WO2014022740A1 - Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles - Google Patents

Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles Download PDF

Info

Publication number
WO2014022740A1
WO2014022740A1 PCT/US2013/053347 US2013053347W WO2014022740A1 WO 2014022740 A1 WO2014022740 A1 WO 2014022740A1 US 2013053347 W US2013053347 W US 2013053347W WO 2014022740 A1 WO2014022740 A1 WO 2014022740A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
substituted
aryl
alkenyl
alkynyl
Prior art date
Application number
PCT/US2013/053347
Other languages
French (fr)
Inventor
Ravindra K. Pandey
Raoul Kopelman
Anurag Gupta
Munawwar Sajjad
Original Assignee
Health Research, Inc.
The Research Foundation Of State Universityof New York
Regents Of The University Of Michigan Office Of Technology Transfer
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 US13/566,411 external-priority patent/US10568963B2/en
Application filed by Health Research, Inc., The Research Foundation Of State Universityof New York, Regents Of The University Of Michigan Office Of Technology Transfer filed Critical Health Research, Inc.
Publication of WO2014022740A1 publication Critical patent/WO2014022740A1/en

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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/58[b]- or [c]-condensed
    • C07D209/60Naphtho [b] pyrroles; Hydrogenated naphtho [b] pyrroles

Definitions

  • 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.
  • 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.
  • endogenous molecules e. g. hemoglobin
  • Fluorescence imaging is a non-invasive and non-ionizing imaging technique that requires only nanomoles of fluorophores for contrast enhancement.
  • the NIR spectral range (-650 - 950 nm) is known as the "biological window" for optical imaging since light absorption due to water, deoxygenated hemoglobin and oxygenated hemoglobin is minimized in this region, as well as tissue autofluorescence and light scattering.
  • Fluorescence the emission 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.
  • HPPH and the iodobenzyl pheophorbide-a have plasma half lives ⁇ 25 h.
  • the long radiological half life of 124 I 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.
  • 124 I complex decay scheme which results in only 25% abundance of positron (compared with 100% positron emission of 18F)
  • in vivo quantitative imaging with 124 I labeled antibodies has been successfully carried out under realistic conditions using a PET/CT scanner A variety of biomolecules have been labeled with 124 !
  • Photodynamic therapy a relatively new modality for the treatment of a variety of oncological, cardiovascular, dermatological and ophthalmic diseases, is based on the preferential localization of photosensitizing molecules, (photosensitizers, PS) in target tissues.
  • the PS Upon light activation, the PS produces reactive singlet oxygen 5 which damages tumor cells and neovasculature, and also initiates antitumor inflammatory and immune responses.
  • We and others have developed relatively tumor-avid PS which selectively accumulate in tumor, and these molecules have been used to carry optical, PET and MR imaging agents to the tumor sites.
  • the tumor selectivity of current PS is not always adequate. Approaches that link PS to antibody fragments or receptor ligands have been disappointing because the number of required PS/cell generally is greater than the number of antigen or receptor binding sites. Conversely, the imaging agent carrying capacity of the individual PS molecules is limited.
  • HPPH developed in our laboratory and currently under Phase VII clinical trials, when conjugated with certain cyanine dyes can be used for both fluorescence imaging and photodynamic therapy.
  • the conjugate showed potential tumor imaging and PDT efficacy, but compared to the imaging dose the required therapeutic dose was 8-fold higher.
  • Increasing the number of HPPH moieties in synthetic photosensitizer-cyanine dye (PS-CD) conjugates did not minimize the therapeutic dose.
  • 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 nanoparticles 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.
  • nanoparticles are uniquely promising in that (i) their hydrophilicity and charge can be altered; (ii) they possess enormous surface area which can be modified with functional groups possessing a diverse array of chemical and biochemical properties, including tumor-selective ligands; (iii) owing to their sub-cellular and sub-micron size, they can penetrate deep into tissues and are generally taken up efficiently by cells.
  • the present invention relates to PAA nanoparticles (polyacrylic acid and its modified carboxy derivatives, e.g. polyacrylamide) containing a photosensitizer and an imaging enhancing agent.
  • PAA nanoparticles polyacrylic acid and its modified carboxy derivatives, 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
  • polyacrylamide 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 tetrapyrollic photosensitizer having the structural formula:
  • n is an integer of 0 to 6;
  • R2 0 is methyl, butyl, heptyl, docecyl or 3,5-bis(trifluoromethyl)-benzyl; and R21 is 3,5,-bis(trifluoromethyl)benzyl;
  • Ria and R2 a 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
  • R7 is a covalent bond, alkylene, azaalkyl, or azaaraalkyl or where R2 0 is 3,5- bis(tri-fluoromethyl)benzyl or -CH2X-R 1 or -YR 1 where Y is an aryl or heteroaryl group;
  • 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.
  • a formulation containing the photosensitizer, HPPH [3-(l '-hexyloxyethyl)pyropheophorbide- a], and the cyanine dye in a ratio of 2: 1 minimized the undesirable quenching of the HPPH electronic excitation energy due to energy migration within the nanoparticles and/or F5rster (fluorescence) resonance energy transfer (FRET) between HPPH and cyanine dye.
  • FRET fluorescence resonance energy transfer
  • 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 RTF 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 ⁇ ).
  • 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 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 ⁇ /kg in PAA NPs and 0.78 ⁇ /kg in ORMOSIL NPs.
  • FIG. 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.
  • 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.
  • 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 14C-labeled HPPH, and 14C-labeled HPPH post- loaded into PAA NPs in BALB/c mice bearing Colon26 tumors.
  • 14C-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
  • 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 structural formulas for HPPH at 1, IR820 cyanine dye (CD) at 2 and cyanine dye at 3 where the chloro group of IR820 is replaced with p-aminothiol.
  • Figure 17B illustrates various polyacrylamide acid (PAA) nanoparticles postloaded with HPPH (1), cyanine dye (2) and cyanine dye (3) alone and in different combinations and ratios.
  • PAA polyacrylamide acid
  • Figure 18A shows whole body fluorescence images of BALB/c mice bearing Colon- 26 tumors.
  • Control mouse A, E, I, and M
  • cyanine dye 2 B-D
  • nanoconstruct 4 F-H
  • cyanine dye 3 J-L
  • nanoconstruct 5 N-P
  • Figure 18B shows fluorescence intensity values for left, cyanine dye 2 and construct 4 and right, cyanine dye 3 and construct 5.
  • Figure 19 shows whole body fluorescence images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C & D), 8 (E & F), 9 (G & H) and 10 (I & J) in BALB/c mice bearing Colon-26 tumors.
  • A-J the excitation wavelength was 782 nm.
  • Images A, B, C, E, G, & I and D, F, H, & J were taken 24 and 48 hours post i.v. injection, respectively.
  • Figure 20 shows whole body FRET images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C), 8 (D), 9 (E) and 10 (F) in BALB/c mice bearing Colon-26 tumors.
  • A-F the excitation wavelength was 665 nm.
  • Images A-F were taken 24 hours post i.v. injection.
  • Figure 21 A shows an absorbance curve for constructs 5, 6, 7, and 8 at various wave lengths.
  • Figure 2 IB shows fluorescent intensity for constructs 5, 6, 7, and 8 at 532 nm excitation.
  • Figure 21C shows fluorescent intensity for nanoconstructs 5, 7, and 8. Fluorescence was more intense for nanoconstructs 7 and 8 than for pure CD nanoconstruct 5, even if difference in absorption at 785 nm was minimal.
  • Figure 2 ID shows detector response for constructs 5, 6, 7, and 8 illustrating singlet oxygen decay rates.
  • Figure 22 A shows tumor response versus time for untreated control, PS1 (HPPH), and nanoconstructs 6, 7, and 8.
  • Figure 22B shows tumor response versus time for untreated control, nanoformulation
  • Fig. 23A shows an SEM for nanoconstruct 7 (representative of all groups).
  • Fig. 23B shows a DLS particle size distribution for Blank PAA NPs.
  • the mean diameter is 33.5 nm.
  • Fig. 23C shows a particle size distribution for nanoconstruct 6.
  • the mean diameter is 32.5 nm
  • Fig. 23D shows a particle size distribution for nanoconstruct 5.
  • the mean diameter is 35.2 nm.
  • Figures 24A-24L show Release/Retention Profiles of PS 1 (24A, 24C, 24E, 24G, & 241) and/or cyanine dye 3 (24B, 24D, 24F, 24H, & 24J) from nanoconstructs 5, 6, 7, 8, 9, and
  • Photosensitizers generally fluorescence 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, (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 photosensitizer 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.
  • biodegradable polymer based nanoparticles avoids multi-step synthesis and has numerous advantages including the ability to create water soluble formulations with desired pharmacokinetic properties, capable of delivering a high payload of the multiple agents (therapeutic PS and imaging agents) to tumors, increased photostability of photoactive agents and fluorophores, and the ability to modify the surface of the NP for conjugation to a variety of biomolecules.
  • NPs and other macromolecular objects can passively target the tumor interstitium, via the "Enhanced Permeability and Retention" (EPR) effect due to the leaky vascular system in tumors. 21a ' b
  • EPR Enhanced Permeability and Retention
  • the poor lymphatic drainage system in tumors causes fluid retention in the tumor interstitial space, which helps to retain polymeric nanoparticles and other macromolecular objects in the tumor compared to normal tissue. 21a ' b
  • NPs are a promising means for delivering therapeutic and other molecular agents to tumors.
  • NPs could deliver a high payload of the drug to tumor
  • a PAA-based nanoconstructs for delivering both the near-infrared (NIR) cyanine dye (CD) fluorophore and the red-light absorbing photosensitizer HPPH.
  • the release of the desired imaging and therapeutic agents may also be controlled by creating a nanoparticle that is pH or temperature sensitive, or by modifying the pores of the NP matrix
  • a parallel study we encapsulated the PS within polymeric NPs, but the retention efficiency was low, therefore a large concentration of NPs was required to achieve the desired therapeutic dose.
  • 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 tumor imaging, 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).
  • a 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.
  • PDT photodynamic therapy
  • 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.
  • PET imaging agent 1241-photosensitizer
  • Our initial investigation with an 1241-labeled photosensitizer 2 indicates its in vivo PDT efficacy and capability of detecting tumorsl04-106 (RIF, Colon26, U87, GL261, pancreatic tumor xenograft)) and tumor metastases (BALB/c mice bearing orthotopic 4T1 (breast) tumors) (Fig 6).
  • tumor-avid porphyrinbased photosensitizer exhibit high uptake in liver and spleen, but are non-toxic until exposed to light. The photosensitizer clears from the system quickly (days) without organ toxicity.
  • 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: [0035] 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:
  • F3-Cys shows target-specificity in 9L glioma cells:
  • F3-cys 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).
  • 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 nanoparticle 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 porphyrin based 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.
  • 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.
  • mice were imaged at 24, 48 and 72h post injection and biodistribution studies were performed at each time point summarized in Figurea 8A-8C (only 72h images shown).
  • 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.
  • Human Serum Albumin, Tween-80 and dimethyl sulfoxide (DMSO) were purchased from Sigma-Aldrich.
  • Bovine Calf Serum (BCS) was purchased and dPBS (pH 7.4, lx, without calcium and magnesium) were purchased from Cellgro.
  • Ethanol (200 proof) was purchased from Pharmco-Aaper. All solutions were prepared with 18 ⁇ water purified by a Millipore Milli-Q Advantage A 10 water purification system. 30 and 100 kDa Amicon Ultra- 15 and Ultra 4 centrifuge filters were purchased from Fisher Scientific.
  • PAA nanoparticles were prepared by following our previous with slight modifications (See e.g. Multifunctional ORMOSIL and PAA nanoparticles, Gupta et al, Photodynamic Therapy: Back to the Future. Edited by Kessel, David FL. Proceedings of the SPIE, Volume 7380 (2009)., pp. 73805H- 73805H-12 (2009). [0046] Post-Loading of the Photosensitizer 1, and the cyanine dyes 2, and 3 to blank
  • AFPAA to create nanoconstructs 4, 5, 6, 9, and 10 In brief, 10 mg of lyophilized PAA NPs were suspended in 1 mL of 1% Tween-80 / water solution and to this solution 10 ⁇ , of 1, 2, or 3 (20 mM in DMSO) is added and magnetically stirred at a constant rpm for 2 hours. The NPs were centrifuge filtered in a 30 kDa Amicon Ultra- 15 centrifuge filter for 30 minutes at 5,000 RPM and then the NPs were reconstituted with water. The nanoparticles were syringe filtered with a 0.2 ⁇ regenerated cellulose syringe filter. Nano formulation 9 and 10 were created by mixing nanoconstruct 1 and 3 such that the molar ratio of 1 to 3 was 2: 1 and 4: 1, respectively. The NPs are stored at 4° C until further use. For details see "Supporting Material Information”.
  • the NPs from all formulations were suspended in a 1% human serum albumin (HSA) - water solution and immediately the absorbance value for the HSA/nanoconstruct solution was measured spectrophotometrically.
  • HSA human serum albumin
  • the NP solution is centrifuge filtered in a 100 kDa Amicon ultra-4 centrifugation filter for 20 minutes at 4,000 RPM. The absorbance of the PS or fluorophore in the filtrate was spectrophotometically measured (filtrate 1).
  • the NPs in the retentate were reconstituted to the original volume with 1% HSA and re-centrifuge filtered (filtrate 2) and measured spectrophotometrically. The amount of 1 and/or 3 retained by the NP was confirmed by measuring the absorbance of the retentate upon reconstitution to the original volume with 1% HSA. If the sum total of all filtrates and the retentate is less than 90% of the stock value for either chromophore then ethanol is added to the centrifuge filter to measure what had adsorbed to the filter. These measurements were taken immediately post-addition of the nanoconstructs in a 1% HSA solution, 4 and 24 hours post addition of the nanoconstructs in the 1% HSA solution.
  • Optical Imaging Setup The fluorescence imaging was conducted in accordance with a protocol approved by the Institutional Animal Care and Use Committee IACUC at Roswell Park Cancer Institute and the Guide for the Use of Laboratory Animals.
  • BALB/c mice (3 mice/group) bearing subcutaneous Colon 26 tumors on the right shoulder were injected i. v. (tail-vein) with either cyanine dyes or nanoconstructs/formulations.
  • cyanine dyes or nanoconstructs/formulations For a detailed description of the groups of mice imaged along with the dose, see the "Supporting Material Information".
  • a SPEX 270M spectrometer (Jobin Yvon) was used for acquisition of fluorescence emission spectra in the far red and MR spectral ranges, utilizing the first output port equipped with an InGaAs photodetector (Electrooptical Systems Inc., USA),.
  • a diode- pumped solid-state laser (Verdi, Coherent) at 532 nm was the excitation source. Generation of singlet oxygen O2) was detected by its phosphorescence emission peaked at 1270 nm.
  • the decays of this emission were acquired using the Infinium oscilloscope (Hewlett-Packard) coupled to the output of the Hamamatsu IR-PMT which is attached to the second output port of the SPEX 270M spectrometer.
  • Nanoconstructs 5 - 8 in polystyrene cuvettes were placed in front of the entrance to the spectrometer.
  • the emission signal was collected at 90-degrees relative to the exciting laser beam with the use of additional long-pass filters (a 950LP filter and/or a 538AELP filter) to attenuate the scattered light and fluorescence from the samples.
  • a second harmonic (532 nm) from the nanosecond pulsed Nd:YAG laser (Lotis Til, Belarus) operating at 20 Hz was used as the excitation source for time-resolved measurements.
  • mice Eight- to twelve-week-old BALB/cAnNCr mice (Jackson Laboratory, Bar Harbor, ME) were inoculated subcutaneously (s.c.) with lxlO 6 Colon 26 cells. When tumors reached 40-70 mm 3 , mice were injected i.v. (tail vein) with PS 1 (formulated in 1% Tween 80/D5W) or PAA nanoconstructs/formulations 6-10 suspended in water and further diluted in D5W. 24 hours post i.v.
  • PS 1 formulated in 1% Tween 80/D5W
  • PAA nanoconstructs/formulations 6-10 suspended in water and further diluted in D5W. 24 hours post i.v.
  • mice (BALB/c mice bearing Colon 26 tumors, 10 mice/group) were restrained in plexiglass holders and tumors were irradiated at 665 nm with a fluence and fluence rate of 135 J/cm 2 at 75 mW/cm 2 , respectively, using a pumped argon-dye laser.
  • the growth of tumors was measured two to three times per week and the mice were monitored for a total of 60 days post PDT treatment. When the tumor regrowth was >400 mm 3 , the mice were euthanized according to the guidelines of the institute approved animal protocol.
  • FIGS 17A and 17B Post-loading of amine functionalized PAA NPs with IR-820, cyanine dye 2, nanoconstruct 4; cyanine dye 3, nanoconstruct 5; HPPH, photosensitizer 1 : nanoconstruct 6; photosensitizer 1 and cyanine dye 3 at a 2: 1 molar ratio, nanoconstruct 7; and HPPH 1 and cyanine dye 3 at a 4: 1 molar ratio, nanoconstruct 8.
  • Nanoformulations 9 and 10 are nanoconstruct 6 and 5 mixed such that the molar ratio of 1 to 3 is 2: 1 and 4: 1, respectively.
  • DLS dynamic light scattering
  • SEM scanning electron microscopy
  • porous nanoparticles are advantageous since release of PS from the
  • NP is not required for the singlet oxygen to diffuse into the tumor cells. However, if the release profile is rapid the NP may not be able to efficiently deliver a high payload of the desired agent to tumor. Therefore, we investigated the release profiles of the photosensitizer 1 and the cyanine dye 3 from nanoconstructs 5-10, respectively, by incubating them in 1% human serum albumin (HSA) at variable time points. The release profiles are summarized in the supplemental section. The release of PS 1 in nanoconstruct 6 showed a two-phase release, where an increase in the release was seen in the first four hours, which subsequently decreased during the following 20 hours.
  • HSA human serum albumin
  • nanoconstruct 6 Compared to nanoconstructs 7-10, nanoconstruct 6 showed the highest retention of PS 1 (HPPH) over a 24 hour time period with approximately 87% being retained. When comparing the percentage of PS 1 retained (at the initial time point, time zero), upon addition of 1% HSA, the nanoconstruct 8 and 10 showed the highest retention ( ⁇ 84 %) of the PS.
  • the nanoformulation 7 which provided the best whole-body fluorescence imaging and PDT response, was also subjected for the release of both the chromophores in 25% bovine calf serum (BCS, 37°C) at 4, 8, 12, and 24 hours post-addition.
  • FIG. 18A shows whole body fluorescence images of BALB/c mice bearing Colon-26 tumors.
  • Control mouse A, E, I, and M
  • cyanine dye 2 B-D
  • nanoconstruct 4 F- H
  • cyanine dye 3 J-L
  • nanoconstruct 5 N-P
  • the fluorescence intensity values shown in Figure 18B are background subtracted from the control mouse (left, cyanine dye 2 and construct 4 and right, cyanine dye 3 and construct 5),
  • the modified version 3 showed higher uptake and improved tumor-imaging ability ( Figure 18A, J-L).
  • Figure 18A at N-P the difference in intensity in the tumor for 5 being statistically higher (p ⁇ 0.05) at 24 hours post-injection.
  • nanoconstructs 7 and 8 in which the PS and CD were post-loaded in a ratio of 2 to 1 and 4 to 1 on excitation of the cyanine dye at 782 nm gave fluorescence at 866 and 870 nm respectively.
  • both nanoconstructs on in vivo excitation at 665 nm produced a significant fluorescence beyond 860 nm, which can be explained by the phenomenon known as the F5rster (Fluorescence) Resonance Energy Transfer (FRET), or by the more general phenomena of energy migration, or excitation percolation, followed by energy trapping, analogous to the energy transport and funneling process in photosynthetic antenna.
  • FRET Fluorescence Resonance Energy Transfer
  • Figure 19 shows whole body fluorescence images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C & D), 8 (E & F), 9 (G & H) and 10 (I & J) in BALB/c mice bearing Colon-26 tumors.
  • the excitation wavelength was 782 nm.
  • Images A, B, C, E, G, & I and D, F, H, & J were taken 24 and 48 hours post i.v. injection, respectively and
  • Figure 20 shows whole body FRET images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C), 8 (D), 9 (E) and 10 (F) in BALB/c mice bearing Colon-26 tumors.
  • the excitation wavelength was 665 nm. Images A-F were taken 24 hours post i.v. injection.
  • the concentration for the cyanine dye was kept constant in all nanoconstructs and the concentration of 1 for nanoconstruct 6 and 8 was kept the same and was two times higher than for nanoconstruct 7.
  • the fluorescence spectrum in Figure 2 IB shows the difference in fluorescence intensity for 6, 7, and 8, which resulted from the different efficiency of the PS— >CD energy transfer in these nanoconstructs. This energy transfer caused a decrease in intensity of the PS fluorescence along with an increase in CD fluorescence intensity.
  • Figures 21A to 2 ID show: 21 A) Electronic absorption spectra of nanoconstructs 5, 6, 7, and 8 in water. 2 IB) Fluorescence emission of nanoconstructs 5, 6, 7, and 8 excited at 532 nm in water. 21C) Fluorescence emission of nanoconstructs 5, 7, and 8 excited at 785 nm. 2 ID) The singlet oxygen production of nanoconstructs 5, 6, 7, and 8 in water was detected by measuring the phosphorescence of singlet oxygen, l 0 2 , at 1270 nm upon excitation by a 532 nm laser. Nanoconstruct 5 was used as the instrument response function (IRF) as it does not produce l 02.
  • IRF instrument response function
  • HPPH derived from chlorophyll-a
  • the PDT response (no tumor regrowth) for HPPH 1 was 40%, 40%, 60%, 30%, 40%, and 30%, respectively.
  • the nanoconstruct 7 containing HPPH and cyanine dye 3 in a ratio of 2-1 was more effective than PS 1 alone in 1% Tween 80 formulation and nanoformulations 9 and 10, and also provided (i) the ability to both image and treat the tumors, which could be extremely useful for a "See and Treat” approach and (ii) compared to the synthetic HPPH- cyanine dye conjugate in which the imaging dose was 8- to 10-fold lower than the therapeutic dose, a single dose (0.47 moles/kg) of nanoconstruct 7 can be used for both tumor imaging and PDT.
  • FIGS 22A and 22B show Kaplan-Meier plots for BALB/c mice bearing subcutaneous Colon-26 tumors treated with PS 1 and various nanoconstructs at the PS dose of 0.47 mol/kg.
  • the tumors were exposed to light at the light fluence and fluence rate of 135 J/cm 2 @ 75 mW/cm 2 .
  • nanoconstruct 7 (containing HPPH and CD in a ratio of 2: 1) showed the best long term PDT efficacy (6/10 mice were tumor-free on day 60).
  • PAA NPs provide a great platform for post-loading because of the porous nature of the polyacrylamide- based hydrogels, wherein the hydrophobic part of the molecule may interact preferentially.
  • the surfactant Tween-80 plays an important role in efficient retention of the compounds within the NPs. Its presence in an aqueous solution apparently causes formation of a micellar layer on the nanoconstruct surface whereby the polyethers form the outer hydrophobic layer and the oleic acid forms the inner, more hydrophobic layer of the construct.
  • NIR fluorophores cyanine dyes in general have shown great potential for fluorescence imaging.
  • IR-820 a near-infrared (NIR) cyanine dye
  • NIR near-infrared
  • nanoconstructs 9 and 10 produced lower in vivo FRET signal as compared to nanoconstructs 7 and 8, however the nanoconstruct 7 still provided the best PDT outcome (60% for PS 1 vs. 40% for nanoconstruct 6). Further studies to improve the target-specificity of the nanoconstructs by introducing certain target-specific agents at the periphery of the PAA NPs are in progress.
  • AFPAA Blank Amine Functionalized Polyacrylamide Nanoparticles
  • hexane was removed by rotary evaporation and the particles were precipitated by addition of ethanol (50 ml).
  • the surfactant and residual monomers were washed away from the particles with ethanol (150 ml) followed by washing with water (100 ml) 5 times each in an Amicon ultra- filtration cell equipped with a Biomax 500 kDa cutoff membrane.
  • the concentrated nanoparticles were lyophilized for two days for storage, and reconstituted by suspending in water before use.
  • the NP solution of 0.2 mg/ml was prepared in water and a drop of the NP solution was placed on the SEM aluminium specimen mount (aluminum) and dried gradually at room temperature. The sample was then sputter coated with gold and the SEM images were taken on the Philips ESEM XL30.
  • the concentration of 1, 2, and 3 was measured spectrophotometrically in ethyl alcohol according to the Beer's-Lambert law using 47,500, 200,000, and 207,455 (L mol- 1 cm “1 ) as the molar extinction coefficient for 1, 2, and 3, respectively, after they were syringe filtered with a 0.2 ⁇ regenerated cellulose syringe filter. If scattering was present in the absorbance spectra, the NPs were centrifuged in a microfuge membrane-filter (Nanosep 100K Omega) at 14,000 RPM for 10 minutes. The filtrate was used to calculate the concentration of 1, 2, or 3. To create nanoconstrust 9 and 10, nanoconstruct 6 and nanoconstrust 3 are mixed together such that the molar ratio of PS 1 and cyanine dye 3 is 2-1 and 4-1, respectively. The NPs are stored at 4° C until further use.
  • Post-Loading of the photosensitizer 1 and the cyanine dye 3 to blank AFPAA to create nanoconstructs 7 and 8 The lyophilized AFPAA NPs were dissolved in 1% Tween- 80 to a final concentration of 10 mg of NPs/1 ml of 1% Tween-80.
  • the hydrodynamic diameter of the blank PAA NPs were measured by DLS prior to use to ensure the NPs are of appropriate size. If aggregation is present, the hydrodynamic diameter may increase; therefore the NPs were sonicated in a water bath sonicator for several minutes to disperse the aggregation.
  • 1 and 3 Prior to post-loading, 1 and 3 were dissolved in DMSO to prepare 20 mM solutions.
  • NP solution For 2 ml of NP solution, 20 or 40 ⁇ of 1 was post-loaded as the first step of making nanoconstructs 7 and 8, respectively, and excess reagents were removed by centrifuge filtration as before until no PS 1 is detected in the filtrate. The concentration of PS 1 was measured spectrophotometrically. If scattering is observed (determined spectrophotometrically), the NPs in ethanol were centrifuged in a microfuge membrane-filter at 14,000 RPM for 10 minutes to remove PS 1 from the NPs. The filtrate was used for calculating the concentration of the PS. The concentration of the PS 1 and 3 in nanoconstructs 7 and 8 was 200 and 400 ⁇ respectively.
  • mice The second group of mice was imaged with cyanine dye 3 and nanoconstruct 5 (dose: 0.3
  • the third and fourth groups of mice were imaged with nanoconstruct 7 containing the cyanine dye 3 post-loaded (dose: 0.216 moles/kg) and the PS 1 post-loaded (dose: 0.47 and the nanoconstruct 8 containing the cyanine dye 3 post-loaded (dose: 0.108 and PS 1 post-loaded (dose: 0.47
  • the fifth and sixth groups of mice were imaged post with nanoconstruct 9 containing the cyanine dye 3 post- loaded (dose: 0.236 and the PS 1 postloaded (dose: 0.47 and the nanoconstruct 10 contained 3 post-loaded (dose: 0.127 and the PS 1 post-loaded
  • Optical Imaging Setup The fluorescence imaging was conducted in accordance with a protocol approved by the Institutional Animal Care and Use Committee IACUC at Roswell Park Cancer Institute and the Guide for the Use of Laboratory Animals.
  • BALB/c mice (3 mice/group) bearing subcutaneous Colon 26 tumors on the right shoulder were injected i. v. (tail-vein) with either cyanine dyes or nanoconstructs/formulations.
  • mice were anesthetized with ketamine/xylazine by intraperitonial injection and imaged with a monochrome scientific grade CCD camera CRI (Nuance, Woburn, MA).
  • the CCD camera was employed in the mono mode and a 782 nm BWF light source (B&W-Tek, Newark, DE) continuous wave laser was used for excitation.
  • the fluorescence emission was collected with an 800 and 830 nm long pass filters in series.
  • FRET fluorescence resonance energy transfer
  • ET energy transfer
  • Figures 23A -23D shows an SEM for nanoconstruct 7
  • Fig. 23 B shows a DLS particle size distribution for Blank PAA NPs
  • Fig. 23C shows a particle size distribution for nanoconstruct 6
  • Fig. 23D shows a particle size distribution for nanoconstruct 5 in Tween-80 / water (concentration of Tween-80 is ⁇ 1%).
  • the mean diameter is 33.5 nm, 32.5 nm, and 35.2 nm for Fig. 23B, Fig. 23C, and Fig. 23D, respectively.
  • the mean hydrodynamic diameter as determined by DLS is similar for all nanoconstruct/formulations.
  • Figures 24A-24L show Release/Retention Profiles of PS 1 (24A, 24C, 24E,
  • 24G, & 241) and/or cyanine dye 3 (24B, 24D, 24F, 24H, & 24J) from nanoconstructs 5, 6, 7, 8, 9, and 10 in a 1% Human Serum Albumin (HSA) solution.
  • HSA Human Serum Albumin
  • the release/retention was measured immediately upon addition of the various nanoconstucts in 1% HSA (*), 4, and 24 hours post-addition of the nanoconstructs in a 1% HSA solution. Each experiment was done in triplicate with the symbols representing the mean. The error bars are standard error of the mean.
  • Figures 24K and 24L show Release Profiles of PS 1 and cyanine dye 3 from nanoconstruct 7 in a 25% Bovine Calf Serum (BCS) solution at 37 °C. The release was measured 4, 8, 12, and 24 hours post-addition of nanoconstruct 7 in 25% BCS. Each experiment was done in triplicate with the symbols representing the mean. The error bars are standard error of the mean.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A composition comprising PAA nanoparticles containing a post loaded tetrapyrollic photosensitizer and a postloaded imaging agent and methods for making and using same.

Description

MULTIFUNCTIONAL NANOPLATFORMS FOR FLUORESCENCE IMAGING AND PHOTODYNAMIC THERAPY DEVELOPED BY POST-LOADING PHOTOSENSITIZER AND FLUOROPHORE TO POLYACRYLAMIDE NANOPARTICLES
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority from U.S. Application No. 13/566,411, 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 Grant Numbers CAl 19358 and CAl 14053 awarded by the U.S. National Institutes of Health. The United States Government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0003] 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.
[0004] In efforts to develop effective photosensitizers with the required photophysical characteristics, compounds having a tetrapyrollic core ring were used. Usually, chlorophyll-a and bacteriochlorophyll-a were used as intermediates in sythesis. 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.
[0005] 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.
[0006] 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.
[0007] Fluorescence imaging is a non-invasive and non-ionizing imaging technique that requires only nanomoles of fluorophores for contrast enhancement. The NIR spectral range (-650 - 950 nm) is known as the "biological window" for optical imaging since light absorption due to water, deoxygenated hemoglobin and oxygenated hemoglobin is minimized in this region, as well as tissue autofluorescence and light scattering.
[0008] Fluorescence, the emission 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.
[0009] 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.
[0010] 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.
[0011] 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.
[0012] Both cancer detection and treatment depend on selective delivery of appropriate agents to the malignancy. Photodynamic therapy (PDT), a relatively new modality for the treatment of a variety of oncological, cardiovascular, dermatological and ophthalmic diseases, is based on the preferential localization of photosensitizing molecules, (photosensitizers, PS) in target tissues. Upon light activation, the PS produces reactive singlet oxygen5 which damages tumor cells and neovasculature, and also initiates antitumor inflammatory and immune responses. We and others have developed relatively tumor-avid PS which selectively accumulate in tumor, and these molecules have been used to carry optical, PET and MR imaging agents to the tumor sites. However, the tumor selectivity of current PS is not always adequate. Approaches that link PS to antibody fragments or receptor ligands have been disappointing because the number of required PS/cell generally is greater than the number of antigen or receptor binding sites. Conversely, the imaging agent carrying capacity of the individual PS molecules is limited.
[0013] It has recently been shown that HPPH, developed in our laboratory and currently under Phase VII clinical trials, when conjugated with certain cyanine dyes can be used for both fluorescence imaging and photodynamic therapy. The conjugate showed potential tumor imaging and PDT efficacy, but compared to the imaging dose the required therapeutic dose was 8-fold higher. Increasing the number of HPPH moieties in synthetic photosensitizer-cyanine dye (PS-CD) conjugates did not minimize the therapeutic dose.
[0014] 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 nanoparticles containing PDT agents, folate receptor-targeted, boron containing dendrimers for neutron capture and nanoparticle-directed thermal therapy.
[0015] 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.
[0016] Nevertheless, nanoparticles (NP) are uniquely promising in that (i) their hydrophilicity and charge can be altered; (ii) they possess enormous surface area which can be modified with functional groups possessing a diverse array of chemical and biochemical properties, including tumor-selective ligands; (iii) owing to their sub-cellular and sub-micron size, they can penetrate deep into tissues and are generally taken up efficiently by cells.
BRIEF SUMMARY OF THE INVENTION
[0017] The present invention relates to PAA nanoparticles (polyacrylic acid and its modified carboxy derivatives, e.g. polyacrylamide) containing a photosensitizer and an imaging enhancing agent.
[0018] 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, e.g. polyacrylamide 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.
[0019] 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.
[0020] The photosensitizer is preferably a tetrapyrollic 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 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 imgf000011_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 =N¾ where ¾ 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 ¾ is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, araalkyl, or ORc where Rc is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl or CO RdRe 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.
[0021] 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.
[0022] 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.
[0023] 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. In particular, a novel post-loading approach for constructing a multifunctional biodegradable polyacrylamide (PAA) nanoplatform for tumor-imaging (fluorescence) and photodynamic therapy (PDT is provided. This approach provides an opportunity to post-load the imaging and therapeutic agents at desired concentrations. Among the PAA nanoparticles, a formulation containing the photosensitizer, HPPH [3-(l '-hexyloxyethyl)pyropheophorbide- a], and the cyanine dye in a ratio of 2: 1 minimized the undesirable quenching of the HPPH electronic excitation energy due to energy migration within the nanoparticles and/or F5rster (fluorescence) resonance energy transfer (FRET) between HPPH and cyanine dye. An excellent tumor-imaging (MR fluorescence) and phototherapeutic efficacy of the nanoconstruct formulation is demonstrated. Under similar treatment parameters the HPPH in 1% Tween 80/5% aqueous dextrose formulation was less effective than the nanoconstruct containing HPPH and cyanine dye in a ratio of 2 to 1. This is the first example showing the utility of the post-loading approach in developing a nanoconstructs for tumor-imaging and therapy.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
As used herein "Figure" and "Fig." are used interchangeably.
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 RTF 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 NP)
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 μηιοΐ/kg in PAA NPs and 0.78 μηιοΐ/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 μθί). 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 μϋϊ, 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 injection.
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 structural formulas for HPPH at 1, IR820 cyanine dye (CD) at 2 and cyanine dye at 3 where the chloro group of IR820 is replaced with p-aminothiol.
Figure 17B illustrates various polyacrylamide acid (PAA) nanoparticles postloaded with HPPH (1), cyanine dye (2) and cyanine dye (3) alone and in different combinations and ratios.
Figure 18A shows whole body fluorescence images of BALB/c mice bearing Colon- 26 tumors. Control mouse (A, E, I, and M), cyanine dye 2 (B-D), nanoconstruct 4 (F-H), cyanine dye 3 (J-L), nanoconstruct 5 (N-P), 24, 48, and 72h post i.v. injection.
Figure 18B shows fluorescence intensity values for left, cyanine dye 2 and construct 4 and right, cyanine dye 3 and construct 5.
Figure 19 shows whole body fluorescence images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C & D), 8 (E & F), 9 (G & H) and 10 (I & J) in BALB/c mice bearing Colon-26 tumors. For A-J the excitation wavelength was 782 nm. Images A, B, C, E, G, & I and D, F, H, & J were taken 24 and 48 hours post i.v. injection, respectively.
Figure 20 shows whole body FRET images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C), 8 (D), 9 (E) and 10 (F) in BALB/c mice bearing Colon-26 tumors. For A-F the excitation wavelength was 665 nm. Images A-F were taken 24 hours post i.v. injection. Figure 21 A shows an absorbance curve for constructs 5, 6, 7, and 8 at various wave lengths.
Figure 2 IB shows fluorescent intensity for constructs 5, 6, 7, and 8 at 532 nm excitation.
Figure 21C shows fluorescent intensity for nanoconstructs 5, 7, and 8. Fluorescence was more intense for nanoconstructs 7 and 8 than for pure CD nanoconstruct 5, even if difference in absorption at 785 nm was minimal.
Figure 2 ID shows detector response for constructs 5, 6, 7, and 8 illustrating singlet oxygen decay rates.
Figure 22 A shows tumor response versus time for untreated control, PS1 (HPPH), and nanoconstructs 6, 7, and 8.
Figure 22B shows tumor response versus time for untreated control, nanoformulation
9 and nanoformulation 10.
Fig. 23A shows an SEM for nanoconstruct 7 (representative of all groups).
Fig. 23B shows a DLS particle size distribution for Blank PAA NPs. The mean diameter is 33.5 nm.
Fig. 23C shows a particle size distribution for nanoconstruct 6. The mean diameter is 32.5 nm, and
Fig. 23D shows a particle size distribution for nanoconstruct 5. The mean diameter is 35.2 nm.
Figures 24A-24L show Release/Retention Profiles of PS 1 (24A, 24C, 24E, 24G, & 241) and/or cyanine dye 3 (24B, 24D, 24F, 24H, & 24J) from nanoconstructs 5, 6, 7, 8, 9, and
10 in a 1% Human Serum Albumin (HSA) solution. Figures 24K and 24L show Release Profiles of PS 1 and cyanine dye 3 from nanoconstruct 7 in a 25% Bovine Calf Serum (BCS) solution at 37 °C. DETAILED DESCRIPTION OF THE INVENTION
[0024] Photosensitizers (photosensitizer) generally fluorescence 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 photosensitizer have relatively short fluorescent wavelengths, < 800 nm, which are not optimal for detection deep in tissues.
[0025] 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.
[0026] We have thus developed multifunctional nanovector platforms that can deliver tumor-avid therapeutic photosensitizers that only become active (and toxic) when illuminated by specific wavelengths of light, and, in addition, carry one or more imaging agents; these nano-platforms thus could enable both diagnosis and image guided therapy.
[0027] Among the nanoparticles, hydrogel polyacrylamide (PAA) in which the monomeric units are linked together with ester bonds have been of particular interest due to their biocompatibility/biodegradability and low toxicity Using biodegradable polymer based nanoparticles (NPs) avoids multi-step synthesis and has numerous advantages including the ability to create water soluble formulations with desired pharmacokinetic properties, capable of delivering a high payload of the multiple agents (therapeutic PS and imaging agents) to tumors, increased photostability of photoactive agents and fluorophores, and the ability to modify the surface of the NP for conjugation to a variety of biomolecules. NPs and other macromolecular objects can passively target the tumor interstitium, via the "Enhanced Permeability and Retention" (EPR) effect due to the leaky vascular system in tumors.21a'b In addition, the poor lymphatic drainage system in tumors causes fluid retention in the tumor interstitial space, which helps to retain polymeric nanoparticles and other macromolecular objects in the tumor compared to normal tissue.21a'b For these reasons, NPs are a promising means for delivering therapeutic and other molecular agents to tumors.
[0028] Because NPs could deliver a high payload of the drug to tumor, we investigated the use of a PAA-based nanoconstructs for delivering both the near-infrared (NIR) cyanine dye (CD) fluorophore and the red-light absorbing photosensitizer HPPH. The release of the desired imaging and therapeutic agents may also be controlled by creating a nanoparticle that is pH or temperature sensitive, or by modifying the pores of the NP matrix In a parallel study we encapsulated the PS within polymeric NPs, but the retention efficiency was low, therefore a large concentration of NPs was required to achieve the desired therapeutic dose. To increase the retention of the PS within the NP, we decided to form the NPs first and then load the PS into the porous PAA-NPs. This novel loading approach of the desired agents was termed "post-loading". In this procedure, both HPPH (phototherapeutic agent) and the cyanine dye (NIR fluorescence imaging agent) moieties were highly retained in the NPs (confirmed by release kinetics) and provided constructs for non-invasive detection of tumors and delineation of the tumor margins by NIR fluorescence imaging.
[0029] 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 tumor imaging, 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
[0030] 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.
[0031] PET imaging and PDT: PAA nanoparticles decreased the liver uptake of the 1241-photosensitizer (PET imaging agent) and enhanced the tumor-specificity. Our initial investigation with an 1241-labeled photosensitizer 2 indicates its in vivo PDT efficacy and capability of detecting tumorsl04-106 (RIF, 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 clears 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 Figures 8A-8C (only 72h images shown).
[0032] 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:
[0033] 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.
[0034] 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: [0035] 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:
[0036] 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 1 1). Cell internalization of F3-targeted nanoparticles was confirmed by fluorescence confocal microscopy.
F3-Cys shows target-specificity in 9L glioma cells:
[0037] 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:
[0038] 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:
[0039] 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 Figures 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.
[0040] 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 nanoparticle 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 porphyrin based 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 (100-150μΟί). The mice were imaged at 24, 48 and 72h post injection and biodistribution studies were performed at each time point summarized in Figurea 8A-8C (only 72h images shown). [0041] 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.
[0042] Importantly, according to the present invention, we compared the photosensitizing and NIR fluorescence imaging potential of several biodegradable PAA nanoparticle formulations, in which the HPPH and CD moieties were post-loaded at a 2 to 1 and a 4 to 1 ratio, respectively. These formulations were significantly different in tumor uptake, in pharmacokinetics and in in vivo imaging and PDT efficacy.
[0043] Additional examples and details associated with the post-loading of both the photosensitizer and fluorescent imaging compound are as follows:
[0044] Human Serum Albumin, Tween-80 and dimethyl sulfoxide (DMSO) were purchased from Sigma-Aldrich. Bovine Calf Serum (BCS) was purchased and dPBS (pH 7.4, lx, without calcium and magnesium) were purchased from Cellgro. Ethanol (200 proof) was purchased from Pharmco-Aaper. All solutions were prepared with 18 ΜΩ water purified by a Millipore Milli-Q Advantage A 10 water purification system. 30 and 100 kDa Amicon Ultra- 15 and Ultra 4 centrifuge filters were purchased from Fisher Scientific.
Animal Studies: All animal studies were performed following the animal protocol guidelines approved by Institutional Animal Care and Use Committee (IACUC).
[0045] The PAA nanoparticles were prepared by following our previous with slight modifications (See e.g. Multifunctional ORMOSIL and PAA nanoparticles, Gupta et al, Photodynamic Therapy: Back to the Future. Edited by Kessel, David FL. Proceedings of the SPIE, Volume 7380 (2009)., pp. 73805H- 73805H-12 (2009). [0046] Post-Loading of the Photosensitizer 1, and the cyanine dyes 2, and 3 to blank
AFPAA to create nanoconstructs 4, 5, 6, 9, and 10: In brief, 10 mg of lyophilized PAA NPs were suspended in 1 mL of 1% Tween-80 / water solution and to this solution 10 μΐ, of 1, 2, or 3 (20 mM in DMSO) is added and magnetically stirred at a constant rpm for 2 hours. The NPs were centrifuge filtered in a 30 kDa Amicon Ultra- 15 centrifuge filter for 30 minutes at 5,000 RPM and then the NPs were reconstituted with water. The nanoparticles were syringe filtered with a 0.2 μιη regenerated cellulose syringe filter. Nano formulation 9 and 10 were created by mixing nanoconstruct 1 and 3 such that the molar ratio of 1 to 3 was 2: 1 and 4: 1, respectively. The NPs are stored at 4° C until further use. For details see "Supporting Material Information".
[0047] Post-Loading of the photosensitizer 1 and the cyanine dye 3 to blank AFPAA to create nanoconstructs 7 and 8: Upon measuring the concentration of PS 1 in nanoconstruct 6, cyanine dye 3 in DMSO (20 mM) was added such that the molar ratio of PS 1 to cyanine dye 3 was either 2: 1 or 4: 1. Once cyanine dye 3 was added, the procedure is the same as for post-loading, PS 1 or cyanine dye 2 and 3. For detailed procedure see the "Supporting Material Information".
[0048] Release Kinetics Procedure: The in vitro release profile of the photosensitizer
1, and the cyanine dye 3 in nanoconstructs/formulations 5-10 was measured. The NPs from all formulations were suspended in a 1% human serum albumin (HSA) - water solution and immediately the absorbance value for the HSA/nanoconstruct solution was measured spectrophotometrically. To measure the release of the photosensitizer 1 and/or the cyanine dye 3 from the NP, the NP solution is centrifuge filtered in a 100 kDa Amicon ultra-4 centrifugation filter for 20 minutes at 4,000 RPM. The absorbance of the PS or fluorophore in the filtrate was spectrophotometically measured (filtrate 1). The NPs in the retentate were reconstituted to the original volume with 1% HSA and re-centrifuge filtered (filtrate 2) and measured spectrophotometrically. The amount of 1 and/or 3 retained by the NP was confirmed by measuring the absorbance of the retentate upon reconstitution to the original volume with 1% HSA. If the sum total of all filtrates and the retentate is less than 90% of the stock value for either chromophore then ethanol is added to the centrifuge filter to measure what had adsorbed to the filter. These measurements were taken immediately post-addition of the nanoconstructs in a 1% HSA solution, 4 and 24 hours post addition of the nanoconstructs in the 1% HSA solution. Additionally, the release of PS 1 and cyanine dye 3 in nanoconstruct 7 was measured in 25% bovine calf serum (BCS) at 37° C. The procedure followed for the release of the PS/fluorophore in 25% BCS was similar to that of 1% HSA, except that the measurements were taken at 4, 8, 12, and 24 h post-addition of nanoconstruct 7.
[0049] Optical Imaging Setup: The fluorescence imaging was conducted in accordance with a protocol approved by the Institutional Animal Care and Use Committee IACUC at Roswell Park Cancer Institute and the Guide for the Use of Laboratory Animals. BALB/c mice (3 mice/group) bearing subcutaneous Colon 26 tumors on the right shoulder were injected i. v. (tail-vein) with either cyanine dyes or nanoconstructs/formulations. For a detailed description of the groups of mice imaged along with the dose, see the "Supporting Material Information".
[0050] Absorbance, Fluorescence, and Singlet Oxygen Measurements: The absorbance measurements were performed on a Varian Cary-50 Bio UV-Visible spectrophotometer. The concentrations of the NP formulations were measured in ethanol utilizing 47,500, 200,000 and 207,455 L mol"1 cm"1 as the respective molar extinction coefficients of 1, 2, and 3.
[0051] A SPEX 270M spectrometer (Jobin Yvon) was used for acquisition of fluorescence emission spectra in the far red and MR spectral ranges, utilizing the first output port equipped with an InGaAs photodetector (Electrooptical Systems Inc., USA),. A diode- pumped solid-state laser (Verdi, Coherent) at 532 nm was the excitation source. Generation of singlet oxygen O2) was detected by its phosphorescence emission peaked at 1270 nm. The decays of this emission were acquired using the Infinium oscilloscope (Hewlett-Packard) coupled to the output of the Hamamatsu IR-PMT which is attached to the second output port of the SPEX 270M spectrometer. Nanoconstructs 5 - 8 in polystyrene cuvettes were placed in front of the entrance to the spectrometer. The emission signal was collected at 90-degrees relative to the exciting laser beam with the use of additional long-pass filters (a 950LP filter and/or a 538AELP filter) to attenuate the scattered light and fluorescence from the samples. A second harmonic (532 nm) from the nanosecond pulsed Nd:YAG laser (Lotis Til, Belarus) operating at 20 Hz was used as the excitation source for time-resolved measurements.
[0052] In-Vivo Photodynamic Therapy: Eight- to twelve-week-old BALB/cAnNCr mice (Jackson Laboratory, Bar Harbor, ME) were inoculated subcutaneously (s.c.) with lxlO6 Colon 26 cells. When tumors reached 40-70 mm3, mice were injected i.v. (tail vein) with PS 1 (formulated in 1% Tween 80/D5W) or PAA nanoconstructs/formulations 6-10 suspended in water and further diluted in D5W. 24 hours post i.v. injection (dose of PS 1 : 0.47 mol/kg), mice (BALB/c mice bearing Colon 26 tumors, 10 mice/group) were restrained in plexiglass holders and tumors were irradiated at 665 nm with a fluence and fluence rate of 135 J/cm2 at 75 mW/cm2, respectively, using a pumped argon-dye laser. The growth of tumors was measured two to three times per week and the mice were monitored for a total of 60 days post PDT treatment. When the tumor regrowth was >400 mm3, the mice were euthanized according to the guidelines of the institute approved animal protocol.
[0053] Preparation of HPPH and Near Infrared Cyanine Dye Post-Loaded PAA NPs.
In an ongoing SAR study with a series of cyanine dyes (CD), we modified IR820 2 with limited imaging potential to a highly avid CD 3 in which the chloro-group of IR820 was replaced with a p-aminothiol functionality. CD 3 formulated in 1% Tween 80 / 5% dextrose was tumor avid, but the corresponding PAA formulation produced enhanced tumor contrast. On the other hand PS 1 (HPPH) and nanoconstruct 6 showed similar PDT efficacy with 40% tumor cure at a dose of 0.47 mol/kg. Although the PAA formulation did not enhance the PDT efficacy at similar treatment parameters, it did show a markedly improved tumor- specificity (determined by fluorescence imaging).23 Our objective was to prepare a single platform for imaging and therapy, therefore we investigated a synthetic approach in which the PS 1 was conjugated with 3. The resulting product showed excellent tumor-imaging ability (dose: 0.3 mol/kg), but the therapeutic dose was 8- to 10-fold higher. The low activity of the conjugate could be due to a part of the singlet oxygen produced by exposing the tumors with light was quenched by the CD, which reduced its activity and thus required a higher dose of the agent (HPPH-CD) for achieving efficacy similar to PS (HPPH) 1. HPPH-CD conjugate also exhibited significant FRET, which indirectly correlates to singlet oxygen production, a key cytotoxic agent for PDT. In other words molecules with higher FRET should show reduced singlet oxygen production and PDT efficacy.
[0054] Therefore, for our present study we were interested in preparing a series of multifunctional PAA nanoplatforms in which the PS and the CD molecules are post-loaded together in variable ratios or separately post-loaded (Figure 17B, nanoconstructs 4-10) and to investigate their tumor imaging and therapeutic potential. We anticipated that among all the nanoconstructs, the nano formulation 10 in which PS and CD were separately post-loaded and then mixed in a ratio of 4 to 1 may show enhanced PDT response due to lower singlet oxygen quenching probability by the cyanine dye or the energy transfer between the two chromophores (PS and CD) which could result in higher singlet oxygen production and improved long term tumor cure.
[0055] See Figures 17A and 17B: Post-loading of amine functionalized PAA NPs with IR-820, cyanine dye 2, nanoconstruct 4; cyanine dye 3, nanoconstruct 5; HPPH, photosensitizer 1 : nanoconstruct 6; photosensitizer 1 and cyanine dye 3 at a 2: 1 molar ratio, nanoconstruct 7; and HPPH 1 and cyanine dye 3 at a 4: 1 molar ratio, nanoconstruct 8. Nanoformulations 9 and 10 are nanoconstruct 6 and 5 mixed such that the molar ratio of 1 to 3 is 2: 1 and 4: 1, respectively.
[0056] To characterize the size and dispersity of the nanoparticles, dynamic light scattering (DLS) and scanning electron microscopy (SEM) was utilized. The DLS showed a mean diameter of 33.5, 32.5, and 35.2 nm for blank nanoparticles, nanoconstruct 6, 5, and 7. The SEM demonstrated that the NPs are uniform and monodisperse, with a mean diameter of -25 nm (see supplemental).
[0057] For PDT, porous nanoparticles are advantageous since release of PS from the
NP is not required for the singlet oxygen to diffuse into the tumor cells. However, if the release profile is rapid the NP may not be able to efficiently deliver a high payload of the desired agent to tumor. Therefore, we investigated the release profiles of the photosensitizer 1 and the cyanine dye 3 from nanoconstructs 5-10, respectively, by incubating them in 1% human serum albumin (HSA) at variable time points. The release profiles are summarized in the supplemental section. The release of PS 1 in nanoconstruct 6 showed a two-phase release, where an increase in the release was seen in the first four hours, which subsequently decreased during the following 20 hours. Compared to nanoconstructs 7-10, nanoconstruct 6 showed the highest retention of PS 1 (HPPH) over a 24 hour time period with approximately 87% being retained. When comparing the percentage of PS 1 retained (at the initial time point, time zero), upon addition of 1% HSA, the nanoconstruct 8 and 10 showed the highest retention (~84 %) of the PS. To mimic the release of PS 1 and cyanine dye 3 from nanoconstruct 7 in vivo, the nanoformulation 7, which provided the best whole-body fluorescence imaging and PDT response, was also subjected for the release of both the chromophores in 25% bovine calf serum (BCS, 37°C) at 4, 8, 12, and 24 hours post-addition. The maximum release (3.2%) for PS 1 occurred at 4 h time point, whereas for the cyanine dye 3, the maximum release (2.8%) was observed at 8 h time point. These results were interesting to show a slower release of both the cyanine dye and the PS under physiologically relevant conditions.
[0058] Fluorescence imaging ability of various formulations.
[0059] To show that cyanine dye 2 (IR-820, Aldrich) has poor tumor selectivity, whole body fluorescence imaging of BALB/c mice bearing subcutaneous Colon 26 tumors was performed. Figure 18A shows whole body fluorescence images of BALB/c mice bearing Colon-26 tumors. Control mouse (A, E, I, and M), cyanine dye 2 (B-D), nanoconstruct 4 (F- H), cyanine dye 3 (J-L), nanoconstruct 5 (N-P), 24, 48, and 72h post i.v. injection. The fluorescence intensity values shown in Figure 18B are background subtracted from the control mouse (left, cyanine dye 2 and construct 4 and right, cyanine dye 3 and construct 5), The error bar is the standard deviation of the mean fluorescence intensity in the tumors, n = 3. * = statistical significance of the difference in mean fluorescence intensity (P < 0.05, student t-test).
[0060] The water insoluble fluorophore 2 was formulated in a 1% Tween-80 / 5% dextrose solution and was injected i.v. at a dose of 0.3
Figure imgf000032_0001
The mice were imaged at 24, 48, and 72 hours post- injection (Figure 18A at B-D). Due to the rather poor tumor localization of 2, we post-loaded it into PAA NPs formulation 4 and compared their tumor uptake and fluorescence imaging abilities. The results summarized in Figure 18B clearly indicate much improved tumor selectivity of the PAA NP formulation 4 over the free fluorophore 2. Under similar imaging parameters, modified cyanine dye 3 and the corresponding nanoconstruct 5 (cyanine dye 3 post-loaded to PAA NPs) were also imaged (Figure 18A). As can be seen, compared to cyanine dye 2 (Figure 18A), the modified version 3 showed higher uptake and improved tumor-imaging ability (Figure 18A, J-L). Upon post- loading the cyanine dye 3 to PAA NPs, its uptake and tumor-imaging ability at 24, 48 and 72 hours post-injection was further enhanced (Figure 18A at N-P) with the difference in intensity in the tumor for 5 being statistically higher (p < 0.05) at 24 hours post-injection. We then decided to investigate further the utility of these biodegradable nanoparticles in developing a "multifunctional" nanoplatform. The fluorescence imaging of PAA nanoconstructs/formulations 7-10 in which HPPH 1 and cyanine dye 3 were post-loaded at a ratio of 2: 1 and 4: 1 (either in a single nanoparticle 7 and 8 or in separate nanoparticles 9 and 10), respectively, was investigated in BALB/c mice bearing Colon 26 tumors. On comparing the images obtained by using the cyanine dye 3 alone and the corresponding nanoconstructs 5, 7-10 the maximum accumulation in the tumor for 3 was observed at 48 h post injection Figure 18A at K, whereas the nanoconstructs 5, 7 and 8 (Figures 19 and 20) produced the maximum tumor uptake at 24h post-injection. This could be due to a significant difference in the pharmacokinetic characteristics of the products in two different formulations. In nanoconstructs 7 and 8 in which the PS and CD were post-loaded in a ratio of 2 to 1 and 4 to 1 on excitation of the cyanine dye at 782 nm gave fluorescence at 866 and 870 nm respectively. Interestingly, both nanoconstructs on in vivo excitation at 665 nm produced a significant fluorescence beyond 860 nm, which can be explained by the phenomenon known as the F5rster (Fluorescence) Resonance Energy Transfer (FRET), or by the more general phenomena of energy migration, or excitation percolation, followed by energy trapping, analogous to the energy transport and funneling process in photosynthetic antenna. On the basis of imaging results summarized in Figure 20, the nanoconstruct 7 provided the greatest contrast between the tumor and non-tumor tissues.
[0061] Specifically, Figure 19 shows whole body fluorescence images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C & D), 8 (E & F), 9 (G & H) and 10 (I & J) in BALB/c mice bearing Colon-26 tumors. For A-J the excitation wavelength was 782 nm. Images A, B, C, E, G, & I and D, F, H, & J were taken 24 and 48 hours post i.v. injection, respectively and Figure 20 shows whole body FRET images of a control mouse (A), nanoconstructs/formulation 5 (B), 7 (C), 8 (D), 9 (E) and 10 (F) in BALB/c mice bearing Colon-26 tumors. For A-F the excitation wavelength was 665 nm. Images A-F were taken 24 hours post i.v. injection.
[0062] It is known that when a PS is in close proximity to a longer wavelength- absorbing fluorophore, with a spectral overlap between the fluorescence of the PS and the absorbance of the fluorophore, then upon exciting the PS, the PS fluorescence decreases due to the energy transfer to the fluorophore and the singlet oxygen yield also diminishes. This phenomenon was evident for the nanoconstructs containing both the PS and the cyanine dye. To determine the degree of energy transfer between PS and fluorophore, the fluorescence of nanoconstructs 7 and 8 were compared with 5 and 6. The concentration for the cyanine dye was kept constant in all nanoconstructs and the concentration of 1 for nanoconstruct 6 and 8 was kept the same and was two times higher than for nanoconstruct 7. The fluorescence spectrum in Figure 2 IB shows the difference in fluorescence intensity for 6, 7, and 8, which resulted from the different efficiency of the PS— >CD energy transfer in these nanoconstructs. This energy transfer caused a decrease in intensity of the PS fluorescence along with an increase in CD fluorescence intensity. The energy transfer was strongest for nanoconstruct 8 since the fluorescence spectrum displayed the least intense fluorescence band from PS moiety oax ~ 670 nm) and the most intense fluorescence band from the CD moiety kmax » 870 nm) upon excitation at 532 nm (Figure 21 A). Also, on excitation of 3 at 785 nm, CD fluorescence was more intense for nanoconstructs 7 and 8 than for pure CD nanoconstruct 5, even if difference in absorption at 785 nm was minimal (Figure 21C). This was an effect of the post- loaded PS molecules; their presence in the post-loaded PAA nanoparticles could result in a more dense environment for the CD molecules, which, in turn, enhanced the radiative rate for the CD fluorophore. Overall, combination of the facts that the PS fluorescence under 532 nm excitation for 7 and 8 was less intense than for 6 and, at the same time, CD fluorescence under 532 nm excitation was more intense for 7 and 8 than for 5 unambiguously demonstrates that energy transfer PS— >CD occurs in nanoconstructs and its efficiency for 8 was higher than that for 7. This higher ET efficiency can be explained by the higher concentration of PS post-loaded to nanoparticles resulting in less average distance between PS chromophores, allowing electronic excitation to migrate from one PS molecule to others, before being trapped by the CD chromophore. Lovell et al30 had reported a similar observation in a series of pyropheophorbide-a conjugated with quenchers. An increase in concentration of the photosensitizer possibly results in a higher probability of electronic excitation energy percolation causing the trapping of the electronic excitations by lower concentration of the quenchers (cyanine dyes).
[0063] To confirm that the more efficient PS— >CD energy transfer in the nanoconstructs correlates with a less efficient production of singlet oxygen, we compared the singlet oxygen generation yield of the nanoconstructs using the singlet oxygen phosphorescence spectroscopy. Phosphorescence decays are shown in Figure 2 ID and demonstrate two clearly distinguishable decay rates for the singlet oxygen. One is shorter with a lifetime of ~4 μ8 (which is close to the lifetime of singlet oxygen in water31) suggesting that it is derived from the excited oxygen molecules decaying in aqueous environment. The second decay rate is much longer with a lifetime of -100 μ8, which is apparently associated with singlet oxygen decaying within the PAA matrix. The production of singlet oxygen was highest for the PS only formulation (nanoconstruct 6) and decreases for three others in the order of 8>7>5. We assume that the production of singlet oxygen by 5 was negligible since it dose not contain PS; thus the decay curve for 5 practically depicts the Instrument Response Function of the setup. These results were understandable since there was twice the amount of PS in nanoconstruct 8 which should produce more singlet oxygen, even if energy transfer from PS is more efficient in 8 than in 7. Overall, it is important to stress that both 8 and 7 nanoconstructs showed singlet oxygen production comparable with that from the PS nanoparticle formulation, nanoconstruct 6. These and our current results demonstrate that in a two-chromophore system, an increase in FRET and/or energy percolation increases the fluorescence intensity of the acceptor or longer wavelength (lower energy) chromophore, which decreases singlet oxygen production.18'19
[0064] Comparative in vivo PDT efficacy of PAA NPs containing HPPH and CD in variable ratios (Nanoconstructs 6 - 10 vs. PS 1)
[0065] Specifically, Figures 21A to 2 ID show: 21 A) Electronic absorption spectra of nanoconstructs 5, 6, 7, and 8 in water. 2 IB) Fluorescence emission of nanoconstructs 5, 6, 7, and 8 excited at 532 nm in water. 21C) Fluorescence emission of nanoconstructs 5, 7, and 8 excited at 785 nm. 2 ID) The singlet oxygen production of nanoconstructs 5, 6, 7, and 8 in water was detected by measuring the phosphorescence of singlet oxygen, l02, at 1270 nm upon excitation by a 532 nm laser. Nanoconstruct 5 was used as the instrument response function (IRF) as it does not produce l02.
[0066] HPPH, derived from chlorophyll-a, is an effective PDT agent with low skin phototoxicity and, in BALB/c mice bearing Colon 26 tumors, a complete PDT response of 40% was observed at a dose of 0.47 moles/kg on exposing the tumors with light at 665 nm, delivered at a fluence and fluence-rate of 135 J/cm2 and 75 mW/cm2 24 hours post-injection. To compare the newly developed nanoconstructs with free HPPH, we used similar treatment parameters as described above. In preliminary screening, the PDT response (no tumor regrowth) for HPPH 1, nana-constructs 6, 7, 8, 9 and 10 was 40%, 40%, 60%, 30%, 40%, and 30%, respectively. The nanoconstruct 7 containing HPPH and cyanine dye 3 in a ratio of 2-1 was more effective than PS 1 alone in 1% Tween 80 formulation and nanoformulations 9 and 10, and also provided (i) the ability to both image and treat the tumors, which could be extremely useful for a "See and Treat" approach and (ii) compared to the synthetic HPPH- cyanine dye conjugate in which the imaging dose was 8- to 10-fold lower than the therapeutic dose, a single dose (0.47 moles/kg) of nanoconstruct 7 can be used for both tumor imaging and PDT.
[0067] To demonstrate both tumor imaging and PDT using the same compositions,
BALB/c mice bearing subcutanaceous Colon-6 tumors were treated as shown in Figures 22A and 22B. Figures 22A and 22B show Kaplan-Meier plots for BALB/c mice bearing subcutaneous Colon-26 tumors treated with PS 1 and various nanoconstructs at the PS dose of 0.47 mol/kg. The tumors were exposed to light at the light fluence and fluence rate of 135 J/cm2 @ 75 mW/cm2. Under same treatment parameters nanoconstruct 7 (containing HPPH and CD in a ratio of 2: 1) showed the best long term PDT efficacy (6/10 mice were tumor-free on day 60).
[0068] Compared to encapsulation, the post-loading approach was more effective when hydrophobic compounds were used in conjunction with PAA nanoparticles. PAA NPs provide a great platform for post-loading because of the porous nature of the polyacrylamide- based hydrogels, wherein the hydrophobic part of the molecule may interact preferentially.
We have found that the surfactant Tween-80 plays an important role in efficient retention of the compounds within the NPs. Its presence in an aqueous solution apparently causes formation of a micellar layer on the nanoconstruct surface whereby the polyethers form the outer hydrophobic layer and the oleic acid forms the inner, more hydrophobic layer of the construct.
[0069] Among all organic NIR fluorophores, cyanine dyes in general have shown great potential for fluorescence imaging. In this series of compounds, IR-820, a near-infrared (NIR) cyanine dye, is of particular interest due to its inherent desirable photophysical characteristics, namely excitation and emission in the NIR range beyond 750 nm, which allows for deeper tissue penetration of activating light; however, its tumor uptake is known to be low.
[0070] It has been shown that when Cy5 was encapsulated within PEG-coated silica nanoparticles (3.3 and 6.0 nm diameter, respectively), the fluorescence intensity of the dye increased by 2.0 - 2.5 times, compared to that of the free dye. This prompted us to investigate the utility of PAA nanoconstructs for the delivery of both imaging and therapeutic agents. We found that the newly constructed nanoparticles were capable of delivering a high payload of both the PS and the CD molecules to the tumor. This is likely due to the "Enhanced Permeability and Retention" (EPR) effect.
[0071] The UV-Visible and fluorescence spectrometry studies confirmed that both hydrophobic PS and hydrophilic fluorophores related to cyanine dyes can be co-loaded into amine functionalized PAA nanoconstructs and maintain their photophysical characteristics. The DLS and SEM images show that upon co-post-loading, the diameter and spherical shape of the NPs remain intact. The in vitro spectroscopic measurements show that excitation of nanoconstruct 8 channels more energy transport/FRET from HPPH to cyanine dye 7, resulting in reduced efficiency of 1 for PDT efficacy. In contrast to synthetic HPPH-cyanine dye conjugates, a single dose of the PAA nanoconstruct can be used simultaneously for tumor imaging and for efficient long-term tumor cure by PDT. Additionally, nanoconstructs 9 and 10 produced lower in vivo FRET signal as compared to nanoconstructs 7 and 8, however the nanoconstruct 7 still provided the best PDT outcome (60% for PS 1 vs. 40% for nanoconstruct 6). Further studies to improve the target-specificity of the nanoconstructs by introducing certain target-specific agents at the periphery of the PAA NPs are in progress.
[0072] Details for preparation of compounds and constructs are as follows: [0073] Materials: Acrylamide, Ν,Ν,Ν',Ν'- tetraethylmethylenediamine (TEMED), ammonium persulfate (APS), polyethylene glycol dodecyl ether (Brij 30), 3-(acryloyloxy)-2- hydroxypropyl methacrylate (AHM), hexane, and dioctyl sulfosuccinate (AOT) were purchased from Sigma-Aldrich (USA). 3-(aminopropyl) methacrylamide (APMA) was obtained from Polysciences Inc., USA, and ethanol (190 proof) was obtained from Fisher Scientific, USA. Nanosep 100K Omega filters were purchased from Pall USA. Amicon ultrafiltration cell equipped with a Biomax 500 kDa cutoff membrane was purchased from Millipore, USA
[0074] Synthesis of Blank Amine Functionalized Polyacrylamide Nanoparticles (AFPAA): The synthesis was similar to the previously reported method by Wisner et al.26 In brief, hexane (45 ml, VWR, USA) was added into a dried 100 ml round bottom flask and stirred under a constant purge of argon. Suitable amounts of AOT (1.6 g, Sigma-Aldrich, USA) and Brij 30 (3.1 g, Sigma-Aldrich, USA) were added to the reaction flask and stirring was continued under argon protection for 20 min. Acrylamide (71 1 mg), 3-(aminopropyl) methacrylamide (89 mg) and biodegradable 3-(acryloyloxy)-2-hydroxypropyl methacrylate (428 mg) were dissolved in phosphate buffered saline (2 ml) (PBS, pH = 7.4) in a glass vial by sonication to obtain a uniform solution. The solution was then added to the hexane reaction mixture and vigorously stirred for another 20 min at room temperature. Polymerization was initiated by adding freshly prepared ammonium persulfate aqueous solution (10% w/v aqueous solution, 40 1) and TEMED (40 1). The resulting solution was stirred vigorously overnight. At the completion of polymerization, hexane was removed by rotary evaporation and the particles were precipitated by addition of ethanol (50 ml). The surfactant and residual monomers were washed away from the particles with ethanol (150 ml) followed by washing with water (100 ml) 5 times each in an Amicon ultra- filtration cell equipped with a Biomax 500 kDa cutoff membrane. The concentrated nanoparticles were lyophilized for two days for storage, and reconstituted by suspending in water before use.
[0075] Characterization of Size and Dispersion of AFPAA: The dynamic light scattering measurements were performed on a Nicomp 370 Submicron Particle Sizer (Nicomp, Santa Barbara, CA). The NP solution was placed in a borosilicate glass capillary tube, and diluted with water to an intensity reading of 300 kHz. The readings were performed in triplicate with each run set for 5 minutes. The volume weighted Gaussian or Nicomp's proprietary analysis mode was utilized to determine the mean hydrodynamic diameter. The morphology and size of the dried NP were characterized by scanning electron microscope (SEM). The NP solution of 0.2 mg/ml was prepared in water and a drop of the NP solution was placed on the SEM aluminium specimen mount (aluminum) and dried gradually at room temperature. The sample was then sputter coated with gold and the SEM images were taken on the Philips ESEM XL30.
[0076] Post-Loading of the photosensitizer 1, and the cyanine dyes 2, and 3 to blank AFPAA to create nanoconstructs 4, 5, 6, 9, and 10: 10 mg of lyophilized PAA NPs were dissolved in 1 ml of 1% Tween-80 / 18 ΜΩ water solution. Prior to post-loading with the NPs, the hydrodynamic diameter was measured to ensure that the NPs are of the appropriate diameter and that the NPs are not aggregated. When they are aggregated, the hydrodynamic diameter increases. The NPs can be sonicated in a water bath sonicator for several minutes to disperse the aggregates. To post-load, 10 μΐ of 1, 2, or 3 was added to the NP solution and magnetically stirred at a constant rpm for at least 2 hours. Excess DMSO, Tween-80, and 1, 2, or 3 that did not post-load was removed via centrifuge filtration (Amicon Ultra- 15 30 kDa centrifuge filter), for 30 minutes at 5,000 rpm. The filtrate was measured spectrophotometically and if 1, 2, or 3 was detected; the retentate was reconstituted to the original volume with water and recentrifuged. This step was repeated until no absorbance for 1, 2, or 3 was detected in the filtrate. The concentration of 1, 2, and 3 was measured spectrophotometrically in ethyl alcohol according to the Beer's-Lambert law using 47,500, 200,000, and 207,455 (L mol-1 cm"1) as the molar extinction coefficient for 1, 2, and 3, respectively, after they were syringe filtered with a 0.2 μιη regenerated cellulose syringe filter. If scattering was present in the absorbance spectra, the NPs were centrifuged in a microfuge membrane-filter (Nanosep 100K Omega) at 14,000 RPM for 10 minutes. The filtrate was used to calculate the concentration of 1, 2, or 3. To create nanoconstrust 9 and 10, nanoconstruct 6 and nanoconstrust 3 are mixed together such that the molar ratio of PS 1 and cyanine dye 3 is 2-1 and 4-1, respectively. The NPs are stored at 4° C until further use.
[0077] Post-Loading of the photosensitizer 1 and the cyanine dye 3 to blank AFPAA to create nanoconstructs 7 and 8: The lyophilized AFPAA NPs were dissolved in 1% Tween- 80 to a final concentration of 10 mg of NPs/1 ml of 1% Tween-80. The hydrodynamic diameter of the blank PAA NPs were measured by DLS prior to use to ensure the NPs are of appropriate size. If aggregation is present, the hydrodynamic diameter may increase; therefore the NPs were sonicated in a water bath sonicator for several minutes to disperse the aggregation. Prior to post-loading, 1 and 3 were dissolved in DMSO to prepare 20 mM solutions. For 2 ml of NP solution, 20 or 40 μΐ of 1 was post-loaded as the first step of making nanoconstructs 7 and 8, respectively, and excess reagents were removed by centrifuge filtration as before until no PS 1 is detected in the filtrate. The concentration of PS 1 was measured spectrophotometrically. If scattering is observed (determined spectrophotometrically), the NPs in ethanol were centrifuged in a microfuge membrane-filter at 14,000 RPM for 10 minutes to remove PS 1 from the NPs. The filtrate was used for calculating the concentration of the PS. The concentration of the PS 1 and 3 in nanoconstructs 7 and 8 was 200 and 400 μΜ respectively. To 7 and 8, 20 μΐ of 3 in 20 mM DMSO was added and magnetically stirred for 2 hours. The same centrifuge filtration steps used for 1 were employed for 3. The concentration of 1 and 3 was measured spectrophotometrically (as described above), after they were syringe filtered with a 0.2 μιη regenerated cellulose syringe filter. The ratio of 1 to 3 in nanoconstructs 7 and 8 was 2-1 and 4-1, respectively. The NPs are stored at 4°C until further use.
[0078] Whole-body fluorescence imaging: The first group of BALB/c mice were imaged with cyanine dye 2 and the nanoconstruct 4 post loaded with cyanine dye 2 (dose: 0.3
The second group of mice was imaged with cyanine dye 3 and nanoconstruct 5 (dose: 0.3 The third and fourth groups of mice were imaged with nanoconstruct 7 containing the cyanine dye 3 post-loaded (dose: 0.216 moles/kg) and the PS 1 post-loaded (dose: 0.47 and the nanoconstruct 8 containing the cyanine dye 3 post-loaded (dose: 0.108
Figure imgf000042_0001
and PS 1 post-loaded (dose: 0.47 The fifth and sixth groups of mice were imaged post with nanoconstruct 9 containing the cyanine dye 3 post- loaded (dose: 0.236
Figure imgf000042_0002
and the PS 1 postloaded (dose: 0.47 and the nanoconstruct 10 contained 3 post-loaded (dose: 0.127
Figure imgf000042_0003
and the PS 1 post-loaded
Figure imgf000042_0004
[0079] Optical Imaging Setup: The fluorescence imaging was conducted in accordance with a protocol approved by the Institutional Animal Care and Use Committee IACUC at Roswell Park Cancer Institute and the Guide for the Use of Laboratory Animals. BALB/c mice (3 mice/group) bearing subcutaneous Colon 26 tumors on the right shoulder were injected i. v. (tail-vein) with either cyanine dyes or nanoconstructs/formulations. At 24, 48 and 72h post- injection, mice were anesthetized with ketamine/xylazine by intraperitonial injection and imaged with a monochrome scientific grade CCD camera CRI (Nuance, Woburn, MA). For tumor imaging with cyanine dyes 2 and 3, the CCD camera was employed in the mono mode and a 782 nm BWF light source (B&W-Tek, Newark, DE) continuous wave laser was used for excitation. The fluorescence emission was collected with an 800 and 830 nm long pass filters in series. To image the in vivo fluorescence resonance energy transfer (FRET) or energy transfer (ET) from the PS 1 to cyanine dye 3, 1 was excited at 665 nm from a pumped argon-dye laser, and the fluorescence emission of 3 was collected with an 800 and 830 nm long pass filters in series. Fluorescent images for each group were set to the same lookup table (LUT, royal) and signal intensity in ImageJ ( IH, USA).
[0080] Figures 23A -23D: Fig. 23A shows an SEM for nanoconstruct 7
(representative of all groups) and Fig. 23 B shows a DLS particle size distribution for Blank PAA NPs, Fig. 23C shows a particle size distribution for nanoconstruct 6, and Fig. 23D shows a particle size distribution for nanoconstruct 5 in Tween-80 / water (concentration of Tween-80 is < 1%). The mean diameter is 33.5 nm, 32.5 nm, and 35.2 nm for Fig. 23B, Fig. 23C, and Fig. 23D, respectively. The mean hydrodynamic diameter as determined by DLS is similar for all nanoconstruct/formulations.
[0081] Figures 24A-24L show Release/Retention Profiles of PS 1 (24A, 24C, 24E,
24G, & 241) and/or cyanine dye 3 (24B, 24D, 24F, 24H, & 24J) from nanoconstructs 5, 6, 7, 8, 9, and 10 in a 1% Human Serum Albumin (HSA) solution. The release/retention was measured immediately upon addition of the various nanoconstucts in 1% HSA (*), 4, and 24 hours post-addition of the nanoconstructs in a 1% HSA solution. Each experiment was done in triplicate with the symbols representing the mean. The error bars are standard error of the mean. Figures 24K and 24L show Release Profiles of PS 1 and cyanine dye 3 from nanoconstruct 7 in a 25% Bovine Calf Serum (BCS) solution at 37 °C. The release was measured 4, 8, 12, and 24 hours post-addition of nanoconstruct 7 in 25% BCS. Each experiment was done in triplicate with the symbols representing the mean. The error bars are standard error of the mean.

Claims

What is claimed is:
1. A composition comprising PAA nanoparticles containing a tetrapyrollic photosensitizer for Photodynamic therapy and an imaging agent for fluorescent imaging.
2. The composition of claim 1 wherein the tetrapyrollic photosensitizer has the structural formula:
Figure imgf000044_0001
or a phamaceutically 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
Figure imgf000045_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 Rb 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 CONRaR; 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.
3. The composition of claim 2 wherein the photosensitizer is post loaded onto the nanoparticle after nanoparticle formation.
4. The composition of claim 3 wherein the imaging agent is a cyanine dye.
5. The composition of claim 3 wherein the imaging agent is a 124I labeled compound.
6. The composition of claim 3 wherein the imaging agent is a PET, fluorescence or MR imaging agent.
7. The composition of claim 6 wherein the nanoparticle contains a targeting moiety.
8. The composition of claim 7 wherein the targeting moiety is a peptide, folic acid or a carbohydrate.
9. A method for imaging comprising:
injecting PAA nanoparticles containing post loaded photosensitizer into a subject, injecting an imaging agent, and
imaging the subject at 24, 48 and 72 h post injections.
10. A method for making PAA nanoparticle' s containing a photosensitizer and an imaging agent by post loading a photosensitizer and a fluorophore onto a preprepared PAA nanoparticle.
11. The method of claim 10 where the photosensitizer has the structural formula:
Figure imgf000047_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
Figure imgf000048_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 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;
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 RdRe 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 = ¾ where ¾ is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, or aryl, or is an amino acid residue.
12. A method for making PAA nanoparticle's by conjugating a fluorophore to a PAA nanoparticle and then postloading a photosensitizer to the PAA nanoparticle.
13. The composition of claim 4 where the cyanine dye is post loaded onto the nanoparticle.
14. The composition of claim 13 where the photosensitizer is HPPH.
15. The composition of claim 13 where the numerical ratio of postloaded photosensitizer moieties to cyanine dye moieties is from 1 to 1 to 10 to 1.
16. The composition of claim 13 where the numerical ratio of postloaded photosensitizer moieties to cyanine dye moieties is from 2 to 1 to 4 to 1.
17. The composition of claim 13 where the cyanine dye has the structural formula:
Figure imgf000049_0001
18. The composition of claim 14 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.
19. A composition comprising a mixture of different PAA nanoparticles, at least one of which contains a postloaded photosensitizer and at least one of which contains a postloaded cyanine dye.
20. The composition of claim 19 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.
21. A method for imaging and treatment of hyperproliferative tissue in an animal comprising: a) injecting a composition according to claim 1 in an amount of 0.05 to 1.0 μιηοΐ68/1¾
b) imaging the animal by fluorescent imaging to define and locate the hyperproliferative tissue, and
c) treating the defined and located hyperproliferative tissue with photodynamic therapy.
PCT/US2013/053347 2012-08-03 2013-08-02 Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles WO2014022740A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13/566,411 2012-08-03
US13/566,411 US10568963B2 (en) 2009-10-21 2012-08-03 Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles

Publications (1)

Publication Number Publication Date
WO2014022740A1 true WO2014022740A1 (en) 2014-02-06

Family

ID=50028550

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/053347 WO2014022740A1 (en) 2012-08-03 2013-08-02 Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles

Country Status (1)

Country Link
WO (1) WO2014022740A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110091373A1 (en) * 2009-10-21 2011-04-21 Health Research, Inc. Paa nanoparticles for enhancement of tumor imaging

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
Gupta et al. Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles
Son et al. Light-responsive nanomedicine for biophotonic imaging and targeted therapy
Li et al. Cyanine conjugates in cancer theranostics
Li et al. Sequential protein-responsive nanophotosensitizer complex for enhancing tumor-specific therapy
Lim et al. Nanophotosensitizers toward advanced photodynamic therapy of Cancer
Jeong et al. Photosensitizer-conjugated human serum albumin nanoparticles for effective photodynamic therapy
Shinde et al. Enhanced permeability and retention effect: A key facilitator for solid tumor targeting by nanoparticles
US20110288234A1 (en) Silica nanoparticles postloaded with photosensitizers for drug delivery in photodynamic therapy
Battogtokh et al. Active-targeted pH-responsive albumin–photosensitizer conjugate nanoparticles as theranostic agents
Pantiushenko et al. Development of bacteriochlorophyll a-based near-infrared photosensitizers conjugated to gold nanoparticles for photodynamic therapy of cancer
US20210052731A1 (en) Inorganic nanophotosensitizers and methods of making and using same
Fathi et al. Current trends in pyrrole and porphyrin-derived nanoscale materials for biomedical applications
US10568963B2 (en) Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles
Thomas et al. NIRF heptamethine cyanine dye nanocomplexes for multi modal theranosis of tumors
Zhao et al. The substituted zinc (II) phthalocyanines using “sulfur bridge” as the linkages. Synthesis, red-shifted spectroscopic properties and structure-inherent targeted photodynamic activities
Silindir‐Gunay et al. Near‐infrared imaging of diseases: A nanocarrier approach
Moukheiber et al. Surfactant-stripped pheophytin micelles for multimodal tumor imaging and photodynamic therapy
Zhang et al. A single-wavelength NIR-triggered polymer for in situ generation of peroxynitrite (ONOO−) to enhance phototherapeutic efficacy
Yan et al. Near-infrared emissive AIE nanoparticles for biomedical applications: From the perspective of different nanocarriers
Geng et al. NIR-triggered ligand-presenting nanocarriers for enhancing synergistic photothermal-chemotherapy
US8906343B2 (en) PAA nanoplatforms containing fluorophores and targeted moieties covalently linked and photosensitizer post-loaded
Grebinyk et al. Molecular and nanoparticulate agents for photodynamic therapy guided by near infrared imaging
WO2014022740A1 (en) Multifunctional nanoplatforms for fluorescence imaging and photodynamic therapy developed by post-loading photosensitizer and fluorophore to polyacrylamide nanoparticles
Marko et al. Multifunctional Agents for Cancer-Imaging and Photodynamic Therapy: Impact of Polyacrylamide-Based Nanoplatforms

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: 13826441

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: 13826441

Country of ref document: EP

Kind code of ref document: A1