough concentration to detect small tumors. Use of ICG alone to image hypervascular or "leaky" angiogenic vessels around tumors has been disappointing, due to its limited intrinsic tumor selectivity.
  • Photosensitizers generally fluoresce and their fluorescence properties in vivo has been exploited for the detection of early-stage cancers in the lung, bladder and other sites. For treatment of early disease or for deep seated tumors the fluorescence can be used to guide the activating light.
  • photosensitizers are not optimal fluorophores for tumor detection for several reasons: (i) They have low fluorescence quantum yields (especially the long wavelength photosensitizers related to bacteriochlorins).
  • Efficient photosensitizer tend to have lower fluorescence efficiency (quantum yield) than compounds designed to be fluorophores, such as cyanine dyes because the excited singlet state energy emitted as fluorescence is instead transferred to the triplet state and then to molecular oxygen .
  • fluorophores such as cyanine dyes
  • They have small Stokes shifts.
  • Porphyrin-based photosensitizer have a relatively small difference between the long wavelength absorption band and the fluorescence wavelength (Stokes shift), which makes it technically difficult to separate the fluorescence from the excitation wavelength
  • Most photosensitizers have relatively short fluorescent wavelengths, ⁇ 800 nm, which are not optimal for detection deep in tissues.
  • PET Positron emission tomography
  • PET is a technique that permits non-invasive use of radioisotope labeled molecular imaging probes to image and assay biochemical processes at the level of cellular function in living subjects20.
  • PET predominately has been used as a metabolic marker, without specific targeting to malignancies.
  • radiolabeled peptide ligands to target malignancies.
  • PET is important in clinical care and is a critical component in biomedical research, supporting a wide range of applications, including studies of tumor hypoxia, apoptosis and angiogenesis21.
  • a long circulation time may be desirable, as it can increase delivery of the agent into tumors.
  • the present invention relates to PAA (polyacrylic acid and derivatives thereof at the carboxy moiety, e.g. polyacrylamide) containing a photosensitizer and an imaging enhancing agent.
  • PAA polyacrylic acid and derivatives thereof at the carboxy moiety, e.g. polyacrylamide
  • PAA nanoparticles have core matrixes that can readily incorporate molecular or small nanoparticle payloads, and can be prepared in 10-150 nm sizes, with good control of size distributions.
  • the surfaces of nanoparticles can be readily functionalized, to permit attachment of targeting ligands, and both are stable to singlet oxygen (102) produced during photodynamic therapy (PDT).
  • PDT photodynamic therapy
  • poly(acrylic acid) nanoparticles have the advantages of (1) A relatively large knowledge base on cancer imaging, PDT, chemical sensing, stability and biodegradation. (2) No known in-vivo toxicity. (3) Long plasma circulation time without surface modification, but with biodegradation and bioelimination rates controllable via the type and amount of selective cross-linking (introduced during polymerization inside reverse micelles). (4) Scale-up to 400g material has been demonstrated, as well as storage stability over extended periods. Limitations have included relative difficulty in incorporating hydrophobic compounds, leaching of small hydrophilic components unless they are "anchored", and unknown limitation on bulk tumor permeability because of hydrogel swelling.
  • photosensitizers have several very desirable properties as therapeutic agents deliverable by PAA nanoparticles.
  • PDT provides dual selectivity in that the photosensitizer is inactive in the absence of light and is innocuous without photoactivation.
  • the photosensitizer contained by the nanoparticle can be locally activated at the site of disease.
  • PDT effects are due to production of singlet oxygen, which, in accordance with the compounds and methods of the invention, can readily diffuse from the pores of the nanoparticle.
  • nanoparticle platforms also provide significant advantages for PDT: (1) High levels of imaging agents can be combined with the photosensitizer in the nanoparticle permitting a "see and treat” approach, with fluorescence image guided placement of optical fibers to direct the photoactivating light to large or subsurface tumors, or to early non clinically evident disease. (2) It is possible to add targeting moieties, such as cRGD or F3 peptide to the nanoparticle so as to increase the selective delivery of the photosensitizer.
  • targeting moieties such as cRGD or F3 peptide
  • the nanoparticle can carry large numbers of photosensitizers, and their surface can be modified to provide the desired hydrophilicity for optimal plasma pharmacokinetics. Thus, they can deliver high levels of photosensitizer to tumors, reducing the amount of light necessary for tumor cure.
  • the photosensitizer is preferably a tetrapyrrolic photosensitizer having the structural formula:
  • X is an aryl or heteroaryl group
  • n is an integer of 0 to 6;
  • R 2 o is methyl, butyl, heptyl, docecyl or 3,5-bis(trifluoromethyl)-benzyl
  • R 2 i is 3,5,-bis(trifluoromethyl)benzyl
  • Ri a and R 2a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;
  • R 3 and R4 are each independently hydrogen or substituted or unsubstituted alkyl;
  • R 3a and R4 a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;
  • R5 is hydrogen or substituted or unsubstituted alkyl
  • R 9 and Rio are each independently hydrogen, or substituted or unsubstituted alkyl and R9 may be -CH2CH2COOR 2 where R 2 is an alkyl group that may optionally substituted with one or more fluorine atoms;
  • the photosensitizer may be conjugated with an image enhancing agent prior to incorporation into the nanoparticle, after incorporation into the nanoparticle or the photosensitizer and/or image enhancing agent may chemically bound to the nano particle and/or one or more of the photosensitizer and image enhancing agent may be physically bound to the nanoparticle.
  • Imaging enhancing agents may be for essentially any imaging process, e.g.
  • imaging enhancing agents are discussed in the background of the invention previously discussed and in the list of references incorporated by reference herein as background art.
  • Figure 1A shows the structural formula of HPPH-CD (cyanine dye) conjugate used as a photosensitizer and imaging agent.
  • Figure IB is a graph showing in vivo photosensitizing efficacy of HPPH-CD conjugate 1 in C3H mice bearing RIF tumors (10 mice/ group) at variable drug doses. The tumors were exposed to light (135J/cm2/75mW/cm2) at 24h post-injection.
  • Figure 1 C shows a scanned image showing localization of the conjugate 1 in a live mouse 24 h after injection (drug dose 0.3 ⁇ ). (Without PAA NP0)
  • Figure 2 shows whole body images of BALB/c mice bearing Colon26 tumors with PAA NPs formulations (HPPH and cyanine dye (CD) were post-loaded in 2 to 1 ratio).
  • the CD concentration was kept constant (0.3 ⁇ /kg) at the images were obtained at variable time points.
  • Figure 4 Slow release of HPPH and CD from PAA NPs (post loaded in 2: 1 ratio) after several washes with 1% HSA.
  • Figure 5 A is a diagram showing structure of PAA nanoparticles (PAA NP's)
  • FIG. 6 shows a series of scans wherein Panel 1 (4T1 tumors): Primary (PT) and metastasized tumors (MT) dissected and Panel 2 (4T1 tumors): PET imaging of the dissected primary and metastasized tumors.
  • Panel 3 (BALB/C mouse bearing 4T1 tumor): Whole body PET imaging. The tumor metastasis in lung was clearly observed.
  • Panel 4 The position of the lung is shown by the transmission scan using 57Co source in mice with no lung metastasis.
  • Panel 5 (BALB/C mouse bearing Colo-26 (non-metastatic tumor): Whole body imaging by PET. A high accumulation of the 1241- photosensitizer in tumor is clearly observed without any significant accumulation in lungs (injected dose: 100 ⁇ ).
  • T Tumor
  • PT Primary tumor
  • MT Metastatic tumor.
  • Figure 8 A shows in vivo comparative in vivo PET imaging (72 h post injection) and biodistribution (24h, 48h and 72h postinjection) of 1241-labeled photosensitizer 2 without PAA nanoparticles in BALB/c mice bearing Colon26 tumors (see the text). (Biodistribution of PET imaging agent 2: No PAA, with PAA).
  • Figure 8B shows in vivo comparative in vivo PET imaging (72 h post injection) and biodistribution (24h, 48h and 72h postinjection) of 1241-labeled photosensitizer 2 with PAA nanoparticles in BALB/c mice bearing Colon26 tumors (see the text).
  • Biodistribution of PET imaging agent 2 No PAA, with PAA.
  • Figure 8C shows biodistribution of PET imaging agent 2, no PAA and with PAA.
  • FIG. 9 Fluorescence intensity of cells targeted by F3- targeted (A series), F3-Cys targeted (B series) and nontargeted NPs (F series) in nucleolin rich MDA-MB-435 cell lines.
  • FIG. 10 Fluorescence (left) & Live/dead cell assay (right) of HPPH conjugated PAA NPs + or - F3-Cys peptide incubated for 15 min with MDA-MB- 435 cells.
  • FIG. 1 Confocal images showing the target-specificity of F3-Cys peptide in 9L Glioma tumor cells. Left: F3-Cys PEG Rhodamine-PAA NPs (9L cells). Right: PEG Rhodamine-PAA NPs (9L Cells).
  • FIG. 12 In vivo biodistribution of 14 C-labeled HPPH, and 14 C-labeled HPPH post- loaded into PAA NPs in BALB/c mice bearing Colon26 tumors. 14 C-labeled PS (3.8 ⁇ /0.2 mL) were administered to 12 mice/group. At 24, 48, 72h after, injection, three mice/time- point were sacrificed. The organs of interest were removed and the radioactivity was measured The raw data were converted to counts/ gram of tissue.
  • Figure 13A shows. In vivo biodistribution of iodinated photosensitizer at 24, 48 and 72h post inj ection.
  • Figure 17A shows the structure of cRGDfK (an ay 3 ligand).
  • Figure 17B shows a confocal fluorescence image of MDA-MB-435 cell treated with cRGD-conjugated FITC PAA NP's. The image using the conjugate is excellent.
  • Figure 17C shows fluorescence image of MDA-MB-435 cells treated with unmodified FITC PAA NP's. The image shows no cell distinction.
  • Figure 18 shows an image of tumor growth in the brain of a mouse using photos ens itizer and cyanine dye with PAA nanoparticles.
  • Figure 19A at A shows a fluorescence image of a control at A with no cyanine dye.
  • Figure 19A at B is a fluorescence image using cyanine dye (CD) containing a carboxylic acid group conjugated at the periphery of the PAA NPs and
  • Figure 19A at C is a fluorescence image using cyanine dye alone. Essentially no tumor uptake is shown.
  • CD cyanine dye
  • Figure 19B shows structural formulas for cyanine dye (CD) which showed no tumor avidity and compound 2 conjugate of cyanine dye with an amine containing PAA nanoparticle.
  • FIGS 20A through 20D show schemes for various approaches for preparation of multifunctional nanoparticles.
  • Figure 21 shows a chart for a method for comparative study of MR multifunctional agents.
  • Figure 22 shows a chart for methodology for imaging and treating brain tumors.
  • Photosensitizers generally fluoresce and their fluorescence properties in vivo has been exploited for the detection of early-stage cancers in the lung, bladder and other sites. For treatment of early disease or for deep seated tumors the fluorescence can be used to guide the activating light.
  • photosensitizers are not optimal fluorophores for tumor detection for several reasons: (i) They have low fluorescence quantum yields (especially the long wavelength photosensitizers related to bacteriochlorins).
  • Efficient photosensitizers tend to have lower fluorescence efficiency (quantum yield) than compounds designed to be fluorophores, such as cyanine dyes because the excited singlet state energy emitted as fluorescence is instead transferred to the triplet state and then to molecular oxygen .
  • fluorophores such as cyanine dyes
  • They have small Stokes shifts.
  • Porphyrin-based photosensitizer have a relatively small difference between the long wavelength absorption band and the fluorescence wavelength (Stokes shift), which makes it technically difficult to separate the fluorescence from the excitation wavelength
  • Most photosensitizers have relatively short fluorescent wavelengths, ⁇ 800 nm, which are not optimal for detection deep in tissues.
  • HPPH conjugated with R absorbing fluorophore(s) can be used as bifunctional agents for tumor-imaging by fluorescence and phototherapy (PDT).
  • PDT fluorescence and phototherapy
  • HPPH was used as a vehicle to deliver the imaging agent to tumor.
  • the limitation of this approach was that the conjugate exhibited significantly different dose requirements for the two modalities.
  • the imaging dose was approximately 10-fold lower than the phototherapeutic dose (Fig. IB and 1C), which could be due to a part of the singlet oxygen (a key cytotoxic agent responsible for the destruction of the tumors) produced on exciting the photosensitizer being quenched by the fluorophore leading to its photo-destruction.
  • HPPH and the cyanine dye were post-loaded in variable ratios (HPPH to CD: 1 : 1; 2: 1 ; 3: 1 and 4: 1 molar concentrations).
  • HPPH was postloaded to PAA nanoparticles first. Free HPPH was removed by spin filtration and then cyanine dye was postloaded. It was spin-filtered again, washed several times with 1% bovine calf serum and the concentration was measured.
  • the 2: 1 formulations produce the best tumor imaging and long-term tumor cure in BALB/c mice bearing Colon26 tumors.
  • This formulation contained in a single dose the therapeutic dose of HPPH (0.47 ⁇ /kg) and the imaging dose of Cyanine dye (0.27 mol/kg), which were similar to the components used alone for tumor imaging and therapy, but with much more tumor selectivity (skin to tumor ratio of HPPH was 4: 1 instead of 2: 1 without nanoparticles). Under similar treatment parameters the Ormosil nanoparticles showed a significantly reduced response (imaging and PDT, not shown).
  • the stability of the drugs in PAA nanoparticle was established by repeated washing with aqueous bovine calf serum through Amicon centrifugal filter units with a lOOKDa or larger cut off membrane and drug in the filtrate was measured spectrophotometrically.
  • Figs. 2-4 The comparative in vivo PDT efficacy of the ORMOSIL and PAA formulations, their tumor imaging potential and stability (in vitro release kinetics) is shown in Figs. 2-4, which clearly illustrate the advantages of PAA nanoparticles in reducing the therapeutic dose by almost 8- fold without diminishing the tumor-imaging potential and also avoiding the Tween-80 formulation required for the HPPH-CD conjugate 1.
  • the HPPH CD conjugate 1 was post-loaded to PAA nanoparticles, which certainly enhanced the tumorimaging, but the therapeutic dose was still 10-fold higher (similar to the HPPH CD conjugate, Fig. 5B).
  • the cyanine dye was conjugated peripherally to the PAA nanoparticles first and then HPPH was post loaded. Again, compared to HPPH-CD conjugate 1, the PAA formulation showed enhanced tumor-specificity (imaging) (Fig.5B).
  • Chang et al reported an effective radius of tumor cell kill in 22 glioma patients of 8 mm compared with the 1.5 cm depth of necrosis noted by Pierria with the intracavitary illumination method. It is believed that tumor resection is important so that the numbers of tumor cells remaining to treat are minimized. With stereotactic implantation of fibers for interstitial PDT there is no cavity to accommodate swelling and a considerable volume of necrotic tumor which causes cerebral edema. However, cerebral edema can be readily controlled with steroid therapy. Compared to chemotherapy and radiotherapy, patients with brain tumors treated with PDT have definitely shown long-term survival, whereas glioma patients treated with adjuvant chemotherapy or radiotherapy do not seem to show additional benefits. On the basis of our preliminary data, the ⁇ 3 targeted nanoparticles may improve tumor-selectivity and PDT outcome.
  • PET imaging and PDT PAA nanoparticles decreased the liver uptake of the 1241- photosensitizer (PET imaging agent) and enhanced the tumor-specificity
  • radioactive photosensitizer such as the 1241- labeled analog 2 (superior to 18F-FDG in PET-imaging of lung, brain, breast and pancreas tumors) with a T1 ⁇ 2 of 4.2 days could cause radiation damage to normal organs.
  • radioactive photosensitizer such as the 1241- labeled analog 2 (superior to 18F-FDG in PET-imaging of lung, brain, breast and pancreas tumors) with a T1 ⁇ 2 of 4.2 days could cause radiation damage to normal organs.
  • PAA nanoparticles made a remarkable difference in tumor contrast with brain, lung and pancreatic tumors). See Fig. 7 for comparative biodistribution. PAA nanoparticles can be targeted to nucleolin with F3-Cys.
  • F3-targeted nanoparticles were prepared using two kinds of F3 peptides: F3 peptide conjugated to nanoparticle via one of the 8 lysines available in its sequence and F3- Cys peptide conjugated to nanoparticle via cysteine. Cysteine capped nanoparticles served as non-targeted control.
  • Three 25 mg batches of each type of nanoparticle contained: 2.6, 5.1 and 7.7 mg F3, (A3-A5) respectively; 2.7, 5.3 and 8 mg F3-Cys (B3-B5) respectively, and 0.29, 0.58 and 0.87 mg Cys (C3-C5) respectively.
  • the fluorescence intensity from PAA nanoparticle incubated in vitro with nucleolin positive MDA-MB-435 cells is shown in Fig. 9.
  • the F3-Cys conjugated nanoparticles show considerably higher binding efficiency than non-targeted nanoparticles, while F3 conjugated nanoparticles do not.
  • Conjugation via a cysteine link preserves the specificity of F3 peptide for nucleolin.
  • excess cysteine on the nanoparticles helphotosensitizer to minimize the non-specific binding. Additional experiments (not shown) suggested that the amount of F3-Cys peptide (5.3 mg/25mg nanoparticle) used for B4 nanoparticles was optimal.
  • HPPH conjugated PAA nanoparticles with F3-Cys peptide at the outer surface show targeted specificity:
  • F3-mediated specificity is retained in the presence of conjugated HPPH.
  • F3 targeted nanoparticles did targeted nanoparticles did not, indicating that F3 -mediated specificity is retained in the presence of conjugated HPPH.
  • F3 targeted nanoparticles did not accumulate in the nucleus.
  • HPPH conjugated PAA nanoparticles with F3-Cyspeptide at the outer surface show targeted specificity:
  • This invention shows the utility of porphyrin-based compounds in a
  • bifunctional agent for imaging breast tumor and tumor metastasis. Similar to most nanoparticles, PAA nanoparticles accumulate in liver and spleen. Their clearance rate from most organs is significantly faster than Ormosil nanoparticles and they do not show long-term organ toxicity. Even tumor-avid porphyrin based photos ens itizers exhibit high uptake in liver and spleen, but are non-toxic until exposed to light. The photos ens itizers clear from the system quickly (days) without organ toxicity.
  • HPPH 660 nm
  • PS photos ens itizer
  • Aim 1 To investigate the utility of PAA nanoparticles in developing multifunctional platforms by (i) post-loading both the monomeric therapeutic and imaging agents, and (ii) conjugating the fluorescence agent at the periphery of the NPs and post- loading the PET/PDT agent.
  • Aim 2 Conjugate cRGD to the best candidate selected from each series (Aims).
  • FIG. 17A shows structure of cRGDfK.
  • Figure 17B shows a confocal fluorescence image of MDA- MB-435 cell treated with cRGD-conjugated FITC PAA NP's and
  • Figure 17C shows fluorescence image of MDA-MB-435 cells treated with unmodified FITC PAA NP's.
  • Figure 19A shows a control at A, cyanine dye-COOH conjugated to PAA nanoparticles (conjugate 2 48 h post injection) at B and free cyanine dye-COOH (48 h post-injection) at C. Dosages for B and C were 0.3 ⁇ 1 ⁇ /13 ⁇ 4.
  • Figure 19B shows structures for cyanine dye and cyanine dye- nanoparticle conjugate.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

A PAA nanoparticle containing a covalently linked fluorescent dye and a post-loaded tetrapyrrolic photosensitizer.ln a first embodiment, a composition comprising PAA nanoparticles containing a tetrapyrollic photosensitizer post loaded to the PAA nanoparticle after nanoparticle formation and an imaging agent covalently bonded to the PAA nanoparticle and a tumor targeting moiety covalently bonded to the PAA nanoparticles.

Description

PAA NANOPLATFORMS CONTAINING FLUOROPHORES AND TARGETED MOIETIES COVALENTLY LINKED AND PHOTOSENSITIZER POST-LOADED CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority from U.S.A. Application No. 13/566,444, filed
August 3, 2012; which application is incorporated by reference herein in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] This invention was made with United States of America Government support under U.S. Grant Numbers CAl 19358 and CAl 14053 awarded by the United States National
Institute of Health. The United States Government has certain rights in the invention.
BACKGROUND OF THE INVENTION [0003] Nanoscience is being developed in conjunction with advanced medical science for further precision in diagnosis and treatment. Nanoplatforms and nanovectors that deliver a therapeutic or imaging agent for biomedical applications show promise for cancer diagnosis and therapy. Therapeutic examples include nanoparticle containing PDT agents, folate receptor-targeted, boron containing dendrimers for neutron capture and nanoparticle-directed thermal therapy.
[0004] Nanoparticles have had disadvantages when considered for use in photodynamic therapy (PDT). In particular, certain nanoparticles have no relatively large knowledge base on cancer imaging, PDT, chemical sensing, stability and biodegradation. (2) have in in- vivo toxicity. (3) Have short plasma circulation time without surface modification and unstable or uncontrollable biodegradation and bioelimination rates (4) Have problems associated with scale-up and are not storage stabile over extended periods. And (5) have additional limitations including relative difficulty in incorporating hydrophobic compounds, leaching of small hydrophilic components unless they are "anchored", and unknown limitation on bulk tumor permeability because of hydrogel swelling.
[0005] A major challenge of cancer therapy is preferential destruction of malignant cells with sparing of normal tissue. Critical for successful eradication of malignant disease are early detection and selective ablation of the malignancy. Photodynamic therapy (PDT) is a clinically effective and still evolving locally selective therapy for cancers. The utility of PDT has been demonstrated with various photosensitizers for multiple types of disease. It is FDA approved for early and late stage lung cancer, obstructive esophageal cancer, high-grade dysplasia associated with Barrett's esophagus, age-related macular degeneration and actinic keratoses. PDT employs tumor localizing photosensitizers that produce reactive singlet oxygen upon absorption of light which is believed to be responsible for the destruction of the tumor. Subsequent oxidation-reduction reactions also can produce superoxide anions, hydrogen peroxide and hydroxyl radicals which contribute to tumor ablation4. Photosensitizers have been designed which localize relatively specifically to certain subcellular structures such as mitochondria, which are highly sensitive targets. On the tumor tissue level, direct photodynamic tumor cell kill, destruction of the tumor supporting vasculature and possibly activation of the innate and adaptive anti-tumor immune system interact to destroy the malignant tissue6. The preferential killing of the targeted cells (e.g. tumor), rather than adjacent normal tissues, is essential for PDT, and the preferential target damage achieved in clinical applications is a major driving force behind the use of the modality. The success of PDT relies on development of tumor-avid molecules that are preferentially retained in malignant cells but cleared from normal tissues.
[0006] Malignant brain tumors (gliomas) are generally resistant to conventional aggressive treatments by surgery, radiation, and chemotherapy. Over 80% of recurrences are within 2 cm of the original tumor margin. The prognosis after glioma surgery is partly determined by the precision of surgical resection, which may be sub-optimal since the intraoperative identification of tumor margins or small foci of cancer cells depends on visual inspection. Given the potent nature of brain tumors, better treatments are essential to improve response. Photodynamic therapy (PDT) for the treatment of a variety of brain tumors, in particular gliomas have been investigated in laboratory studies and clinical trials. The main advantage of PDT lies in its ability to select out tumor cells that are infiltrating brain parenchyma and that are responsible for local tumor recurrence, which is the main therapeutic dilemma in the treatment of gliomas.
[0007] In efforts to develop effective photosensitizers with the required photophysical characteristics, compounds having a tetrapyrrolic core ring were used. Usually, chlorophyll-a and bacteriochlorophyll-a were used as intermediates in synthesis. Extensive QSAR studies on a series of the alkyl ether derivatives of pyropheophorbide-a (660 nm) led to selection of HPPH (hexyl ether derivative), now in promising Phase II clinical trials. Photosensitizer development now extends to purpurinimide (700 nm) and bacteriopurpurinimde (780-800 nm) series with high singlet oxygen producing capability. Long wavelength absorption is important for treating large deep seated tumors, because longer wavelength light increases penetration and minimizes the number of optical fibers needed for light delivery within the tumor.
[0008] Various efforts have been made to target tumor cells so that an agent may destroy the tumor cells while sparing normal cells. Such systems are reliant upon specific receptors and as such must reach receptor location. This is a disadvantage since even though the agent may reach the targeted cell, it may not be effective unless the particular receptor is reached and bound. [0009] Multiple, complementary techniques for tumor detection, including magnetic resonance, scintigraphic and optical imaging are under active development. Each approach has particular strengths and advantages. Optical imaging includes measurement of absorption of endogenous molecules (e. g. hemoglobin) or administered dyes, detection of bioluminescence in preclinical models, and detection of fluorescence from endogenous fluorophores or from targeted exogenous molecules. Fluorescence, the mission of absorbed light at a longer wavelength, can be highly sensitive: a typical cyanine dye with a lifetime of 0.6 nsec can emit up to 1032 photons/second/mole. A sensitive optical detector can image <103 photons/second. Thus even with low excitation power, low levels of fluorescent molecular beacons can be detected. A challenge is to deliver the dyes selectively and in high enough concentration to detect small tumors. Use of ICG alone to image hypervascular or "leaky" angiogenic vessels around tumors has been disappointing, due to its limited intrinsic tumor selectivity. Multiple approaches have been employed to improve optical probe- localization, including administering it in a quenched form that is activated within tumors, or coupling it to antibodies or small molecules such as receptor ligands. Recent studies have focused on developing dye conjugates of small bioactive molecules, to improve rapid diffusion to target tissue and use combinatorial and high throughput strategies to identify, optimize, and enhance in vivo stability of the new probes. Some peptide analogs of ICG derivatives have moderate tumor specificity and are entering pre-clinical studies. However, none of these compounds are designed for both tumor detection and therapy. It is important to develop targeting strategies that cope with the heterogeneity of tumors in vivo, where there are inconsistent and varying expressions of targetable sites.
[0010] Photosensitizers (photosensitizer) generally fluoresce and their fluorescence properties in vivo has been exploited for the detection of early-stage cancers in the lung, bladder and other sites. For treatment of early disease or for deep seated tumors the fluorescence can be used to guide the activating light. However, photosensitizers are not optimal fluorophores for tumor detection for several reasons: (i) They have low fluorescence quantum yields (especially the long wavelength photosensitizers related to bacteriochlorins). Efficient photosensitizer tend to have lower fluorescence efficiency (quantum yield) than compounds designed to be fluorophores, such as cyanine dyes because the excited singlet state energy emitted as fluorescence is instead transferred to the triplet state and then to molecular oxygen . (ii) They have small Stokes shifts. Porphyrin-based photosensitizer have a relatively small difference between the long wavelength absorption band and the fluorescence wavelength (Stokes shift), which makes it technically difficult to separate the fluorescence from the excitation wavelength, (iii) Most photosensitizers have relatively short fluorescent wavelengths, < 800 nm, which are not optimal for detection deep in tissues.
[0011] Attempts have been made to develop bifunctional conjugates that use tumor- avid photosensitizer to target the MR fluorophores to the tumor. The function of the fluorophore is to visualize the tumor location and treatment site. The presence of the photosensitizer allows subsequent tumor ablation. The optical imaging allows the clinician performing PDT to continuously acquire and display patient data in real-time. This "see and treat" approach may determine where to treat superficial carcinomas and how to reach deep- seated tumors in sites such as the breast, lung and brain with optical fibers delivering the photo-activating light. A similar approach was also used for developing potential PDT/MRI conjugates in which HPPH was conjugated with Gd(III)DTPA Due to a significant difference between imaging and therapeutic doses, the use of a single molecule that includes both modalities is problematic.
[0012] Positron emission tomography (PET) is a technique that permits non-invasive use of radioisotope labeled molecular imaging probes to image and assay biochemical processes at the level of cellular function in living subjects20. PET predominately has been used as a metabolic marker, without specific targeting to malignancies. Recently, there has been growing use of radiolabeled peptide ligands to target malignancies. Currently, PET is important in clinical care and is a critical component in biomedical research, supporting a wide range of applications, including studies of tumor hypoxia, apoptosis and angiogenesis21. For targeting, a long circulation time may be desirable, as it can increase delivery of the agent into tumors. HPPH and the iodobenzyl pheophorbide-a have plasma half lives ~25 h. The long radiological half life of 124I is well matched to the pheophorbides; it permits sequential imaging with time for clearance from normal tissue. Labeling techniques with radioiodine are well defined with good yield and radiochemical purity22. Despite the complex decay scheme of 124I which results in only 25% abundance of positron (compared with 100% positron emission of 18F), in vivo quantitative imaging with 124I labeled antibodies has been successfully carried out under realistic conditions using a PET/CT scanner A variety of biomolecules have been labeled with124! We have devised a coupling reaction which rapidly and efficiently links 124I to a tumor-avid photosensitizer23-25, and used the conjugate to target and image murine breast tumor and its metastasis to lung Acquisition of clinical PET images can be slow, but combination PET-CT scanners allow real time guidance of therapeutic interventions. Also, new developments in tracking may permit real time interventions guided by PET data sets. BRIEF SUMMARY OF THE INVENTION
[0013] The present invention relates to PAA (polyacrylic acid and derivatives thereof at the carboxy moiety, e.g. polyacrylamide) containing a photosensitizer and an imaging enhancing agent.
[0014] In accordance with the invention, therapeutic and imaging potential of encaphotosensitizerulated, post-loaded and covalently linked photosensitizer-nanoparticles have been evaluated. In PAA nanoparticle the post-loading efficiency showed enhanced in vitro/in vivo therapeutic and imaging potential. PAA nanoparticle have core matrixes that can readily incorporate molecular or small nanoparticle payloads, and can be prepared in 10-150 nm sizes, with good control of size distributions. The surfaces of nanoparticles can be readily functionalized, to permit attachment of targeting ligands, and both are stable to singlet oxygen (102) produced during photodynamic therapy (PDT). PAA-nanoparticles, i.e. poly(acrylic acid) nanoparticles, have the advantages of (1) A relatively large knowledge base on cancer imaging, PDT, chemical sensing, stability and biodegradation. (2) No known in-vivo toxicity. (3) Long plasma circulation time without surface modification, but with biodegradation and bioelimination rates controllable via the type and amount of selective cross-linking (introduced during polymerization inside reverse micelles). (4) Scale-up to 400g material has been demonstrated, as well as storage stability over extended periods. Limitations have included relative difficulty in incorporating hydrophobic compounds, leaching of small hydrophilic components unless they are "anchored", and unknown limitation on bulk tumor permeability because of hydrogel swelling.
[0015] In accordance with the invention, photosensitizers have several very desirable properties as therapeutic agents deliverable by PAA nanoparticles. In particular, (1) Only a very small fraction of administered targeted non-photodynamic drug makes it to tumor sites and the remainder can cause systemic toxicity. However, PDT provides dual selectivity in that the photosensitizer is inactive in the absence of light and is innocuous without photoactivation. Thus the photosensitizer contained by the nanoparticle can be locally activated at the site of disease. (2) PDT effects are due to production of singlet oxygen, which, in accordance with the compounds and methods of the invention, can readily diffuse from the pores of the nanoparticle. Thus, in contrast to chemotherapeutic agents, release of encaphotosensitizerulated drug from the nanoparticle, is not necessary. Instead, stable nanoparticles with long plasma residence times can be used, which increases the amount of drug delivered to the tumors. (3) PDT is effective regardless of the intracellular location of the photosensitizer. While mitochondria are a principal target of singlet oxygen, photosensitizer incorporated in lysosomes are also active the photodynamic process causes rupture of the lysosomes with release of proteolytic enzymes and redistribution of the photosensitizer within the cytoplasm, nanoparticle platforms also provide significant advantages for PDT: (1) High levels of imaging agents can be combined with the photosensitizer in the nanoparticle permitting a "see and treat" approach, with fluorescence image guided placement of optical fibers to direct the photoactivating light to large or subsurface tumors, or to early non clinically evident disease. (2) It is possible to add targeting moieties, such as cRGD or F3 peptide to the nanoparticle so as to increase the selective delivery of the photosensitizer. (3) The nanoparticle can carry large numbers of photosensitizers, and their surface can be modified to provide the desired hydrophilicity for optimal plasma pharmacokinetics. Thus, they can deliver high levels of photosensitizer to tumors, reducing the amount of light necessary for tumor cure.
[0016] The photosensitizer is preferably a tetrapyrrolic photosensitizer having the structural formula:
Figure imgf000010_0001
or a pharmaceutically acceptable derivative thereof, wherein:
Ri and R2 are each independently substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, -C(0)Ra or -COORa or -CH(CH3)(ORa) or -CH(CH3)(0(CH2)nXRa) where Ra is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, or substituted or unsubstituted cycloalkyl; where R2 may be -CH=CH2, -CH(OR20)CH3, -C(0)Me, -C(=NR2i)CH3 or -CH(NHR21)CH3
where X is an aryl or heteroaryl group;
n is an integer of 0 to 6;
where R2o is methyl, butyl, heptyl, docecyl or 3,5-bis(trifluoromethyl)-benzyl; and
R2i is 3,5,-bis(trifluoromethyl)benzyl;
Ria and R2a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;
R3 and R4 are each independently hydrogen or substituted or unsubstituted alkyl; R3a and R4a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;
R5 is hydrogen or substituted or unsubstituted alkyl;
R6 and R6a are each independently hydrogen or substituted or unsubstituted alkyl, or together form =0;
R7 is a covalent bond, alkylene, azaalkyl, or azaaraalkyl or =NR2o where R2o is 3,5- bis(tri-fluoromethyl)benzyl or -CH2X-R1 or -YR1 where Y is an aryl or heteroaryl group;
Rs and Rsa are each independently hydrogen or substituted or unsubstituted alkyl or together form =0; R9 and Rio are each independently hydrogen, or substituted or unsubstituted alkyl and R9 may be -CH2CH2COOR2 where R2 is an alkyl group that may optionally substituted with one or more fluorine atoms;
each of R1-R10, when substituted, is substituted with one or more substituents each independently selected from Q, where Q is alkyl, haloalkyl, halo, photosensitizereudohalo, or -COORb where R, is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, araalkyl, or ORc where Rc is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl or CONRjRe where Rd and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or NRfRg where Rf and Rg are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or =NRh where Rh is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or is an amino acid residue;
each Q is independently unsubstituted or is substituted with one or more substituents each independently selected from Qi, where Qi is alkyl, haloalkyl, halo, photosensitizereudohalo, or -COORb where Rb is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, araalkyl, or ORc where Rc is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl or CONRdRe where Rj and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or NRfRg where Rf and Rg are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or =NRh where Rh is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or is an amino acid residue.
[0017] The photosensitizer may be conjugated with an image enhancing agent prior to incorporation into the nanoparticle, after incorporation into the nanoparticle or the photosensitizer and/or image enhancing agent may chemically bound to the nano particle and/or one or more of the photosensitizer and image enhancing agent may be physically bound to the nanoparticle. [0018] Imaging enhancing agents may be for essentially any imaging process, e.g.
Examples of such imaging enhancing agents are discussed in the background of the invention previously discussed and in the list of references incorporated by reference herein as background art.
[0019] It is to be understood that other agents may be incorporated into the nanoparticle such as tumor targeting moieties and tumor inhibiting or tumor toxic moieties.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
Figure 1A shows the structural formula of HPPH-CD (cyanine dye) conjugate used as a photosensitizer and imaging agent.
Figure IB is a graph showing in vivo photosensitizing efficacy of HPPH-CD conjugate 1 in C3H mice bearing RIF tumors (10 mice/ group) at variable drug doses. The tumors were exposed to light (135J/cm2/75mW/cm2) at 24h post-injection.
Figure 1 C shows a scanned image showing localization of the conjugate 1 in a live mouse 24 h after injection (drug dose 0.3 μιηοΐε^). (Without PAA NP0)
Figure 2 shows whole body images of BALB/c mice bearing Colon26 tumors with PAA NPs formulations (HPPH and cyanine dye (CD) were post-loaded in 2 to 1 ratio). The CD concentration was kept constant (0.3 μιηοΐ/kg) at the images were obtained at variable time points. A = 24 h, B = 48 h and C = 72 h post injection (λεχ: 785 nm; λΕιη: 830 nm). L = Low and H = High.
Figure 3 is a graph showing in vivo PDT efficacy of HPPH and CD post loaded in a ratio of 2: 1 and 4: 1 in PAA and ORMOSIL NPs. Note: HPPH dose: 0.47 μιηοΐ/kg in PAA NPs and 0.78 μιηοΐ/kg in ORMOSIL NPs.
Figure 4. Slow release of HPPH and CD from PAA NPs (post loaded in 2: 1 ratio) after several washes with 1% HSA. Figure 5 A is a diagram showing structure of PAA nanoparticles (PAA NP's)
Figure 5B shows comparative in vivo imaging at variable time points of BALB/c mice bearing Colon26 tumors with HPPH-CD conjugate 1 and CD-conjugated with PAA NPs/post;-loaded with HPPH. The NPs were more tumor specific. (Mouse 1)
Figure 6 shows a series of scans wherein Panel 1 (4T1 tumors): Primary (PT) and metastasized tumors (MT) dissected and Panel 2 (4T1 tumors): PET imaging of the dissected primary and metastasized tumors. Panel 3 (BALB/C mouse bearing 4T1 tumor): Whole body PET imaging. The tumor metastasis in lung was clearly observed. Panel 4: The position of the lung is shown by the transmission scan using 57Co source in mice with no lung metastasis. Panel 5: (BALB/C mouse bearing Colo-26 (non-metastatic tumor): Whole body imaging by PET. A high accumulation of the 1241- photosensitizer in tumor is clearly observed without any significant accumulation in lungs (injected dose: 100 μθ). T = Tumor, PT = Primary tumor; MT = Metastatic tumor.
Figure 7. In vivo biodistribution of 18F-FDG (100 μθϊ, half-life 2 h) at 110 min and 124I-PS 2 (100 μθϊ, half-life 4.2 d) at 48h in BALB/c mice bearing Colon 26 tumor (3 mice/group). Tumor-uptake was similar for both agents. However, the higher uptake of FDG over 124I-PS 2 in normal organs is clearly evident.
Figure 8 A shows in vivo comparative in vivo PET imaging (72 h post injection) and biodistribution (24h, 48h and 72h postinjection) of 1241-labeled photosensitizer 2 without PAA nanoparticles in BALB/c mice bearing Colon26 tumors (see the text). (Biodistribution of PET imaging agent 2: No PAA, with PAA).
Figure 8B shows in vivo comparative in vivo PET imaging (72 h post injection) and biodistribution (24h, 48h and 72h postinjection) of 1241-labeled photosensitizer 2 with PAA nanoparticles in BALB/c mice bearing Colon26 tumors (see the text). (Biodistribution of PET imaging agent 2: No PAA, with PAA). Figure 8C shows biodistribution of PET imaging agent 2, no PAA and with PAA.
Figure 9. Fluorescence intensity of cells targeted by F3- targeted (A series), F3-Cys targeted (B series) and nontargeted NPs (F series) in nucleolin rich MDA-MB-435 cell lines.
Figure 10. Fluorescence (left) & Live/dead cell assay (right) of HPPH conjugated PAA NPs + or - F3-Cys peptide incubated for 15 min with MDA-MB- 435 cells.
Figure 1 1. Confocal images showing the target-specificity of F3-Cys peptide in 9L Glioma tumor cells. Left: F3-Cys PEG Rhodamine-PAA NPs (9L cells). Right: PEG Rhodamine-PAA NPs (9L Cells).
Figure 12. In vivo biodistribution of 14C-labeled HPPH, and 14C-labeled HPPH post- loaded into PAA NPs in BALB/c mice bearing Colon26 tumors. 14C-labeled PS (3.8 μΟί/0.2 mL) were administered to 12 mice/group. At 24, 48, 72h after, injection, three mice/time- point were sacrificed. The organs of interest were removed and the radioactivity was measured The raw data were converted to counts/ gram of tissue.
Figure 13A shows. In vivo biodistribution of iodinated photosensitizer at 24, 48 and 72h post inj ection.
Figure 13B shows. In vivo biodistribution of iodinated photosensitizer using variable sizes of PAA NPs at 24, 48 and 72h post injection 531-ME Post-Loaded into 30 nm PAA Nanoparticles.
Figure 13C shows. In vivo biodistribution of iodinated photosensitizer using variable sizes of PAA NPs at 24, 48 and 72h post injection 531-ME Post-Loaded into 150 nm PAA Nanoparticles.
Figure 14 shows the structural formula of HPPH.
Figure 15 is a diagram of Multifunctional PAA Nanoparticles.
Figure 16 shows flow diagrams for preparation of postloaded nanoparticles.
Figure 17A shows the structure of cRGDfK (an ay 3 ligand). Figure 17B shows a confocal fluorescence image of MDA-MB-435 cell treated with cRGD-conjugated FITC PAA NP's. The image using the conjugate is excellent.
Figure 17C shows fluorescence image of MDA-MB-435 cells treated with unmodified FITC PAA NP's. The image shows no cell distinction.
Figure 18 shows an image of tumor growth in the brain of a mouse using photos ens itizer and cyanine dye with PAA nanoparticles.
Figure 19A at A shows a fluorescence image of a control at A with no cyanine dye. Figure 19A at B is a fluorescence image using cyanine dye (CD) containing a carboxylic acid group conjugated at the periphery of the PAA NPs and Figure 19A at C is a fluorescence image using cyanine dye alone. Essentially no tumor uptake is shown.
Figure 19B shows structural formulas for cyanine dye (CD) which showed no tumor avidity and compound 2 conjugate of cyanine dye with an amine containing PAA nanoparticle.
Figures 20A through 20D show schemes for various approaches for preparation of multifunctional nanoparticles.
Figure 21 shows a chart for a method for comparative study of MR multifunctional agents.
Figure 22 shows a chart for methodology for imaging and treating brain tumors.
DETAILED DESCRIPTION OF THE INVENTION
[0020] Photosensitizers (photosensitizer) generally fluoresce and their fluorescence properties in vivo has been exploited for the detection of early-stage cancers in the lung, bladder and other sites. For treatment of early disease or for deep seated tumors the fluorescence can be used to guide the activating light. However, photosensitizers are not optimal fluorophores for tumor detection for several reasons: (i) They have low fluorescence quantum yields (especially the long wavelength photosensitizers related to bacteriochlorins). Efficient photosensitizers tend to have lower fluorescence efficiency (quantum yield) than compounds designed to be fluorophores, such as cyanine dyes because the excited singlet state energy emitted as fluorescence is instead transferred to the triplet state and then to molecular oxygen . (ii) They have small Stokes shifts. Porphyrin-based photosensitizer have a relatively small difference between the long wavelength absorption band and the fluorescence wavelength (Stokes shift), which makes it technically difficult to separate the fluorescence from the excitation wavelength, (iii) Most photosensitizers have relatively short fluorescent wavelengths, < 800 nm, which are not optimal for detection deep in tissues.
[0021] We have previously shown that certain tumor-avid photosensitizer(s) (e. g.,
HPPH) conjugated with R absorbing fluorophore(s) (non-tumor specific cyanine dyes) can be used as bifunctional agents for tumor-imaging by fluorescence and phototherapy (PDT). Here, HPPH was used as a vehicle to deliver the imaging agent to tumor. The limitation of this approach was that the conjugate exhibited significantly different dose requirements for the two modalities. The imaging dose was approximately 10-fold lower than the phototherapeutic dose (Fig. IB and 1C), which could be due to a part of the singlet oxygen (a key cytotoxic agent responsible for the destruction of the tumors) produced on exciting the photosensitizer being quenched by the fluorophore leading to its photo-destruction. Exposing the tumor at 780 nm (excitation wavelength for the cyanine dye) produced in vivo emission at 860 nm and, as expected, no significant photobleaching of the fluorophore (CD) or the photosensitizer (HPPH) was observed.
[0022] For investigating the utility of PAA nanoparticles three different approaches were used. First HPPH and the cyanine dye (fluorophore) were post-loaded in variable ratios (HPPH to CD: 1 : 1; 2: 1 ; 3: 1 and 4: 1 molar concentrations). In brief, HPPH was postloaded to PAA nanoparticles first. Free HPPH was removed by spin filtration and then cyanine dye was postloaded. It was spin-filtered again, washed several times with 1% bovine calf serum and the concentration was measured. The 2: 1 formulations produce the best tumor imaging and long-term tumor cure in BALB/c mice bearing Colon26 tumors. This formulation contained in a single dose the therapeutic dose of HPPH (0.47 μιηοΐ/kg) and the imaging dose of Cyanine dye (0.27 mol/kg), which were similar to the components used alone for tumor imaging and therapy, but with much more tumor selectivity (skin to tumor ratio of HPPH was 4: 1 instead of 2: 1 without nanoparticles). Under similar treatment parameters the Ormosil nanoparticles showed a significantly reduced response (imaging and PDT, not shown). The stability of the drugs in PAA nanoparticle was established by repeated washing with aqueous bovine calf serum through Amicon centrifugal filter units with a lOOKDa or larger cut off membrane and drug in the filtrate was measured spectrophotometrically. The comparative in vivo PDT efficacy of the ORMOSIL and PAA formulations, their tumor imaging potential and stability (in vitro release kinetics) is shown in Figs. 2-4, which clearly illustrate the advantages of PAA nanoparticles in reducing the therapeutic dose by almost 8- fold without diminishing the tumor-imaging potential and also avoiding the Tween-80 formulation required for the HPPH-CD conjugate 1. In the 2nd approach the HPPH CD conjugate 1 was post-loaded to PAA nanoparticles, which certainly enhanced the tumorimaging, but the therapeutic dose was still 10-fold higher (similar to the HPPH CD conjugate, Fig. 5B). In the 3rd approach the cyanine dye was conjugated peripherally to the PAA nanoparticles first and then HPPH was post loaded. Again, compared to HPPH-CD conjugate 1, the PAA formulation showed enhanced tumor-specificity (imaging) (Fig.5B).
Effect of nanoparticles on tumor selectivity
[0023] A photosensitizer (photosensitizer) with increased selectivity and longer wavelength could be a more suitable candidate for brain and deeply seated tumors (especially breast, brain and lung). The evolution of light sources and delivery systems is also critical to the progression of photodynamic therapy (PDT) in the medical field. Two different techniques: interstitial and intracavitary light delivery have been used for treatment of brain tumors. Powers using interstitial PDT on patients with recurrent brain tumors showed that the majority of patients had tumor recurrence within two months of treatment. However, it was later observed that treatment failures appeared to occur outside the region of the effective light treatment. Chang et al reported an effective radius of tumor cell kill in 22 glioma patients of 8 mm compared with the 1.5 cm depth of necrosis noted by Pierria with the intracavitary illumination method. It is believed that tumor resection is important so that the numbers of tumor cells remaining to treat are minimized. With stereotactic implantation of fibers for interstitial PDT there is no cavity to accommodate swelling and a considerable volume of necrotic tumor which causes cerebral edema. However, cerebral edema can be readily controlled with steroid therapy. Compared to chemotherapy and radiotherapy, patients with brain tumors treated with PDT have definitely shown long-term survival, whereas glioma patients treated with adjuvant chemotherapy or radiotherapy do not seem to show additional benefits. On the basis of our preliminary data, the ανβ3 targeted nanoparticles may improve tumor-selectivity and PDT outcome.
PET imaging and PDT: PAA nanoparticles decreased the liver uptake of the 1241- photosensitizer (PET imaging agent) and enhanced the tumor-specificity
[0024] Our initial investigation with an 1241-labeled photosensitizer 2 indicates its in vivo PDT efficacy and capability of detecting tumors l04-106 (RTF, Colon26, U87, GL261, pancreatic tumor xenograft) and tumor metastases (BALB/c mice bearing orthotopic 4T1 (breast tumors) (Fig 6). Interestingly, compared to 18F FDG photosensitizer 2 showed enhanced contrast in most of the tumors including those where 18F FDG-PET provides limited imaging potential (e.g., brain, lung and pancreatic tumors). See Fig. 7 for comparative biodistribution. This is the first report showing the utility of porphyrin-based compounds as a "BIFU CTIONAL AGENT" for imaging breast tumor and tumor metastasis. Similar to most nanoparticles, PAA nanoparticles accumulate in liver and spleen. Their clearance rate from most organs is significantly faster than Ormosil nanoparticle and they do not show long-term organ toxicity. Even tumor-avid porphyrinbased photosensitizer exhibit high uptake in liver and spleen, but are non-toxic until exposed to light. The photosensitizer clear from the system quickly (days) without organ toxicity. However, radioactive photosensitizer such as the 1241- labeled analog 2 (superior to 18F-FDG in PET-imaging of lung, brain, breast and pancreas tumors) with a T½ of 4.2 days could cause radiation damage to normal organs. Based on the observation of high uptake of PAA nanoparticles in liver and spleen (below) we postulated that saturating the organs with the non-toxic PAA nanoparticles before injecting the PET agent might reduce uptake and radiation damage by 1241- imaging agent. For proof-of principle blank PAA nanoparticles were first injected (i.v.) into mice bearing Colon26 tumors followed 24 h later by i.v. 1241-analog (100- 50μΟί). The mice were imaged at 24, 48 and 72h post injection and biodistribution studies were performed at each time point summarized in Figure 8A-8C (only 72h images shown).
[0025] The presence of PAA nanoparticles made a remarkable difference in tumor contrast with brain, lung and pancreatic tumors). See Fig. 7 for comparative biodistribution. PAA nanoparticles can be targeted to nucleolin with F3-Cys.
[0026] F3-targeted nanoparticles were prepared using two kinds of F3 peptides: F3 peptide conjugated to nanoparticle via one of the 8 lysines available in its sequence and F3- Cys peptide conjugated to nanoparticle via cysteine. Cysteine capped nanoparticles served as non-targeted control. Three 25 mg batches of each type of nanoparticle contained: 2.6, 5.1 and 7.7 mg F3, (A3-A5) respectively; 2.7, 5.3 and 8 mg F3-Cys (B3-B5) respectively, and 0.29, 0.58 and 0.87 mg Cys (C3-C5) respectively. The fluorescence intensity from PAA nanoparticle incubated in vitro with nucleolin positive MDA-MB-435 cells is shown in Fig. 9. The F3-Cys conjugated nanoparticles show considerably higher binding efficiency than non-targeted nanoparticles, while F3 conjugated nanoparticles do not. Conjugation via a cysteine link preserves the specificity of F3 peptide for nucleolin. In addition excess cysteine on the nanoparticles helphotosensitizer to minimize the non-specific binding. Additional experiments (not shown) suggested that the amount of F3-Cys peptide (5.3 mg/25mg nanoparticle) used for B4 nanoparticles was optimal.
[0027] Optical properties of post-loaded PAA nanoparticles. The absorption spectrum of PAA nanoparticles post-loaded with both HPPH and cyanine dye (even at 0.5 mg/ml), clearly shows characteristic signatures for both the photos ens itizer and dye, without aggregation-induced broadening, while the fluorescence spectrum shows strong signals from both components.
HPPH conjugated PAA nanoparticles with F3-Cys peptide at the outer surface show targeted specificity:
[0028] F3-mediated specificity is retained in the presence of conjugated HPPH. F3 targeted nanoparticles did targeted nanoparticles did not, indicating that F3 -mediated specificity is retained in the presence of conjugated HPPH. F3 targeted nanoparticles did not accumulate in the nucleus. On activation of cells with light at 660 nm only F3-targeted nanoparticle caused cell kill (Fig 11). Cell internalization of F3- targeted nanoparticles was confirmed by fluorescence confocal microscopy.
HPPH conjugated PAA nanoparticles with F3-Cyspeptide at the outer surface show targeted specificity:
[0029] The specificity of targeted nanoparticles was tested by fluorescent imaging
(Fig. 10). F3 targeted HPPH conjugated PAA nanoparticle specifically bound to MDA-MB- 435 cells (expressing nucleolin) while non-targeted nanoparticles did not, indicating that F3- mediated specificity is retained in the presence of conjugated HPPH. F3 targeted nanoparticles did not accumulate in the nucleus. On activation of cells with light at 660 nm only F3-targeted nanoparticle caused cell kill (Fig 11). Cell internalization of F3-targeted nanoparticles was confirmed by fluorescence confocal microscopy.
F3-Cys shows target-specificity in 9L glioma cells:
[0030] Similar to F3-cys, a pegylated form of F3-Cys PEG on PAA nanoparticles also showed remarkable target-specificity in 9L rat glioma cells which also expresses nucleolin, Fig 11. (Note: HPPH is replaced with a Rhodamine moiety).
Biodistribution studies: PAA nanoparticle Enhances tumor uptake of HPPH:
[0031] The biodistbiodistribution of 14C-HPPH and 14C-HPPH post-loaded PAA nanoparticle was performed in BALB/c mice bearing Colon26 tumors at 24, 48 and 72 h post injection (3 mice/time point) and the results are summarized in Fig. 12. As can be seen presence of PAA nanoparticles made a significant increase in tumor uptake with reduced uptake in other organs.
Size of PAA nanoparticles made remarkable difference in tumor-enhancement:
[0032] The biodistribution of 1241-photosensitizer was investigated using variable sizes of nanoparticles either injecting the nanoparticles first and then administrating the labeled photosensitizer or postloading the labeled photos ens itizer to PAA nanoparticles and then perform in vivo biodistribution in mice at 24, 48 and 72 h. The results summarized in Figure 13A-13C clearly indicate that the size of PAA nanoparticles makes a significant impact in tumor enhancement. Experiments related to in vivo PDT efficacy of these formulations are currently in progress.
[0033] This invention shows the utility of porphyrin-based compounds in a
"bifunctional agent" for imaging breast tumor and tumor metastasis. Similar to most nanoparticles, PAA nanoparticles accumulate in liver and spleen. Their clearance rate from most organs is significantly faster than Ormosil nanoparticles and they do not show long-term organ toxicity. Even tumor-avid porphyrin based photos ens itizers exhibit high uptake in liver and spleen, but are non-toxic until exposed to light. The photos ens itizers clear from the system quickly (days) without organ toxicity. However, radioactive photos ens itizers such as the 1241-labeled analog 2 (superior to 18F-FDG in PET-imaging of lung, brain, breast and pancreas tumors) with a T½ of 4.2 days could cause radiation damage to normal organs. Based on the observation of high uptake of PAA nanoparticles in liver and spleen (below) we postulated that saturating the organs with the non-toxic PAA nanoparticles before injecting the PET agent might reduce uptake and radiation damage by 1241- imaging agent. For proof- of principle blank PAA nanoparticles were first injected (i.v.) into mice bearing Colon26 tumors followed 24 h later by i.v. 1241-analog (100-150μΟί). The mice were imaged at 24, 48 and 72h post injection and biodistribution studies were performed at each time point summarized in Figure 8A-8C (only 72h images shown).
[0034] The presence of PAA nanoparticles makes a remarkable difference in tumor contrast with significantly reduced uptake in spleen and liver and improved tumor- uptake/contrast at 24, 48 and 72 h post injection (3 mice/group Similar studies (tumor- imaging and PDT efficacy) in which the labeled photosensitizer is post-loaded to variable sizes. Similar studies (tumor-imaging and PDT efficacy) in which the labeled photosensitizer is post-loaded to variable sizes PAA nanoparticles are currently in progress.
[0035] In more detail, the development of effective multifunctional agents for imaging and PDT by following two approaches is being pursued. In the first approach, 3-( - hexyloxyethyl) pyropheophorbide-a [HPPH, a chlorophyll-a derivative (660 nm) currently in Phase I/II clinical trials] was used as a vehicle in delivering the desired fluorescence and nuclear imaging (PET) agents to tumors The second approach was focused on selecting a biocompatible/biodegradable non-toxic nanoplatform, choosing between organically modified silica (ORMOSIL) and polyacrylamide (PAA) nano-platforms for image-guided therapy. In both approaches, HPPH (660 nm) was used as a photos ens itizer (PS) and the detailed investigations gave promising results. Among the NPs investigated, the PAA nanoplatform was given preference, due to its biocompatibility and limited toxicity. HPPH may be replaced with 800 nm PDT agents and their biological efficacy compared with and without the nanoparticle formulation. This study is aimed to select the "ideal" candidate for image-guided PDT of brain tumors, with and without surgery.
[0036] It is well established that photon migration into tumor tissue is highly dependent on the absorption and scattering properties of tissue components. The near- infrared (MR) region of the spectrum offers certain advantages for photon penetration. Therefore, efforts are currently underway in various laboratories (including ours) to develop PDT agents with long wavelength absorption, near 800 nm. The availability of inexpensive light emitting diodes (LEDs) in this region will also make PDT more applicable and economical.
[0037] Initial results with PAA NPs indicate that multifunctional nanoparticles incorporating both phototherapeutic and imaging agents are uniquely promising in delivering high payloads of these agents to tumors. The success of combined PDT and imaging relies on the development of tumor-avid molecules (EPR effect or target-specific) that are preferentially retained in malignant cells but cleared from surrounding cells and normal tissues.
[0038] On the basis of preliminary data, we extrapolated that a PAA nanoplatform containing both NIR therapeutic (800 nm) and imaging agents as separate monomers will help to treat brain cancer patients in combination of Surgery and PDT via image-guided therapy. Tumor-specificity can further be enhanced by targeting the neovasulature and/or av 3 integrin, known for its over-expression in highly metastatic brain tumors. This study was divided in following two aims:
[0039] Aim 1 : To investigate the utility of PAA nanoparticles in developing multifunctional platforms by (i) post-loading both the monomeric therapeutic and imaging agents, and (ii) conjugating the fluorescence agent at the periphery of the NPs and post- loading the PET/PDT agent.
[0040] Aim 2: Conjugate cRGD to the best candidate selected from each series (Aims
1 & 2) and compare their target-specificity/pharmacokinetics and PDT efficacy in U87 (amp3 +), GL261 ((av 3 low) and Α431(ανβ3 -) tumors. Specificity, we prepared FITC conjugated PAA NPs and then attached cRGDf (an ayp3 ligand), see structure in Figure 17B, on the surface of the NPs. To test if the surface-modified NPs selectively target tumor tissue, as compared to the unmodified NPs, we treated MDA-435 cells (ay 3 expressing) with both types of NPs (20 min incubation at 0.1 mg/mL). The RGD-conjugated NPs specifically bound to MDA-435 cells but not the unmodified NPs [Figure 17B and 17C, respectively] . These results indicate that cRGD modified PAA NPs may enable the targeted delivery of incorporated image contrast agents or drugs for imaging and therapy. Specifically, Figure 17A shows structure of cRGDfK. Figure 17B shows a confocal fluorescence image of MDA- MB-435 cell treated with cRGD-conjugated FITC PAA NP's and Figure 17C shows fluorescence image of MDA-MB-435 cells treated with unmodified FITC PAA NP's. Recent results obtained show great potential of combined PS (660 nm) and CD either in a form of conjugates or post-loaded to biocompatible PAA NPs, for tumor-imaging (Fig.18 , λβΧο:783 nm, Xem 865 nm) and PDT. This approach can be extended to develop brain cancer target- specific multifunction agents (800 nm agents) for image-guided (PET/ fluorescence) PDT. The approach is illustrated in Chart- 1 Fig 21. The best agent(s) selected from this study can be translated in the future to cancer patients by following an approach shown in Chart 2, Figure 22. [0041] We have developed a novel post-loading approach for constructing a multifunctional biodegradable PAA nanoplatform for tumor-imaging and PDT. This approach provides an opportunity to post-load both imaging and therapeutic agents at desired concentrations. HPPH (a chlorophyll-a derivative, developed in our laboratory and in Phase VII human clinical trials) was used as a substrate. We have also shown that, compared to free
124 124
I-PS, the I-PS post-loaded in PAA NPs shows enhanced tumor uptake/tumor imaging and reduced uptake in other organs (especially spleen and liver). We also confirmed the tumor-avidity of PAA NPs by covalently conjugating with a non-tumor avid fluorophore Compound 2, Figure 19B. Finally, one of the main concerns of using nanoparticles has been their unknown toxicity in vivo. We have evaluated the toxicity of PAA NPs at variable doses and even at a high dose no normal organ toxicity was observed in BALB/c mice. On the basis of our exciting findings we hypothesize that extending these approaches by using near 800 nm PS will produce improved PAA nanoplatforms for imaging brain and other tumors. Various approaches for the preparation of multifunctional NP's are shown in Figures 20A- 20D.
[0042] Conjugation of NIR fluorophore at the periphery of PAA NPs drastically enhances its imaging capability: In this approach to investigate the utility of PAA NPs in tumor-specificity, a cyanine dye (CD) containing a carboxylic acid group (Fig. 19B), which did not show any tumor uptake by itself (C in Fig. 19A), was conjugated at the periphery of the PAA NPs (Figure 19B containing amino functionalities) and the imaging potential of all three preparations was investigated in BALB/c mice bearing Colon26 tumors. The CD-PAA conjugate 2, in Figure 19A at B, showed the best tumor localization at both 24 and 48 h post injection B in Fig. 19A (only 48 h images are shown). In particular, Figure 19A shows a control at A, cyanine dye-COOH conjugated to PAA nanoparticles (conjugate 2 48 h post injection) at B and free cyanine dye-COOH (48 h post-injection) at C. Dosages for B and C were 0.3 μηιο1ε/1¾. Figure 19B shows structures for cyanine dye and cyanine dye- nanoparticle conjugate.

Claims

What is claimed is:
1. A composition comprising PAA nanoparticles containing a tetrapyrollic photosensitizer post loaded to the PAA nanoparticle after nanoparticle formation and an imaging agent covalently bonded to the PAA nanoparticle and a tumor targeting moiety covalently bonded to the PAA nanoparticles.
2. The composition of claim 1 where the PAA is a polyacrylamide nanoparticle.
3. The composition of claim 2 wherein the tetrapyrollic photosensitizer has the structural formula:
Figure imgf000028_0001
or a pharmaceutically acceptable derivative thereof, wherein:
Ri and R2 are each independently substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, -C(0)Ra or -COORa or -CH(CH3)(ORa) or -CH(CH3)(0(CH2)nXRa) where Ra is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, or substituted or unsubstituted cycloalkyl; where R2 may be -CH=CH2, -CH(OR20)CH3, -C(0)Me, -C(=NR2i)CH3 or -CH(NHR21)CH3
where X is an aryl or heteroaryl group;
n is an integer of 0 to 6; where R20 is methyl, butyl, heptyl, docecyl or 3,5-bis(trifluoromethyl)-benzyl; and R21 is 3,5,-bis(trifluoromethyl)benzyl;
Ria and R2a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;
R3 and R4 are each independently hydrogen or substituted or unsubstituted alkyl;
R3a and R4a are each independently hydrogen or substituted or unsubstituted alkyl, or together form a covalent bond;
R5 is hydrogen or substituted or unsubstituted alkyl;
R6 and R6a are each independently hydrogen or substituted or unsubstituted alkyl, or together form =0;
R7 is a covalent bond, alkylene, azaalkyl, or azaaraalkyl or
Figure imgf000029_0001
where R20 is 3,5- bis(tri-fluoromethyl)benzyl or -CH2X-R1 or -YR1 where Y is an aryl or heteroaryl group;
Rs and Rsa are each independently hydrogen or substituted or unsubstituted alkyl or together form =0;
R9 and Rio are each independently hydrogen, or substituted or unsubstituted alkyl and
R9 may be -CH2CH2COOR2 where R2 is an alkyl group that may optionally substituted with one or more fluorine atoms;
each of R1-R10, when substituted, is substituted with one or more substituents each independently selected from Q, where Q is alkyl, haloalkyl, halo, photosensitizereudohalo, or -COORb where R, is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, araalkyl, or ORc where Rc is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl or CONRjRe where Rd and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or NRfRg where Rf and Rg are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or =NRh where Rh is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or is an amino acid residue; each Q is independently unsubstituted or is substituted with one or more substituents each independently selected from Qi, where Qi is alkyl, haloalkyl, halo, photosensitizereudohalo, or -COORb where Rb is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, araalkyl, or ORc where Rc is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl or CO RaRe where R<j and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or NRfRg where Rf and Rg are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or =NRh where Rh is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or is an amino acid residue.
4. The composition of claim 2 wherein the imaging agent is a cyanine dye.
5. The composition of claim 4 wherein the imaging agent is a 124I labeled compound.
6. The composition of claim 4 wherein the imaging agent is a PET, fluorescence or MR imaging agent.
7. The composition of claim 4 wherein the targeting moiety is a peptide, folic acid or a carbohydrate.
8. A method for making PAA nanoparticles containing a photosensitizer and an imaging agent by covalently bonding a fluorophore onto a preprepared PAA nanoparticle and then post loading a photosensitizer onto the pre-prepared PAA nanoparticle.
9. The method of claim 8 where the PAA nanoparticle is a polyacrylamide nanoparticle.
10. The method of claim 9 where the photosensitizer is HPPH.
11. The composition of claim 2 where the photosensitizer is HPPH.
12. The composition of claim 2 where the numerical ratio of postloaded photosensitizer to imaging agent is from 1 to 1 to 10 to 1.
13. The composition of claim 12 where the numerical ratio of postloaded photosensitizer moieties to imaging agent is from 2 to 1 to 4 to 1.
14. The composition of claim 2 where the photosensitizer is a HPPH, purpurinimide having an absorbance between 680 and 720 nm, bacteriopurpurinimde having an absorbance between 780 and 800 nm or mixtures thereof.
15. The nanoparticle of claim 2 where the tetrapyrrolic photosensitizer has an emission between 700 and 800 nm.
16. The nanoparticle of claim 2 where the tumor targeting agent is an RGD peptide.
17. The composition of claim 2 wherein the photosensitizer is HPPH, a purpurinimide having an absorbance between 680 and 720 nm, bacteriopurpurinimde having an absorbance between 780 and 800 nm or mixtures thereof.
18. The composition of claim 2 wherein the targeted moiety is cRGD or F3 or F3- targeted (A series), F3-Cys targeted (B series), peptide, folic acid or a carbohydrate.
19. A method for imaging and treatment of hyperproliferative tissue in an animal comprising:
a) injecting a composition according to claim 2 in an amount of 0.05 to 1.0 μπιο^^, b) imaging the animal utilizing the covalently bonded imaging agent to define and locate the hyperproliferative tissue, and
c) treating the defined and located hyperproliferative tissue with photodynamic therapy.
20. A method for imaging and treatment of hyperproliferative tissue in an animal comprising:
a) injecting a composition according to claim 6 in an amount of 0.05 to 1.0 μπιο^^, b) imaging the animal utilizing the covalently bonded imaging agent to define and locate the hyperproliferative tissue, and
c) treating the defined and located hyperproliferative tissue with photodynamic therapy.
21. A method for imaging and treatment of hyperproliferative tissue in an animal comprising:
a) injecting a composition according to claim 16 in an amount of 0.05 to 1.0 μιηοΐ68/1¾ b) imaging the animal utilizing the covalently bonded imaging agent to define and locate the hyperproliferative tissue, and
c) treating the defined and located hyperproliferative tissue with photodynamic therapy.
PCT/US2013/053350 2012-08-03 2013-08-02 Paa nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded WO2014022742A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13/566,444 US8906343B2 (en) 2009-10-21 2012-08-03 PAA nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded
US13/566,444 2012-08-03

Publications (1)

Publication Number Publication Date
WO2014022742A1 true WO2014022742A1 (en) 2014-02-06

Family

ID=50028552

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/053350 WO2014022742A1 (en) 2012-08-03 2013-08-02 Paa nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded

Country Status (1)

Country Link
WO (1) WO2014022742A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050196343A1 (en) * 2004-02-27 2005-09-08 Molecular Therapeutics, Inc. Degradable nanoparticles
US20090043090A1 (en) * 2004-07-16 2009-02-12 Health Research, Inc. Adduct of fluorescent dye and tumor avid tetrapyrrole
WO2009105209A1 (en) * 2008-02-19 2009-08-27 Health Research, Inc. Silica nanoparticles postloaded with photosensitizers for drug delivery in photodynamic therapy
US20110091373A1 (en) * 2009-10-21 2011-04-21 Health Research, Inc. Paa nanoparticles for enhancement of tumor imaging

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050196343A1 (en) * 2004-02-27 2005-09-08 Molecular Therapeutics, Inc. Degradable nanoparticles
US20090043090A1 (en) * 2004-07-16 2009-02-12 Health Research, Inc. Adduct of fluorescent dye and tumor avid tetrapyrrole
WO2009105209A1 (en) * 2008-02-19 2009-08-27 Health Research, Inc. Silica nanoparticles postloaded with photosensitizers for drug delivery in photodynamic therapy
US20110091373A1 (en) * 2009-10-21 2011-04-21 Health Research, Inc. Paa nanoparticles for enhancement of tumor imaging

Similar Documents

Publication Publication Date Title
EP2437653B1 (en) Paa nanoparticles for enhancement of tumor imaging
Sarbadhikary et al. Recent advances in photosensitizers as multifunctional theranostic agents for imaging-guided photodynamic therapy of cancer
Son et al. Light-responsive nanomedicine for biophotonic imaging and targeted therapy
Lin et al. Nano-photosensitizers for enhanced photodynamic therapy
Rai et al. Development and applications of photo-triggered theranostic agents
Gupta et al. Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles
Wang et al. Advances in Prostate‐Specific Membrane Antigen (PSMA)‐Targeted Phototheranostics of Prostate Cancer
Yin et al. Phototheranostics for multifunctional treatment of cancer with fluorescence imaging
US20110288234A1 (en) Silica nanoparticles postloaded with photosensitizers for drug delivery in photodynamic therapy
Sandland et al. Porphyrins and related macrocycles: Combining photosensitization with radio-or optical-imaging for next generation theranostic agents
Battogtokh et al. Active-targeted pH-responsive albumin–photosensitizer conjugate nanoparticles as theranostic agents
JF Calvete et al. Tetrapyrrolic macrocycles: Potentialities in medical imaging technologies
US9045488B2 (en) PAA nanoparticles for tumor treatment and imaging
US10568963B2 (en) Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles
US8906343B2 (en) PAA nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded
Wang et al. Theranostics with photodynamic therapy for personalized medicine: to see and to treat
Pratihar et al. Small molecules and conjugates as theranostic agents
Liu et al. Porphyrins for imaging, photodynamic therapy, and photothermal therapy
Grebinyk et al. Molecular and nanoparticulate agents for photodynamic therapy guided by near infrared imaging
Lim et al. Recent advances in bioimaging for cancer research
WILSON Photonic and non-photonic based nanoparticles in cancer imaging and therapeutics
WO2014022742A1 (en) Paa nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded
Abbas et al. Organic Nanomaterials for Cancer Phototheranostics
Dey et al. Challenges, conclusions, and future aspects of photodynamic therapy
US20130202526A1 (en) Paa nanoparticles for pet imaging and pdt treatment

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13825781

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13825781

Country of ref document: EP

Kind code of ref document: A1