WO2019014413A1 - Procédés de radiothérapie pour déclencher l'action de médicaments photo-activables - Google Patents

Procédés de radiothérapie pour déclencher l'action de médicaments photo-activables Download PDF

Info

Publication number
WO2019014413A1
WO2019014413A1 PCT/US2018/041753 US2018041753W WO2019014413A1 WO 2019014413 A1 WO2019014413 A1 WO 2019014413A1 US 2018041753 W US2018041753 W US 2018041753W WO 2019014413 A1 WO2019014413 A1 WO 2019014413A1
Authority
WO
WIPO (PCT)
Prior art keywords
energy
subject
light
activating
applying
Prior art date
Application number
PCT/US2018/041753
Other languages
English (en)
Inventor
Mark Oldham
Justus ADAMSON
Mark W. Dewhirst
Paul YOON
Harold Walder
Frederic A. Bourke, Jr.
Zakaryae Fathi
Wayne F. Beyer
Original Assignee
Immunolight, Llc
DUKE, University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immunolight, Llc, DUKE, University filed Critical Immunolight, Llc
Publication of WO2019014413A1 publication Critical patent/WO2019014413A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0057Photodynamic therapy with a photosensitizer, i.e. agent able to produce reactive oxygen species upon exposure to light or radiation, e.g. UV or visible light; photocleavage of nucleic acids with an agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N5/0613Apparatus adapted for a specific treatment
    • A61N5/062Photodynamic therapy, i.e. excitation of an agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • A61N2005/1092Details
    • A61N2005/1095Elements inserted into the radiation path within the system, e.g. filters or wedges
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • A61N2005/1092Details
    • A61N2005/1098Enhancing the effect of the particle by an injected agent or implanted device

Definitions

  • the present invention is related to methods and systems for treating a disorder or condition in a subject.
  • Radiotherapy to trigger light activated drugs has much potential for the treatment of many diseases, such as cancer.
  • PET tracers such as 2-deoxy-2-[(18)F]fluoro-D-glucose ((18) (FDG)
  • 18 F-FDG is a modified glucose molecule which
  • radioactive tracers have been used to photoactivate caged luciferin in a breast cancer animal model expressing luciferase
  • CR Cherenkov radiation
  • charged particles released upon radioactive decay may include electrons (such as ⁇ - particles, Auger electrons and conversion electrons), positrons ( ⁇ +), and a-particles.
  • electrons such as ⁇ - particles, Auger electrons and conversion electrons
  • positrons ⁇ +
  • a-particles As these particles travel, the charged particles lose energy through interactions with the surrounding matter. In the biological context this matter is mostly water. At speeds below the speed of light in water, the randomly oriented polar water molecules will align with the passing of the charged particle. After the particle passes, these aligned water molecules along this path will relax back to a lowest energy state. In cases, when the particle is traveling at super-relativistic phase velocities (i.e. the particle travels faster than the speed of light in a particular), the polarized molecules relax by releasing energy in the form of visible radiation luminescence.
  • the following background discussions describe the conventional understanding of 1) psoralens and their photoreactivity and 2) alkylating agents and their photoreactivity.
  • the present invention can utilize those and other pathways to cause reactions of the photoreactive drugs with target cells.
  • U.S. Pat. No. 6,235,508 describes that psoralens are naturally occurring compounds which have been used therapeutically for millennia in Asia and Africa.
  • the action of psoralens and light has been used to treat vitiligo and psoriasis (PUVA therapy; Psoralen Ultra Violet A).
  • Psoralen is capable of binding to nucleic acid double helices by intercalation between base pairs; adenine, guanine, cytosine and thymine (DNA) or uracil (RNA).
  • psoralen in its excited state reacts with a thymine or uracil double bond and covalently attaches to both strands of a nucleic acid helix.
  • the crosslinking reaction appears to be specific for a thymine (DNA) or a uracil (RNA) base. Binding may proceed when psoralen is intercalated in a site containing thymine or uracil, but an initial photoadduct must absorb a second UVA photon to react with a second thymine or uracil on the opposing strand of the double helix in order to crosslink each of the two strands of the double helix, as shown below. This is a sequential absorption of two single photons as shown, as opposed to simultaneous absorption of two or more photons.
  • U.S. Pat. No. 4,748, 120 of Wiesehan is an example of the use of certain substituted psoralens by a photochemical decontamination process for the treatment of blood or blood products.
  • Additives such as antioxidants are sometimes used with psoralens, such as 8-MOP, AMT and I-IMT, to scavenge singlet oxygen and other highly reactive oxygen species formed during photoactivation of the psoralens. It is well known that UV activation creates such reactive oxygen species, which are capable of seriously damaging otherwise healthy cells Much of the viral deactivation may be the result of these reactive oxygen species rather than any effect of photoactivation of psoralens.
  • Some of the best known photoactivatable compounds are derivatives of psoralen or coumarin, which are nucleic acid intercalators. For psoralens and coumarins, this chemical pathway is likely to lead to the formation of a variety of ring-opened species, such as shown below for coumarin:
  • U.S. Pat. No. 5,216, 176 describes a large number of psoralens and coumarins that have some effectiveness as photoactivated inhibitors of epidermal growth factor. Halogens and amines are included among the vast functionalities that could be included in the
  • U. S. Pat. No. 5,984,887 describes using extracorporeal photopheresis with 8-MOP to treat blood infected with CMV.
  • the treated cells as well as killed and/or attenuated virus, peptides, native subunits of the virus itself (which are released upon cell break-up and/or shed into the blood) and/or pathogenic noninfectious viruses are then used to generate an immune response against the virus, which was not present prior to the treatment.
  • INA is hydrophobic compound that preferentially partitions into lipid bilayers of the Ebola virus. These authors reported that the "INA treatment renders ZEBOV completely noninfectious without structural perturbation” and that "INA- inactivated ZEBOV was immunogenic and protected mice from lethal challenge.”
  • U.S. Pat. No. 7,049, 110 entitled “Inactivation of West Nile virus and malaria using photosensitizers” describes the inactivation of microorganisms in fluids or on surfaces, preferably the fluids that contain blood or blood products and biologically active proteins. An effective, non-toxic amount of a photosensitizer was added to the fluid, and the fluid was exposed to photoradiation sufficient to activate the photosensitizer whereby microorganisms were inactivated.
  • the ' 110 patent describes a7,8-dimethyl-10-ribityl isoalloxazine photosensitizers and other photosensitizers including endogenous alloxazine or isoalloxazine photosensitizers.
  • the ⁇ 10 patent describes the treatment of a host carrying various microorganisms including viruses (both extracellular and intracellular), bacteria, bacteriophages, fungi, blood-transmitted parasites such as malaria, and protozoa.
  • viruses include acquired immunodeficiency (HIV) virus, hepatitis A, B and C viruses, sinbis virus, cytomegaloviris, vesicular stomatitis virus, herpes simplex viruses, e.g.
  • Bacteriophages include ⁇ 174, ⁇ 6, ⁇ , R17, T4, and T2.
  • Exemplary bacteria include P. aeruginosa, S. aureus, S. epidermis, L.
  • microorganisms is non-screened microorganisms— those microorganisms that are not screened by current blood banking processes. Some non-screened microorganisms include malaria and West Nile virus. One class of microorganisms includes those transmitted by mosquitoes, including malaria and West Nile virus.
  • the ⁇ 10 patent describes that the preferable use endogenous photosensitizers, including endogenous photosensitizers which function by interfering with nucleic acid replication.
  • endogenous photosensitizers including endogenous photosensitizers which function by interfering with nucleic acid replication.
  • the chemistry believed to occur between 7,8-dimethyl-10- ribityl isoalloxazine and nucleic acids does not proceed via singlet oxygen-dependent processes (i.e. Type II mechanism), but rather by direct sensitizer-substrate interactions (Type I mechanisms).
  • 7,8-dimethyl-lO-ribityl isoalloxazine appears not to produce large quantities of singlet oxygen upon exposure to UV light, but rather exerts its effects through direct interactions with substrate (e.g., nucleic acids) through electron transfer reactions with excited state sensitizer species.
  • the '602 patent describes methods for inactivating an infective agent or cancer cell that involve exposing the agent or cell to a hydrophobic photoactivatable compound, for example, 1,5- iodonaphthylazide (INA) activated by ultraviolet light.
  • a hydrophobic photoactivatable compound for example, 1,5- iodonaphthylazide (INA) activated by ultraviolet light.
  • INA 1,5- iodonaphthylazide
  • Psoralens are biologically inert molecules that are well known for anti-cancer therapeutic effects when photo-activated by ultra-violet radiation (Bethea, D., et al., J Dermatol Sci, 1999. 19(2): p. 78-88). Photo-activated psoralen has been shown to bind to various cellular components including DNA (17%), intra-cellular proteins (57%), and lipids (26%) (Gasparro, F.P., et al., Recent Results Cancer Res, 1997. 143: p. 101-27).
  • Immunogenic responses have been observed in patients treated with psoralen with proposed mechanisms including apoptosis, upregulation of Major Histocompatibility Complex I (MHC I), upregulation of immunogenic transcription factors (e.g. NF-kB, NF-AT, AP-1), and promotion of T cell development, maturation and proliferation (Bethea, D., et al., J Dermatol Sci, 1999. 19(2): p. 78- 88; Gasparro, F.P., et al., Recent Results Cancer Res, 1997. 143: p. 101-27; Schmitt, I.M., et al., J Photochem Photobiol B, 1995. 27(2): p.
  • MHC I Major Histocompatibility Complex I
  • immunogenic transcription factors e.g. NF-kB, NF-AT, AP-1
  • the present disclosure relates to the use of Cherenkov radiation (CR) to trigger light activation drugs inside a patient or subject.
  • CR Cherenkov radiation
  • the methods and systems of the present disclosure do not need or rely on light from radioactive traces to trigger light activation drugs.
  • the methods described herein exploit the energy deposition properties of high energy X-rays, generated for example by linear accelerators to generate light inside the subject being treated and to thereby activate drugs in vivo.
  • a method for treating a subject with a disorder which provides within the subject at least one photoactivatable drug for treatment of the subject applies initiation energy from at least one source to generate inside the subject a preferential x-ray flux for generation of Cherenkov radiation (CR) light capable of activating the at least one photoactivatable drug, and from the CR light, activating inside the subject the at least one photoactivatable drug to thereby treat the disorder.
  • CR Cherenkov radiation
  • a system for treating a subject with a disorder which provides within the subject at least one photoactivatable drug for treatment of the subject, applies initiation energy from at least one source to generate inside the subject a preferential x-ray flux for Cherenkov radiation (CR) light capable of activating the at least one photoactivatable drug, and from the CR light, activating inside the subject the at least one photoactivatable drug to thereby treat the disorder.
  • CR Cherenkov radiation
  • FIG. lA and IB are flow cytometry graphs showing activation of 4T1 cells with 3.3Gy irradiation with and without AMT (psoralen).
  • FIG. 2A is a graph showing that AMT (psoralen) exposure was minimized (removed immediately after irradiation).
  • FIG. 3 is a schematic of a system according to one exemplary embodiment of the invention.
  • FIG. 4 is a schematic of an exemplary system according to one embodiment of the invention for treatment of a biological medium.
  • FIG. 5 is a schematic illustrating an exemplary computer system for implementing various embodiments of the invention.
  • FIG. 6A is a schematic of the experimental setup used to ascertain the relative Cherenkov radiation output per x-ray dose.
  • FIG. 6B is a plot of the measured Cherenkov radiation output normalized to account for differences in the total x-ray dose through different low atomic number (low atomic mass) filters:
  • FIG. 6C is a comparison of the UV-Vis Cherenkov light spectrum with and without a 10 cm thick polyurethane filter.
  • FIG. 7A is a plot of cell kill as a function of TMP concentration with and without exposure to UV-Vis Cherenkov light.
  • FIG. 7B is a plot of the flow cytometry data acquired from B16 melanoma cells indicating a similar effect to the cytotoxicity depicted in Figure 7A.
  • FIG. 7C is a plot of the results of FIG. 7B with the data presented in terms of cell kill and MHC fraction.
  • FIGS. 8A and 8B show an experimental set-up for in-vitro investigation of CLAP.
  • FIG. 9 shows an experimental set-up to measure the CL output per unit radiation dose.
  • FIG. 10A shows Cell-Titer Glo® ATP luminescence assay results at varying
  • FIG. 10B shows Cell-Titer Glo® ATP luminescence assay results at varying concentrations of psoralen (TMP) for 4T1 cells.
  • TMP psoralen
  • FIGS. 11 A and 1 IB show flow cytometry results for B16 melanoma, demonstrating CLAP causes a substantial increase in MHC I expression over and above that caused by radiation alone.
  • FIGS. 12A and 12 B show B16 clonogenic survival data, all cells receiving ⁇ psoralen.
  • FIG. 13 A shows relative psoralen absorbance spectrum of 8-MOP at lC ⁇ g/mL compared to Cherenkov emission for 15MV clinical photon beam in water (obtained using GEANT4/GAMOS Monte Carlo simulations) and psoralen-UVA (PUVA) light source.
  • FIG. 13B shows CL output per MV radiation dose physically measured from the set-up illustrated in Figure 9, demonstrating effects of beam energy and polyurethane (low-Z) filter.
  • Articles "a” and “an” are used herein to refer to one or to more than one (i.e. at least one) of the grammatical object of the article.
  • an element means at least one element and can include more than one element.
  • This invention describes an enhanced therapeutic paradigm for radiotherapy, where the therapeutic treatments are delivered as normal, but an additional highly localized damage component is generated through Cherenkov Light Activation of specific drugs that are activated by UV light.
  • Cherenkov light activation solves the major technical limitation of these drugs (limited depth penetration of UV light) because Cherenkov UV radiation is produced naturally when high energy photons liberate secondary high energy electrons throughout the beam path in tissue. While other groups (Ran et al.
  • the present invention in one embodiment provides a more effective treatment since the Cherenkov light intensity from a linear accelerator high energy photon or electron beam is an order of magnitude greater than that of PET radionuclides, and can be further optimized through techniques such as x-ray beam filtering (described below).
  • Drugs that can be activated by Cherenkov light include any UV activated bio-therapeutic, of which psoralen is only one example. Other drugs which are activated by visible radiation may also be indirectly activated by down conversion of the Cherenkov UV light using the energy modulation agents described below.
  • Psoralen is a biologically inert natural compound which transforms to a powerful anti-cancer therapeutic when photo-activated (illuminated with UV light). It has found wide clinical application in treatment sites amenable to UVA light exposure (skin cancer and extracorporeal photopheresis (ECP, FDA approved as UVADEX®). Use of more potent forms of psoralen such as AMT can also increase the bio-therapeutic effect.
  • Aminomethyltrioxsalen when activated by Cherenkov light caused by 15 MV photons. Cell exposure to psoralen was minimized for this experiment; the AMT was removed
  • Figures 1 A and IB depict flow cytometry for 4T1 cells irradiated with 3.3Gy of 15MV photons with (A) and without (B) psoralen included. Psoralen was removed immediately following irradiation by washing the cells in media. The medium was removed from the well plates, leaving only those cells that are adhered to the plate surface. The increased early apoptotic signal in the A group with psoralen indicates the Cherenekov light activation of psoralen.
  • One embodiment of the invention optimizes the photon spectrum from the LINAC to achieve maximum useful UV light generation per unit dose (Gy). Spectrum modification is achieved utilizing low-atomic number filters (e.g. carbon) in-place of the conventional flattening filter, which preferentially absorbs low energy photons.
  • Current medical LINACs contain a flattening filter made from aluminum and copper which flatten the beam through beam- hardening to achieve a flat profile at typical treatment depth of 10 cm.
  • the flattening filter is placed in the photon beam path, located after the electron target. It serves to create a flat dose profile over a clinically useable treatment field size (e.g., up to 40 cm ⁇ 40 cm).
  • Flattening filters are typically cone shaped; they attenuate the center of the field the greatest, so as to achieve the same fluence intensity on the central axis as at the field edge. They are typically composed of dense metals with high atomic weight (such as Tungsten), so as to achieve maximum attenuation in the smallest thickness necessary.
  • Figure 2B is a plot showing the production of Cherenkov radiation in various dielectric media as a function of electron energy.
  • the Cherenkov intensity asymptotically increases with saturation occurring soon after lMeV (from
  • a preferential x-ray flux in a target medium for inducing a biological change produces more Cherenkov radiation per x-ray dose than its original x-ray spectrum from its original source would have produced upon absorption in the same target medium.
  • a preferential x-ray flux in a target medium for inducing a biological change produces between 5-10% more Cherenkov radiation per x-ray dose than its original x-ray spectrum from its original source would have produced upon absorption in the same target medium.
  • a preferential x-ray flux in a target medium for inducing a biological change produces between 5-20% more Cherenkov radiation per x-ray dose than its original x-ray spectrum from its original source would have produced upon absorption in the same target medium.
  • a preferential x-ray flux in a target medium for inducing a biological change produces between 5-50% more Cherenkov radiation per x-ray dose than its original x-ray spectrum from its original source would have produced upon absorption in the same target medium.
  • a preferential x-ray flux has removed from its original source a higher percentage of lower energy x-rays that do not contribute to Cherenkov radiation (e.g. x-rays of 0.3 MeV or lower) than of higher energy x-rays (e.g. x-rays of 1 MeV or higher) which do contribute to Cherenkov radiation.
  • a preferential x-ray flux has removed from its original source a higher percentage of lower energy x-rays that do not contribute to Cherenkov radiation (e.g. x-rays of 0.5 MeV or lower) than of higher energy x-rays (e.g. x-rays of 1 MeV or higher) which do contribute to Cherenkov radiation.
  • Cherenkov radiation e.g. x-rays of 0.5 MeV or lower
  • higher energy x-rays e.g. x-rays of 1 MeV or higher
  • a preferential x-ray flux has removed from its original source a higher percentage of lower energy x-rays that do not contribute to Cherenkov radiation (e.g. x-rays of 1.0 MeV or lower) than of higher energy x-rays (e.g. x-rays of 5 MeV or higher) which do contribute to Cherenkov radiation.
  • Cherenkov radiation e.g. x-rays of 1.0 MeV or lower
  • higher energy x-rays e.g. x-rays of 5 MeV or higher
  • a preferential x-ray flux has removed from its original source a higher percentage of lower energy x-rays that do not contribute to Cherenkov radiation (e.g. x-rays of 1.0 MeV or lower) than of higher energy x-rays (e.g. x-rays of 10 MeV or higher) which do contribute to Cherenkov radiation.
  • Cherenkov radiation e.g. x-rays of 1.0 MeV or lower
  • higher energy x-rays e.g. x-rays of 10 MeV or higher
  • the low-atomic number filter would have a completely different purpose from the conventional flattening filter. More specifically, the purpose of the low-atomic number filter would be to alter the x-ray fluence spectrum of the LINAC beam in order to maximize Cherenkov light production in the tumor per unit dose of radiation.
  • the low-atomic number filter of the invention would have a thickness and mass composition that would remove the lower energy x-ray photons that result in only a small amount of or no Cherenkov radiation from the beam while transmitting the higher energy x-ray photons.
  • Low-atomic number filters (such for example filters made with a substantial fraction of carbon) would exhibit this kind of x-ray photon transmittance useful in the present invention. While not limited to the following thicknesses, depending on the materials selected, the thickness of the low mass filter preferentially absorbing lower energy x-rays can range from mm to cm or more in thickness.
  • a preferential x-ray flux in a target medium for inducing a biological change produces more Cherenkov radiation per x-ray dose than its original x-ray spectrum filtered by a flattening filter would have produced upon absorption in the same target medium.
  • the filter comprises a carbon filter (e.g., a graphite or amorphous carbon filter) having a thickness in the range of 0.5 to 50 cm, or 1 to 20 cm, or 2 to 10 cm, or 5 to 7 cm, or ranges in between and overlapping.
  • the x-ray photons transit the thickness of the carbon filter where the lower energy x-ray photons are preferentially absorbed.
  • the filter comprises a natural or synthetic polymer filter (e.g., a polyurethane filter or polytetrafluorethylene filter or a silicone filter) having a thickness in the range of 0.5 to 50 cm, or 1 to 20 cm, or 2 to 10 cm, or 5 to 7 cm, or ranges in between and overlapping.
  • the x-ray photons transit the thickness of the polymer filter where the lower energy x-ray photons are preferentially absorbed.
  • the invention utilizes "flattening filter free" radiotherapy beams, for which the flattening filter is eliminated. These beams have the advantage of increased dose rate and the passing of higher energy x-rays which would produce a higher percentage of Cherenkov radiation, but at the cost of the beam being un-flattened.
  • fluorophores capture portions of the Cherenkov spectrum and re- emit in the ultraviolet and the visible range which is useful for psoralen (or equivalent) activation.
  • fluorophores that can absorb in the UV-blue range and emit at a lower energy (e.g., toward red) would be suitable for activating drugs that are sensitive to the visible light (i.e., for drugs which have peak absorption in the visible).
  • organic molecules can be used that down-convert from X-Ray into UV and Visible. Organic compounds can be used to achieve the same down conversion.
  • Anthracene and anthracene based compounds can be used.
  • Anthracene exhibits a blue (400-500 nm peak) fluorescence under ultraviolet light.
  • Antharacene also exhibits fluorescence under X- Ray energy.
  • both x-rays in the target medium and Cherenkov radiation in the target medium can be down-converted to light matched to the photoactive drug or determined to be capable of activating the photoactive drug.
  • plastic scintillators plastic scintillator fibers and related materials are made of polyvinyltoluene or styrene and fluors can be used. These and other formulations are commercially available, such as from Saint Gobain Crystals, as BC-414, BC-420, BC-422, or BCF-10.
  • organic molecules could then be used to assist in activation of a drug such as psoralen because these organic molecules would be able to capture a part of the CR spectrum and a part of the x-rays escaping without use and provide an additional source of internal UV light generated inside the patient or subject.
  • Another embodiment involves selection of the linear accelerator dose rate to optimize the drug activation by the Cherenkov light. The following examples are added by way of illustration and not limitation.
  • a filter made of low-Z material e.g. carbon as discussed above
  • the filter replaces the standard flattening filter for 15+ MV photon beams or could be used in addition to the standard flattening filter.
  • CLAP Cherenkov Light Activation of Psoralen
  • SBRT liver Stereotactic Body Radiation Therapy
  • SRS stereotactic radiosurgery
  • the doses are typically higher which could be important because Cherenkov production is proportional to dose.
  • the Cherenkov light photo-activates powerful anti-cancer bio-therapeutics (e.g., psoralen) with potential to add a long-term immunogenic response to SBRT/SRS treatment.
  • the above-noted fluorophores or down converting energy modulation agents in this embodiment maybe used to capture the Cherenkov light emitted at wavelengths outside the range for drug activation, and re-emit at energies within the activation range.
  • the SBRT and SRS treatments are delivered as normal, but an additional highly localized "damage" component (due to photoactivation of psoralen for example) is generated through Cherenkov Light
  • psoralen is a biologically inert natural compound which transforms to a powerful anti-cancer therapeutic when photo-activated (illuminated with LT light).
  • psoralen under exposure to the Cherenkov radiation, can be made to form monoadducts or photoadducts 4', 5' or photoadducts 3,4 or crosslink (where both types of photoadducts.
  • Psoralen and its derivatives have found wide clinical application in treatment sites amenable to UVA light exposure (skin cancer and extracorporeal photopheresis (ECP, FDA approved as UVADEX®).
  • the CLAP enhanced therapeutic treatment of the present invention addresses this limitation by using Cherenkov UV and blue radiation produced when high energy photons liberate secondary high energy electrons throughout the beam path in tissue.
  • the Cherenkov light from radiotherapy can permit real-time surface dose measurements, thereby monitoring of the total Gy exposure.
  • the Cherenkov light reflected off the surface of the patient can be imaged using a UV sensitive camera.
  • Cherenkov light is proportional to the radiation dose delivered. Workers have described in Medical Physics 38 (7) pages 4127-4132 (2011 ), the entire contents of which are incorporated herein by reference, this approach for determining a dose.
  • SBRT/SRS treatments are delivered with an optimized LINAC photon spectrum (using for example the low-mass filter described above) and generate sufficient psoralen photo-activation which, in turn, produces a long-term immunogenic component induced by the patient's autoimmune response to the "damaged" cells.
  • a system for imaging or treating a tumor in a human or animal body.
  • the system includes a pharmaceutical carrier including a photoactivatable drug and an optional pharmaceutical carrier, an x-ray or high energy electron or proton source capable of producing energies for the x-rays, electrons, or protons which yield in a target material CR light, and a processor programmed to control a dose of x-rays or electrons to the tumor for production of CR light inside or in the vicinity of the tumor to activate the photoactivatable drug.
  • the method in one embodiment of the invention includes injecting into a vicinity of and inside the tumor a pharmaceutical carrier including the photoactivatable drug, applying x-ray or high energy electrons or protons to the tumor, and producing the CR light inside or in the vicinity of the tumor to activate the photoactivatable drug.
  • the low mass filter predominantly transmits x-ray photons having energies predominantly greater than 0.5 MeV, or greater than 1.0 MeV, or greater than 1.5 MeV, or greater than 2.0 MeV.
  • the present invention can also use energy modulation agents (e.g., phosphors or other down conversion media), combinations of different down conversion media, upconversion media, combinations of different up conversion media, and/or combinations of different up and down conversion media.
  • energy modulation agents e.g., phosphors or other down conversion media
  • combinations of different down conversion media e.g., upconversion media, combinations of different up conversion media, and/or combinations of different up and down conversion media.
  • Radiation from the energy modulation agents can assist or supplement the CR radiation to alter the biological activity of the medium, as described in more detail below.
  • a system or method for light stimulation within a medium has a high energy x-ray or electron or proton source which provides high energy x-rays or electrons or protons into the medium to be treated to produce CR light inside the medium to be treated, especially a biological medium.
  • the tissue such that radiation dose can be maximized in the target area, while being minimized in skin and superficial dose.
  • Such targeting can be preferably done with appropriate collimation, using as an associated imaging system, a fan beam or cone beam x-ray system, or combinations thereof.
  • Other targeting mechanisms include axial and angular mA modulation of a Computed Tomograph (CT) system, and spectrum shaping through k-edge or crystalline filtering to "tune" the x-ray energy precisely to where the medium to be treated shows optimum CR light production or energy- converting or energy modulation agent in the medium shows maximum sensitivity.
  • CT Computed Tomograph
  • the initiation energy is capable of penetrating completely through the medium.
  • the phrase "capable of penetrating completely through the medium” is used to refer to energy capable of penetrating a container to any distance necessary to activate the activatable agent within the medium. It is not required that the energy applied actually pass completely through the medium, merely that it be capable of doing so in order to permit penetration to any desired distance to internally generate CR light in a vicinity of the activatable agent, such as by targeting the focus of the x-ray beam and thus the desired x-ray dose in the desired tissue.
  • the type of energy source chosen will depend on the medium itself.
  • psoralen and psoralen derivatives are of interest for many of the biological applications of this invention.
  • an initiation energy source can provide an energy that generates CR light to activate an activatable pharmaceutical agent to treat target cells within a subject.
  • the initiation energy is applied indirectly to the activatable pharmaceutical agent, preferably in proximity to the target cells.
  • the phrase “applied indirectly” or variants of this phrase, such as “applying indirectly”, “indirectly applies”, “indirectly applied”, “indirectly applying”, etc.), when referring to the application of the initiation energy, means the penetration by the initiation energy into the subject beneath the surface of the subject and to the activatable pharmaceutical agent within a subject.
  • subject is not intended to be limited to humans, but may also include animals, plants, or any suitable biological organism.
  • cell proliferation disorder refers to any condition where the growth rate of a population of cells is less than or greater than a desired rate under a given physiological state and conditions. Although, preferably, the proliferation rate that would be of interest for treatment purposes is faster than a desired rate, slower than desired rate conditions may also be treated by methods of the invention.
  • Exemplary cell proliferation disorders may include, but are not limited to, cancer, bacterial infection, immune rejection response of organ transplant, solid tumors, viral infection, autoimmune disorders (such as arthritis, lupus, inflammatory bowel disease, Sjogrens syndrome, multiple sclerosis) or a combination thereof, as well as aplastic conditions wherein cell proliferation is low relative to healthy cells, such as aplastic anemia.
  • Particularly preferred cell proliferation disorders for treatment using the present methods are cancer, staphylococcus aureus (particularly antibiotic resistant strains such as methicillin resistant staphylococcus aureus or MRSA), and autoimmune disorders.
  • an “activatable agent” is an agent that normally exists in an inactive state in the absence of an activation signal (e.g., one or more photons).
  • an activation signal e.g., one or more photons.
  • the agent is capable of producing a desired pharmacological, cellular, chemical, electrical, or mechanical effect in a medium (i.e. a predetermined change in the medium).
  • Signals that may be used to activate a corresponding agent may include, but are not limited to, photons of specific wavelengths (e.g. x-rays, ultraviolet, or visible light).
  • an activatable agent such as a photosensitizer, may be activated by UV-A radiation
  • an activatable agent such as a photosensitizer
  • UV-B or UV-C radiation may be activated by UV-B or UV-C radiation. Once activated, the agent in its active-state may then directly proceed to produce a predetermined change.
  • the activatable agent may effect changes that include, but are not limited to an increase in organism activity, a decrease in organism activity, apoptosis, and/or a redirection of metabolic pathways.
  • an “activatable pharmaceutical agent” is an agent that normally exists in an inactive state in the absence of an activation signal. When the agent is activated, it is capable of affecting the desired pharmacological effect on a target cell (i.e. preferably a predetermined cellular change).
  • a photoactive compound that achieves its pharmaceutical effect by binding (with mono adducts formation or cross links formation) to certain cellular structure in its active state may require physical proximity to the target cellular structure when the activation signal is delivered.
  • Some examples of activating conditions may include, but are not limited to, temperature, pH, location, state of the cell, presence or absence of co-factors. Selection of an activatable pharmaceutical agent greatly depends on a number of factors such as the desired cellular change, the desired form of activation, as well as the physical and biochemical constraints that may apply.
  • the activatable pharmaceutical agent When activated for example by CR light, the activatable pharmaceutical agent may affect cellular changes that include, but are not limited to, apoptosis, redirection of metabolic pathways, up-regulation of certain genes, down-regulation of certain genes, secretion of cytokines, alteration of cytokine receptor responses, production or modulation of reactive oxygen species or combinations thereof.
  • an activatable pharmaceutical agent may achieve its desired effect.
  • Such mechanisms may include direct action on a
  • a preferred direct action mechanism is by binding the agent to a critical cellular structure such as nuclear DNA, mRNA, rRNA, ribosome, mitochondrial DNA, or any other functionally important structures.
  • Indirect mechanisms may include modulation of or releasing metabolites upon activation to interfere with normal metabolic pathways, releasing chemical signals (e.g. agonists or antagonists) upon activation to alter the targeted cellular response, and other suitable biochemical or metabolic alterations.
  • the activatable pharmaceutical agent is capable of chemically binding to the DNA or mitochondrial at a therapeutically effective amount.
  • the activatable pharmaceutical agent preferably a photoactivatable agent, is exposed in situ to light internally generated for example by CR and/or an energy modulation agent.
  • An activatable agent may be a small molecule; a biological molecule such as a protein, a nucleic acid or lipid; a supramolecular assembly; a nanoparticle; a nanostructure, or
  • the activatable agent may be derived from a natural or synthetic origin. Any such molecular entity that may be activated by a suitable activation signal source to effect a predetermined cellular change may be advantageously employed in the invention.
  • Suitable photoactive agents include, but are not limited to: psoralens and psoralen derivatives, pyrene cholesteryloleate, acridine, porphyrin, fluorescein, rhodamine, 16- diazorcortisone, ethidium, transition metal complexes of bleomycin, transition metal complexes of deglycobleomycin, organoplatinum complexes, alloxazines such as 7,8-dimethyl-10-ribityl isoalloxazine (riboflavin), 7,8,10-trimethylisoalloxazine (lumiflavin), 7,8-dimethylalloxazine (lumichrome), isoalloxazine-adenine dinucleotide (flavine adenine dinucleotide [FAD], alloxazine mononucleotide (also known as flavine mononucleotide [FMN] and riboflavine-5- phosphat
  • alloxazine includes isoalloxazines.
  • Additional photoactive agents include, but are not limited to, carbene precursors, nitrene precursors, thio derivatives, benzophenones, and halogenated pyrimidines.
  • photochemistries are routinely employed to achieve protein-DNA photocross-links but none has been achieved using an indirect method as presented herein, for example where X-Ray radiation is converted to UV radiation to activate the species and achieve DNA photocross-links.
  • Endogenously-based derivatives include synthetically derived analogs and homologs of endogenous photoactivated molecules, which may have or lack lower (1 to 5 carbons) alkyl or halogen substituents of the photosensitizers from which they are derived, and which preserve the function and substantial non-toxicity. Endogenous molecules are inherently non-toxic and may not yield toxic photoproducts after photoradiation. The nature of the predetermined cellular change will depend on the desired
  • Exemplary cellular changes may include, but are not limited to, morphologic changes, apoptosis, necrosis, up-regulation of certain genes, down-regulation of certain genes, modulation of or secretion of cytokines, alteration of cytokine receptor responses, or a combination thereof.
  • the activatable pharmaceutical agent When activated for example by CR light, the activatable pharmaceutical agent may effect cellular changes that include, but are not limited to, apoptosis, redirection of metabolic pathways, up-regulation of certain genes, down-regulation of certain genes, secretion of cytokines, alteration of cytokine receptor responses, production of reactive oxygen species or combinations thereof.
  • a preferred method of treating a cell proliferation disorder of the invention administers a photoactivatable agent to a patient, stimulates the photoactivatable agent by CR light to induce cell damage (or kill), and generates an auto vaccine effect.
  • energy modulation agents may be included in the medium to be treated.
  • the energy modulation agents could be used to supplement the internally generated CR by downconvenon of x-rays into ultraviolet or visible light.
  • the energy modulation agents could be used to down-convert a portion of the CR spectrum or up-convert a portion of the CR spectrum.
  • an “energy modulation agent” refers to an agent that is capable of receiving an energy input from a source and then re-emitting a different energy to a receiving target.
  • Energy transfer among molecules may occur in a number of ways.
  • the form of energy may be electronic, thermal, electromagnetic, kinetic, or chemical in nature.
  • Energy may be transferred from one molecule to another (intermolecular transfer) or from one part of a molecule to another part of the same molecule (intramolecular transfer).
  • a modulation agent may receive electromagnetic energy and re-emit the energy in the form of thermal energy which otherwise contributes to heating the environment in vicinity of the light emission.
  • the energy modulation agents receive higher energy (e.g. x-ray) and re-emits in lower energy (e.g. UV-A).
  • Some modulation agents may have a very short energy retention time (on the order of fs, e.g.
  • the energy modulation agent materials can preferably include any materials that can absorb X ray and emit light in order to excite the PA molecule.
  • Quantum dots, semiconductor nanostructures and various materials related to quantum dots, semiconductor materials, etc. can be used as energy modulation agents.
  • Scintillator materials can be used as energy modulation agents.
  • Various scintillator materials can be used as energy modulation agents since they absorb X-ray and emit luminescence emission, which can be used to excite the PA system.
  • single crystals of molybdates can be excited by X-ray and emit luminescence around 400 nm [Mirkhin et al, Nuclear Instrum. Meth. In Physics Res. A, 486, 295 (2002, the entire contents of which are incorporated herein by referencey.
  • CdS CsCl
  • XEOL materials such as lanthanides or rare earth materials can be used as energy modulation agents.
  • Suitable energy modulation agents include, but are not limited to, a phosphor, a scintillator, a biocompatible fluorescing metal nanoparticle, fluorescing dye molecule, gold nanoparticle, quantum dots, such as a water soluble quantum dot encapsulated by
  • polyamidoamine dendrimers a luciferase, a biocompatible phosphorescent molecule, a combined electromagnetic energy harvester molecule, an up-converter, a lanthanide chelate capable of intense luminescence, metals (gold, silver, etc); semiconductor materials; materials that exhibit X-ray excited luminescence (XEOL); organic solids, metal complexes, inorganic solids, crystals, rare earth materials (lanthanides), polymers, and materials that exhibit excitonic properties.
  • XEOL X-ray excited luminescence
  • the energy modulation agents include down converters (such as for example phosphors which can convert x-ray or other high energy photon or particle into visible light. These down converters when used in combination can activate a variety of UV- stimulated photoreactions as well as activate any visible light activated reactions.
  • luminescing particles can include gold particles (such as for example the nanoparticles of gold), BaFBrEu particles, CdSe particles, Y 2 0 3 :Eu 3+ particles, and/or other known stimulated luminescent materials such as for example ZnS: Mn 2+ ; ZnS: Mn 2+ ,Yb 3+ , Y 2 0 3 : Eu 3+ ; BaFBr:Tb 3+ ; and YF 3 :Tb 3 +. More specific examples of the
  • downconverters include, but are not limited to: BaFCl:Eu 2+ , BaS0 4 " :Eu 2+ , LaOBr:Tm 3+ , YTa0 4 , YTa0 4 :Nb, CaW0 4 , LaOBr:Tb 3+ , Y 2 0 2 S:Tb 3+ , ZnS:Ag, (Zn,Cd)S:Ag, Gd 2 0 2 S:Tb 3+ , La 2 0 2 S:Tb 3+ .
  • a downconverting energy modulation agent can comprise inorganic particulates selected from the group consisting of: metal oxides; metal sulfides; doped metal oxides; and mixed metal chalcogenides.
  • the inorganic particulates selected from the group consisting of: metal oxides; metal sulfides; doped metal oxides; and mixed metal chalcogenides.
  • downconverting material can comprise at least one of Y 2 0 3 , Y 2 0 2 S, NaYF , NaYbF , YAG, YAP, Nd 2 0 3 , LaF 3 , LaCl 3 , La 2 0 3 , Ti0 2 , LuP0 4 , YV0 4 , YbF 3 , YF 3 , Na-doped YbF 3 , ZnS; ZnSe; MgS; CaS; CaW0 4 , CaSi0 2 :Pb, and alkali lead silicate including compositions of Si0 2 , B 2 0 3 , Na 2 0, K 2 0, PbO, MgO, or Ag, and combinations or alloys or layers thereof.
  • the downconverting material can include a dopant including at least one of Er, Eu, Yb, Tm, Nd, Mn Tb, Ce, Y, U, Pr, La, Gd and other rare-earth species or a combination thereof.
  • the dopant can be included at a concentration of 0.01%-50% by mol concentration.
  • the downconverting energy modulation agent can comprise materials such as ZnSeS:Cu, Ag, Ce, Tb; CaS: Ce,Sm; La 2 0 2 S:Tb; Y 2 0 2 S:Tb;
  • the downconverting material can comprise phosphors such as ZnS:Ag and ZnS:Cu, Pb.
  • the downconverting material can be alloys of the ZnSeS family doped with other metals.
  • suitable materials include ZnSe x S y :Cu, Ag, Ce, Tb, where the following x, y values and intermediate values are acceptable: x:y; respectively 0: 1; 0.1 :0.9; 0.2:0.8; 0.3 :0.7; 0.4:0.6; 0.5:0.5; 0.6:0.4; 0.7:0.3; 0.8:0.2; 0.9:0.1; and 1.0:0.0.
  • the downconverting energy modulation agent can be materials such as sodium yttrium fluoride (NaYF 4 ), lanthanum fluoride (LaF 3 ), lanthanum oxysulfide (La 2 0 2 S), yttrium oxysulfide (Y 2 0 2 S), yttrium fluoride (YF 3 ), yttrium gallate, yttrium aluminum garnet (YAG), gadolinium fluoride (GdF 3 ), barium yttrium fluoride (BaYF 5 , BaY 2 F 8 ), gadolinium oxysulfide (Gd 2 0 2 S), calcium tungstate (CaW0 4 ), yttrium oxide:terbium (Yt 2 0 3 Tb), gadolinium oxysulphide:europium (Gd 2 0 2 S:Eu), lanthanum oxysulphide:europium (La 2 0 2 S)
  • the downconverting energy modulation agent can be near-infrared (NIR) downconversion (DC) phosphors such as KSrP0 4 :Eu 2+ , Pr 3+ , or NaGdF :Eu or Zn 2 Si0 4 :Tb 3+ ,Yb 3+ or p-NaGdF 4 co-doped with Ce 3+ and Tb 3+ ions or Gd 2 0 2 S:Tm or
  • NIR near-infrared
  • DC downconversion
  • BaYF 5 :Eu 3+ or other down converters which emit NIR from visible or UV light exposure (as in a cascade from x-ray to UV to NIR) or which emit NIR directly after x-ray or e-beam exposure.
  • some of the phosphors noted above can absorb in the 390 to 410 nm range and then in turn down convert the CR radiation into red shifted emissions for activation in the visible.
  • the excitation wavelength can be between 300 nm and 450 nm, and the emission can be centered around 650 nm as is the case for 6MgO.
  • an up-converting energy modulation agent can be used which is activated by for example an infrared or near infrared source such as a laser.
  • the up- converting energy modulation agent can be at least one of Y 2 O 3 , Y 2 O 2 S, NaYF 4 , NaYbF 4 , YAG, YAP, Nd 2 0 3 , LaF 3 , LaCl 3 , La 2 0 3 , Ti0 2 , LuP0 4 , YV0 4 , YbF 3 , YF 3 , Na-doped YbF 3 , or Si0 2 or alloys or layers thereof.
  • the luminescing particles (down converters, mixtures of down converters, up converters, mixtures of up converters, and combinations thereof) of the invention described here can be coated with insulator materials such as for example silica which will reduce the likelihood of any chemical interaction between the luminescing particles and the medium.
  • insulator materials such as for example silica which will reduce the likelihood of any chemical interaction between the luminescing particles and the medium.
  • silica for biological applications of inorganic nanoparticles, one of the major limiting factors is their toxicity.
  • nanoparticles are more or less toxic.
  • nanoparticles with toxicity as low as possible are desirable or else the nanoparticles have to remain separated from the medium.
  • Pure T1O 2 , ZnO, and Fe 2 0 3 are biocompatible.
  • CdTe and CdSe are toxic, while ZnS, CaS, BaS, SrS and Y 2 0 3 are less toxic.
  • the toxicity of nanoparticles can result from their inorganic stabilizers, such as TGA, or from dopants such as Eu 2+ , Cr 3+ or Nd 3+ .
  • Suitable energy modulation agents which would seem the most biocompatible are zinc sulfide, ZnS:Mn 2+ , ferric oxide, titanium oxide, zinc oxide, zinc oxide containing small amounts of A1 2 0 3 and Agl nanoclusters encapsulated in zeolite.
  • lanthanum and gadolinium oxyhalides activated with thulium
  • Er 3+ doped BaTi0 3 nanoparticles Yb 3+ doped CsMnCl 3 and RbMnCl 3
  • BaFBr:Eu 2+ nanoparticles cesium iodide, bismuth germanate, cadmium tungstate, and CsBr doped with divalent Eu.
  • the following luminescent polymers are also suitable as energy modulation agents: poly(phenylene ethynylene), poly(phenylene vinylene), poly(p-phenylene), poly(thiophene), poly(pyridyl vinylene), poly(pyrrole), poly(acetylene), poly(vinyl carbazole), poly(fluorenes), and the like, as well as copolymers and/or derivatives thereof.
  • 7,008,559 (the entire contents of which are incorporated herein by reference) describes the upconversion performance of ZnS where excitation at 767 nm produces emission in the visible range.
  • the materials described in U.S. Pat. No. 7,008,559 including the ZnS as well as Er doped BaTi0 3 nanoparticles and Yb 3+ doped CsMnCl 3 are suitable in various embodiments of the invention.
  • the up converters can be used in combination with the down converters (or mixtures of down converters) or in combination with various up converters.
  • Various up converters suitable for this invention include CdTe, CdSe, ZnO, CdS, Y 2 O 3 , MgS, CaS, SrS and BaS.
  • Such up conversion materials may be any one of the following up conversion materials.
  • sulfide, telluride, selenide, and oxide semiconductors and their nanoparticles such as Zni -x Mn x S y , Zni -x Mn x Se y , Zn ⁇ x Mn x Tey, Cdi -x MnS y , Cdi -x Mn x Se y , Cdi -x Mn x Tey, Pbi -x Mn x S y , Pbi -x Mn x Se y , Pbi -x Mn x Te y , Mgi.
  • Additional conversion materials include insulating and nonconducting materials such as BaF 2 , BaFBr, and BaTi0 3 , to name but a few exemplary compounds.
  • Transition and rare earth ion co-doped semiconductors suitable for the invention include sulfide, telluride, selenide and oxide semiconductors and their
  • nanoparticles such as ZnS; Mn; Er; ZnSe; Mn, Er; MgS; Mn, Er; CaS; Mn, Er; ZnS; Mn, Yb; ZnSe; Mn,Yb; MgS; Mn, Yb; CaS; Mn,Yb etc., and their complex compounds: (Mi -z N z )i.
  • nanoparticles such as ZnS:Tb 3+ , Er 3+ ; ZnS:Tb 3+ ; Y 2 0 3 :Tb 3+ ; Y 2 0 3 :Tb 3+ , Er3 + ; ZnS:Mn 2+ ; ZnS:Mn,Er 3+ are known in the art to have two functions, capable of functioning for both down-conversion luminescence and upconversion luminescence.
  • one embodiment of the invention described here coats these nanoparticles with silica.
  • Silica is used as a coating material in a wide range of industrial colloid products from paints and magnetic fluids to high-quality paper coatings. Further, silica is both chemically and biologically inert and also is optically transparent.
  • Other coatings suitable for this invention include a polymethyl methacrylate (PMMA) coating and an ethyl-cellulose coating.
  • the energy modulation agent can be used alone or as a series of two or more energy modulation agents wherein the energy modulation agents provide an energy cascade from the light of the phosphors or scintillators.
  • the first energy modulation agent in the cascade will absorb the CR, convert it to a different energy which is then absorbed by the second energy modulation in the cascade, and so forth until the end of the cascade is reached with the final energy modulation agent in the cascade emitting the energy necessary to activate the activatable pharmaceutical agent.
  • a chemical reaction cascade can be triggered.
  • the CR can activate a chemical which in turn can activate a bio-therapeutic in parallel to or independent of a photonic pathway.
  • the energy modulation agents or the photoactivatable agent may further be coupled to a carrier for cellular targeting purposes.
  • a UV-A emitting energy modulation agent may be concentrated in the tumor site by physical insertion or by conjugating the UV-A emitting energy modulation agent with a tumor specific carrier, such as an antibody, nucleic acid, peptide, a lipid, chitin or chitin-derivative, a chelate, a surface cell receptor, molecular imprints, aptamers, or other functionalized carrier that is capable of concentrating the UV-A emitting source in a specific target tumor.
  • a tumor specific carrier such as an antibody, nucleic acid, peptide, a lipid, chitin or chitin-derivative, a chelate, a surface cell receptor, molecular imprints, aptamers, or other functionalized carrier that is capable of concentrating the UV-A emitting source in a specific target tumor.
  • a method in accordance with one embodiment of the invention utilizes the principle of energy transfer to and among molecular agents to control delivery and activation of cellular changes by irradiation such that delivery of the desired effect is more intensified, precise, and effective than the conventional techniques.
  • At least one energy modulation agent can be administered to the subject which adsorbs, intensifies or modifies the CR into an energy that effects a predetermined cellular change in the target structure.
  • the energy modulation agent may be located around, on, or in the target structure. Further, the energy modulation agent can transform CR into a photonic energy that effects a predetermined change in the target structure. In one embodiment, the energy modulation agent decreases the wavelength of the CR (down convert).
  • the energy modulation agent can increase the wavelength of the CR (up convert).
  • the energy modulation agent is one or more members selected from a biocompatible fluorescing metal nanoparticle, fluorescing metal oxide nanoparticle, fluorescing dye molecule, gold nanoparticle, silver nanoparticle, gold-coated silver nanoparticle, a water soluble quantum dot encapsulated by polyamidoamine dendrimers, a luciferase, a biocompatible phosphorescent molecule, a combined electromagnetic energy harvester molecule, and a lanthanide chelate exhibiting intense luminescence.
  • photoactivatable agents may be stimulated by light from CR and/or light from the energy modulation agents, leading to subsequent irradiation, resonance energy transfer, exciton migration, electron injection, or chemical reaction, to an activated energy state that is capable of effecting the predetermined cellular change desired.
  • the photoactivatable agent upon activation, binds to DNA or RNA or other structures in a cell.
  • the activated energy state of the agent is capable of causing damage to cells, inducing apoptosis.
  • the mechanism of apoptosis is associated with an enhanced immune response that reduces the growth rate of cell proliferation disorders and may shrink solid tumors, depending on the state of the patient's immune system, concentration of the agent in the tumor, sensitivity of the agent to stimulation, and length of stimulation.
  • This excess VR energy is released as thermal energy to the surrounding medium.
  • the molecule deactivates rapidly to the isoenergetic vibrational level of a lower electronic state such as S n -l vian internal conversion (IC) process.
  • IC processes are transitions between states of the same multiplicity.
  • the molecule subsequently deactivates to the lowest vibronic levels of via VR process.
  • the molecule deactivates rapidly to the ground state Si.. This process results in excess VR and IC energy released as thermal energy to the surrounding medium leading to the overheating of the local environment surrounding the light absorbing drug molecules. The heat produced results in local cell or tissue destruction.
  • the light absorbing species include natural chromophores in tissue or exogenous dye compounds such as indocyanine green, naphthalocyanines, and porphyrins coordinated with transition metals and metallic nanoparticles and nanoshells of metals. Natural chromophores, however, suffer from very low absorption.
  • the choice of the exogenous photothermal agents is made on the basis of their strong absorption cross sections and highly efficient light-to-heat conversion. This feature greatly minimizes the amount of energy needed to induce local damage of the diseased cells, making therapy method less invasive.
  • "microwave upconversion" can be used to supplement the CR-driven activation.
  • 20150283392 describes up and down conversion systems for production of emitted light from various energy sources including radio frequency, microwave energy and magnetic induction sources for upconversion.
  • the systems described therein including the plasma-gas containing capsules can be used here.
  • This up converting "capsule" structure once in the patient or subject can be exposed to a combination of microwave energy and/or high magnetic field in order to produce light (for example UV, VIS, or IR light or a combination thereof) from the plasma gas in the gas- filled container to activate a photoactivatable drug such as psoralen.
  • light for example UV, VIS, or IR light or a combination thereof
  • the containers or capsules can include materials ("secondary electron emitters") which, upon exposure to x-rays (as from the CR radiationO would assist in the generation of a gaseous plasma in the capsule.
  • the '392 application describes that, when inner walls of the gas containers are coated with a material that would generate secondary electrons upon X-Ray exposure, the secondary electrons enter into high energy excitations due to radio frequency RF and/or microwave MW energy, thereby producing lower power plasma ignitions. Higher energy excitations are possible in the presence of a magnetic field.
  • the energy source can be an internal source of radiation, often referred to as Brachytherapy.
  • Brachytherapy involves placing radiation sources as close as possible to the tumor site. Sometimes, these sources may be inserted directly into the tumor.
  • the radioactive sources or isotopes are in the form of wires, seeds (or molds), or rods. This technique is commonly used in treating cancers of the cervix, uterus, vagina, rectum, eye, and certain head and neck cancers. It is also occasionally used to treat cancers of the breast, brain, skin, anus, esophagus, lung, bladder, and prostate.
  • brachytherapy There are several types of brachytherapy characterized by different methods of placing radiation inside the body: interstitial brachytherapy, intracavitary brachytherapy, intraluminal radiation therapy, and radioactively tagged molecules given intravenously.
  • brachytherapy can be combined with external beam radiation therapy to generate radiation around the treatment area with a boost of radiation delivered to the tumor area itself.
  • the selection of radioactive seeds is known to those skilled in the art and typically based upon the anatomy of the treatment area, the energy of emission and the duration of treatment. In the present invention, these seeds can be used as the source of CR radiation or as a supplement to CR radiation.
  • the photoactive drug molecules can be given to a patient by oral ingestion, skin application, or by intravenous injection.
  • the photoactive drug molecules drugs travel through the blood stream inside the body towards the targeted tumor (either via passive or active targeting strategies).
  • the invention treatment may also be used for inducing an auto vaccine effect for malignant cells, including those in solid tumors.
  • any rapidly dividing cells or stem cells may be damaged by a systemic treatment, then it may be preferable to direct the stimulating energy directly toward the tumor, preventing damage to most normal, healthy cells or stem cells by avoiding photoactivation or resonant energy transfer of the photoactivatable agent.
  • a treatment may be applied that slows or pauses mitosis.
  • a treatment is capable of slowing the division of rapidly dividing healthy cells or stem cells during the treatment, without pausing mitosis of cancerous cells.
  • a blocking agent is administered preferentially to malignant cells prior to administering the treatment that slows mitosis.
  • an aggressive cell proliferation disorder can be treated with CR- activation of the photoactivatable agent which has a much higher rate of mitosis, which leads to selective destruction of a disproportionate share of the malignant cells during even a
  • Stem cells and healthy cells may be spared from wholesale programmed cell death, even if exposed to photoactivated agents, provided that such
  • photoactivated agents degenerate from the excited state to a lower energy state prior to binding, mitosis or other mechanisms for creating damage to the cells of a substantial fraction of the healthy stem cells.
  • an auto-immune response may not necessarily have to be induced.
  • methods in accordance with the invention may further include adding an additive to alleviate treatment side-effects.
  • additives may include, but are not limited to, antioxidants, adjuvant, or combinations thereof.
  • psoralen is used as the activatable pharmaceutical agent
  • UV-A from CR is used as the activating energy
  • antioxidants are added to reduce the unwanted side-effects of irradiation.
  • compositions suitable for administration can be incorporated into pharmaceutical compositions suitable for administration.
  • Such compositions typically comprise the activatable pharmaceutical agent and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition also comprises at least one additive having a complementary therapeutic or diagnostic effect, wherein the additive is one selected from an antioxidant, an adjuvant, or a combination thereof.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such medical agents for pharmaceutically active substances is well known in the art.
  • compositions can be used in the compositions.
  • Supplementary active compounds can also be incorporated into the compositions. Modifications can be made to the compound of the invention to affect solubility or clearance of the compound. These molecules may also be synthesized with D- amino acids to increase resistance to enzymatic degradation. If necessary, the activatable pharmaceutical agent can be co-administered with a solubilizing agent, such as cyclodextran.
  • a pharmaceutical composition of the invention can be formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, rectal administration, and direct injection into the affected area, such as direct injection into a tumor.
  • subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, or phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound (activatable drug and/or energy modulation agent) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • Oral compositions of the drug and/or energy modulation agent can generally include an inert diluent or an edible carrier.
  • the oral compositions can be enclosed in gelatin capsules or compressed into tablets.
  • the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules.
  • Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
  • Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration of the activatable drug and/or energy modulation agent can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds drug and/or energy modulation agent
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • suppositories e.g., with conventional suppository bases such as cocoa butter and other glycerides
  • retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
  • Liposomal suspensions including liposomes targeted to infected cells with monoclonal antibodies to viral antigens
  • These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811, the entire contents of which are incorporated herein by reference.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • compositions can be included in a container, pack, kit or dispenser together with instructions for administration.
  • Methods of administering agents are not limited to the conventional means such as injection or oral infusion, but include more advanced and complex forms of energy transfer.
  • genetically engineered cells that carry and express energy modulation agents may be used.
  • Cells from the host may be transfected with genetically engineered vectors that express bioluminescent agents. Transfection may be accomplished via in situ gene therapy techniques such as injection of viral vectors or gene guns, or may be performed ex vivo by removing a sample of the host's cells and then returning to the host upon successful transfection. Such transfected cells may be inserted or otherwise targeted at the site where diseased cells are located.
  • the order of administering the different agents is not particularly limited. It will be appreciated that different combinations of ordering may be advantageously employed depending on factors such as the absorption rate of the agents, the localization and molecular trafficking properties of the agents, and other pharmacokinetics or pharmacodynamics considerations.
  • CR Cherenkov radiation
  • Another object of the invention is to treat a condition by CR-activation, disorder or disease in a subject.
  • exemplary conditions, disorders or diseases may include, but are not limited to, cancer, autoimmune diseases, cardiac ablasion (e.g., cardiac arrhythmiand atrial fibrillation), photoangioplastic conditions (e.g., de novo atherosclerosis, restinosis), intimal hyperplasia, arteriovenous fistula, macular degeneration, psoriasis, acne, hopeciareata, portwine spots, hair removal, rheumatoid and inflammatory arthritis, joint conditions, lymph node conditions, and cognitive and behavioral conditions.
  • cardiac ablasion e.g., cardiac arrhythmiand atrial fibrillation
  • photoangioplastic conditions e.g., de novo atherosclerosis, restinosis
  • intimal hyperplasia arteriovenous fistula, macular degeneration, psoriasis, acne, hopeciareat
  • a disease or condition refers to a condition, disorder or disease that may include, but are not limited to, cancer, soft and bone tissue injury, chronic pain, wound healing, nerve regeneration, viral and bacterial infections, fat deposits (liposuction), varicose veins, enlarged prostate, retinal injuries and other ocular diseases, Parkinson's disease, and behavioral, perceptional and cognitive disorders.
  • exemplary conditions also may include nerve (brain) imaging and stimulation, a direct control of brain cell activity with light, control of cell death (apoptosis), and alteration of cell growth and division.
  • target structure refers to an eukaryotic cell, prokaryotic cell, a subcellular structure, such as a cell membrane, a nuclear membrane, cell nucleus, nucleic acid, mitochondria, ribosome, or other cellular organelle or component, an extracellular structure, virus or prion, and combinations thereof.
  • predetermined cellular change induced by the CR radiation will depend on the desired pharmaceutical outcome.
  • exemplary cellular changes may include, but are not limited to, apoptosis, necrosis, up-regulation of certain genes, down-regulation of certain genes, secretion of cytokines, alteration of cytokine receptor responses, regulation of cytochrome c oxidase and flavoproteins, activation of mitochondria, stimulation antioxidant protective pathway, modulation of cell growth and division, alteration of firing pattern of nerves, alteration of redox properties, generation of reactive oxygen species, modulation of the activity, quantity, or number of intracellular components in a cell, modulation of the activity, quantity, or number of extracellular components produced by, excreted by, or associated with a cell, or a combination thereof.
  • Predetermined cellular changes may or may not result in destruction or inactivation of the target structure.
  • the photoactivatable agent can be a photocaged complex having an active agent contained within a photocage.
  • the active agent can bulked up with other molecules that prevent it from binding to specific targets, thus masking its activity.
  • the photocage complex is photoactivated by CR and/or light from the energy modulation agents, the bulk falls off, exposing the active agent.
  • the photocage molecules can be photoactive (i.e. when photoactivated, they are caused to dissociate from the photocage complex, thus exposing the active agent within), or the active agent can be the photoactivatable agent (which when photoactivated causes the photocage to fall off), or both the photocage and the active agent are photoactivated, with the same or different wavelengths.
  • a toxic chemotherapeutic agent can be photocaged, which will reduce the systemic toxicity when delivered. Once the agent is concentrated in the tumor, the agent is irradiated with an activation energy. This causes the "cage” to fall off, leaving a cytotoxic agent in the tumor cell.
  • Suitable photocages include those disclosed by Young and Deiters in "Photochemical Control of
  • the use of CR light for uncaging a compound or agent is used for elucidation of neuron functions and imaging, for example, two-photon glutamine uncaging (Harvey CD, et al., Nature, 450: 1195-1202 (2007); Eder M, et al., Rev. Neurosci., 15: 167-183 (2004)).
  • Other signaling molecules can be released by UV light stimulation, e.g., GABA, secondary messengers (e.g., Ca 2+ and Mg 2+ ), carbachol, capsaicin, and ATP (Zhang F., et al., 2006). Chemical modifications of ion channels and receptors may be carried out to render them light-responsive.
  • Ca 2+ is involved in controlling fertilization, differentiation, proliferation, apoptosis, synaptic plasticity, memory, and developing axons.
  • Ca 2+ waves can be induced by UV irradiation (single-photon absorption) and NIR irradiation (two-photon absorption) by releasing caged Ca 2+ , an extracellular purinergic messenger InsP3 (Braet K., et al., Cell Calcium, 33 :37-48 (2003)), or ion channel ligands (Zhang F., et al., 2006).
  • a light-sensitive protein is introduced into cells or live subjects via number of techniques including
  • lentiviral technology provides a convenient combination a conventional combination of stable long-term expression, ease of high-titer vector production and low immunogenicity.
  • the light-sensitive protein may be, for example, channelrhodopsin-2 (ChR2) and chloride pump halorhodopsin (NpHR).
  • ChR2 channelrhodopsin-2
  • NpHR chloride pump halorhodopsin
  • the light protein encoding gene(s) along with a cell-specific promoter can be incorporated into the lentiviral vector or other vector providing delivery of the light-sensitive protein encoding gene into a target cell.
  • ChR2 containing a light sensor and a cation channel provides electrical stimulation of appropriate speed and magnitude to activate neuronal spike firing, when the cells harboring Ch2R are pulsed with light.
  • a lanthanide chelate capable of intense luminescence and excited by CR light can be used.
  • a lanthanide chelator may be covalently joined to a coumarin or coumarin derivative or a quinolone or quinolone-derivative sensitizer.
  • Sensitizers may be a 2- or 4-quinolone, a 2- or 4- coumarin, or derivatives or combinations of these examples.
  • a carbostyril 124 (7-amino-4-methyl-2-quinolone), a coumarin 120 (7-amino-4-methyl-2- coumarin), a coumarin 124 (7-amino-4-(trifluoromethyl)-2-coumarin),
  • Chelates may be selected to form high affinity complexes with lanthanides, such as terbium or europium, through chelator groups, such as DTPA. Such chelates may be coupled to any of a wide variety of probes or carriers, and may be used for resonance energy transfer to a psoralen or psoralen-derivative, such as 8-MOP, or other photoactive molecules capable of binding DNA.
  • lanthanides such as terbium or europium
  • chelator groups such as DTPA.
  • Such chelates may be coupled to any of a wide variety of probes or carriers, and may be used for resonance energy transfer to a psoralen or psoralen-derivative, such as 8-MOP, or other photoactive molecules capable of binding DNA.
  • the lanthanide chelate is localized at the site of the disease using an appropriate carrier molecule, particle or polymer, and a source of electromagnetic energy is introduced by minimally invasive procedures to irradiate the target structure, after exposure to the lanthanide chelate and a photoactive molecule.
  • a biocompatible, endogenous fluorophore emitter can be selected to stimulate resonance energy transfer from the CR light to a photoactivatable molecule.
  • a biocompatible emitter e.g. the phosphors or scintillators
  • One or more halogen atoms may be added to any cyclic ring structure capable of intercalation between the stacked nucleotide bases in a nucleic acid (either DNA or RNA) to confer new photoactive properties to the intercalator.
  • any intercalating molecule may be selectively modified by halogenation or addition of non-hydrogen bonding ionic substituents to impart advantages in its reaction photochemistry and its competitive binding affinity for nucleic acids over cell membranes or charged proteins, as is known in the art.
  • the initiation energy source may be a linear accelerator equipped with at least kV image guided computer-control capability to deliver a precisely calibrated beam of radiation to a pre-selected coordinate.
  • linear accelerators include the SMARTBEAMTM EVIRT (intensity modulated radiation therapy) system (from Varian Medical Systems, Inc., Palo Alto, California) or Varian OBI technology (OBI stands for "On-board Imaging", and is found on many commercial models of Varian machines).
  • the initiation energy source may be commercially available components of X-ray machines or non-medical X-ray machines. X-ray machines that produce from 10 to 150 keV X-rays are readily available in the marketplace. For instance, the General Electric
  • DEFINIUM series or the Siemens MULTIX series are two non-limiting examples of typical X- ray machines designed for the medical industry, while the EAGLE PACK series from Smith Detection is an example of a non-medical X-ray machine.
  • Another suitable commercially available device is the SIEMENS DEFINITION FLASH, (a CT system), by Siemens Medical Solutions. As such, the invention is capable of performing its desired function when used in conjunction with commercial X-ray equipment.
  • Current medical linear accelerators produce high energy electron and photon beams in the energy range 6-20 MeV.
  • the threshold energy for Cherenkov production is -0.8 MeV, with higher energies producing more Cherenkov radiation inside the medium.
  • FIG. 3 illustrates a system according to one exemplary embodiment of the invention.
  • an exemplary system according to one embodiment of the invention may have an initiation energy source 1 directed at the subject 4.
  • An activatable pharmaceutical agent 2 and an energy modulation agent 3 can be administered to the subject 4.
  • the initiation energy source may additionally be controlled by a computer system 5 that is capable of directing the delivery of the initiation energy (e.g., X-rays).
  • dose calculation and robotic manipulation devices (such as the
  • CYBER-KNIFE robotic radiosurgery system available from Accuray, or similar types of devices may also be included in the system to adjust the distance between the initiation energy source 1 and the subject 4 and/or to adjust the energy and/or dose of the initiation energy source such that the x-rays incident on the target site are within an energy band bounded by a lower energy threshold capable of inducing desirable reactions and an upper energy threshold leading to denaturization of the medium. Further refinements in the x-ray energy and dose can be had by adjusting the distance to the subject 5 or the intervening materials between the target site and the initiation energy source 1.
  • a computer implemented system for designing and selecting suitable combinations of initiation energy source, energy transfer agent, and activatable pharmaceutical agent comprising:
  • CPU central processing unit
  • a database of excitable compounds e.g., a database of excitable compounds; a first computation module for identifying and designing an excitable compound (e.g., a photoactivatable drug) that is capable of binding with a target cellular structure or component; and
  • an excitable compound e.g., a photoactivatable drug
  • a second computation module predicting the initiation energy and dose producing the CR light needed for excitation of the excitable compound
  • system upon selection of a target cellular structure or component, computes an excitable compound that is capable of interacting with the target structure.
  • the computer-implemented system may have a central processing unit (CPU) connected to a memory unit, configured such that the CPU is capable of processing user inputs and selecting a combination of initiation source (or initiation energies or distances), activatable pharmaceutical agent, and energy modulation or energy transfer agents for use in a method of the invention.
  • CPU central processing unit
  • memory unit configured such that the CPU is capable of processing user inputs and selecting a combination of initiation source (or initiation energies or distances), activatable pharmaceutical agent, and energy modulation or energy transfer agents for use in a method of the invention.
  • the computer-implemented system includes (or is programmed to act as) an x-ray source (or high energy source such as an electron beam) control device configured to calculate an x-ray (radiation) exposure condition including a distance between the initiation energy source 1 and the subject 4 and the energy band bounded by the above-noted lower energy threshold capable of inducing desirable reactions and the above-noted upper energy threshold leading to denaturization of the medium.
  • the control device operates the x-ray or high energy source (the initiation energy source 1) within the exposure condition to provide a requisite energy and/or dose of x-rays to the subject or a target site of the subject.
  • the computer system 5 shown in FIG. 3 can include a central processing unit (CPU) having a storage medium on which is provided: a database of excitable compounds, a first computation module for a photoactivatable agent or energy transfer agent, and a second computation module predicting the requisite energy flux needed to sufficiently activate the energy transfer agent or photoactivatable agent.
  • CPU central processing unit
  • an exemplary system may have an initiation energy source 1 directed at a biological medium 4.
  • Activatable agents 2 and an energy modulation agents 3 are dispersed throughout the biological medium 4.
  • the initiation energy source 1 may additionally be connected via a network 8 to a computer system 5 capable of directing the delivery of the initiation energy.
  • the energy modulation agents 3 are encapsulated energy modulation agents 6, depicted in FIG. 4 as silica encased energy modulation agents.
  • initiation energy 7 in the form of radiation from the initiation energy source 1 permeated throughout the biological medium 4.
  • the initiation energy source 1 can be an external energy source or an energy source located at least partially in the biological medium 4.
  • activatable agents 2 and/or the energy modulation agents 3 can include plasmonics agents which enhance either the applied energy or the energy emitted from the energy modulation agents 3 so as to directly or indirectly produce a change in the biological medium.
  • FIG. 5 illustrates a computer system 1201 for implementing various embodiments of the invention.
  • the computer system 1201 may be used as the computer system 5 to perform any or all of the functions described above.
  • the computer system 1201 includes a bus 1202 or other communication mechanism for communicating information, and a processor 1203 coupled with the bus 1202 for processing the information.
  • the computer system 1201 also includes a main memory 1204, such as a random access memory (RAM) or other dynamic storage device (e.g., dynamic RAM (DRAM), static RAM (SRAM), and synchronous DRAM (SDRAM)), coupled to the bus 1202 for storing information and instructions to be executed by processor 1203.
  • the main memory 1204 may be used for storing temporary variables or other intermediate information during the execution of instructions by the processor 1203.
  • the computer system 1201 further includes a read only memory (ROM) 1205 or other static storage device (e.g., programmable read only memory (PROM), erasable PROM (EPROM), and electrically erasable PROM (EEPROM)) coupled to the bus 1202 for storing static information and instructions for the processor 1203.
  • ROM read only memory
  • PROM programmable read only memory
  • EPROM erasable PROM
  • EEPROM electrically erasable PROM
  • the computer system 1201 also includes a disk controller 1206 coupled to the bus 1202 to control one or more storage devices for storing information and instructions, such as a magnetic hard disk 1207, and a removable media drive 1208 (e.g., floppy disk drive, read-only compact disc drive, read/write compact disc drive, compact disc jukebox, tape drive, and removable magneto-optical drive).
  • a removable media drive 1208 e.g., floppy disk drive, read-only compact disc drive, read/write compact disc drive, compact disc jukebox, tape drive, and removable magneto-optical drive.
  • the storage devices may be added to the computer system 1201 using an appropriate device interface (e.g., small computer system interface (SCSI), integrated device electronics (IDE), enhanced-IDE (E-IDE), direct memory access (DMA), or ultra-DMA).
  • SCSI small computer system interface
  • IDE integrated device electronics
  • E-IDE enhanced-IDE
  • DMA direct memory access
  • ultra-DMA ultra-DMA
  • the computer system 1201 may also include special purpose logic devices (e.g., application specific integrated circuits (ASICs)) or configurable logic devices (e.g., simple programmable logic devices (SPLDs), complex programmable logic devices (CPLDs), and field programmable gate arrays (FPGAs)).
  • ASICs application specific integrated circuits
  • SPLDs simple programmable logic devices
  • CPLDs complex programmable logic devices
  • FPGAs field programmable gate arrays
  • the computer system 1201 may also include a display controller 1209 coupled to the bus
  • the computer 1202 to control a display, such as a cathode ray tube (CRT), for displaying information to a computer user.
  • the computer system includes input devices, such as a keyboard and a pointing device, for interacting with a computer user and providing information to the processor 1203.
  • the pointing device for example, may be a mouse, a trackball, or a pointing stick for
  • a printer may provide printed listings of data stored and/or generated by the computer system 1201.
  • the computer system 1201 performs a portion or all of the processing steps (or functions) of this invention in response to the processor 1203 executing one or more sequences of one or more instructions contained in a memory, such as the main memory 1204. Such instructions may be read into the main memory 1204 from another computer readable medium, such as a hard disk 1207 or a removable media drive 1208.
  • processors in a multi -processing arrangement may also be employed to execute the sequences of instructions contained in main memory 1204.
  • hard-wired circuitry may be used in place of or in combination with software instructions. Thus, embodiments are not limited to any specific combination of hardware circuitry and software.
  • the computer system 1201 includes at least one computer readable medium or memory for holding instructions programmed according to the teachings of the invention and for containing data structures, tables, records, or other data described herein.
  • Examples of computer readable media are compact discs, hard disks, floppy disks, tape, magneto-optical disks, PROMs (EPROM, EEPROM, flash EPROM), DRAM, SRAM, SDRAM, or any other magnetic medium, compact discs (e.g., CD-ROM), or any other optical medium, or other physical medium with patterns of holes, a carrier wave (described below), or any other medium from which a computer can read.
  • the invention Stored on any one or on a combination of computer readable media, the invention includes software for controlling the computer system 1201, for driving a device or devices for implementing the invention, and for enabling the computer system 1201 to interact with a human user.
  • software may include, but is not limited to, device drivers, operating systems, development tools, and applications software.
  • Such computer readable media further includes the computer program product of the invention for performing all or a portion (if processing is distributed) of the processing performed in implementing the invention.
  • the computer code devices of the invention may be any interpretable or executable code mechanism, including but not limited to scripts, interpretable programs, dynamic link libraries (DLLs), Java classes, and complete executable programs. Moreover, parts of the processing of the invention may be distributed for better performance, reliability, and/or cost.
  • Non-volatile media includes, for example, optical, magnetic disks, and magneto-optical disks, such as the hard disk 1207 or the removable media drive 1208.
  • Volatile media includes dynamic memory, such as the main memory 1204.
  • Transmission media includes coaxial cables, copper wire and fiber optics, including the wires that make up the bus 1202. Transmission media also may also take the form of acoustic or light waves, such as those generated during radio wave and infrared data communications.
  • Various forms of computer readable media may be involved in carrying out one or more sequences of one or more instructions to processor 1203 for execution.
  • the instructions may initially be carried on a magnetic disk of a remote computer.
  • the remote computer can load the instructions for implementing all or a portion of the invention remotely into a dynamic memory and send the instructions for example over a telephone line using a modem.
  • a modem local to the computer system 1201 may receive the data on the telephone line and use an infrared transmitter to convert the data to an infrared signal.
  • An infrared detector coupled to the bus 1202 can receive the data carried in the infrared signal and place the data on the bus 1202.
  • the bus 1202 carries the data to the main memory 1204, from which the processor
  • 1204 may optionally be stored on storage device 1207 or 1208 either before or after execution by processor 1203.
  • the computer system 1201 also includes a communication interface 1213 coupled to the bus 1202.
  • the communication interface 1213 provides a two-way data communication coupling to a network link 1214 that is connected to, for example, a local area network (LAN) 1215, or to another communications network 1216 such as the Internet.
  • the communication interface 1213 may be a network interface card to attach to any packet switched LAN.
  • the communication interface 1213 may be an asymmetrical digital subscriber line (ADSL) card, an integrated services digital network (ISDN) card or a modem to provide a data communication connection to a corresponding type of communications line.
  • Wireless links may also be implemented.
  • the communication interface 1213 sends and receives electrical, electromagnetic or optical signals that carry digital data streams representing various types of information.
  • the network link 1214 typically provides data communication through one or more networks to other data devices.
  • the network link 1214 may provide a connection to another computer through a local network 1215 (e.g., a LAN) or through equipment operated by a service provider, which provides communication services through a communications network 1216.
  • the local network 1214 and the communications network 1216 use, for example, electrical, electromagnetic, or optical signals that carry digital data streams, and the associated physical layer (e.g., CAT 5 cable, coaxial cable, optical fiber, etc).
  • the signals through the various networks and the signals on the network link 1214 and through the communication interface 1213, which carry the digital data to and from the computer system 1201 may be implemented in baseband signals, or carrier wave based signals.
  • the baseband signals convey the digital data as unmodulated electrical pulses that are descriptive of a stream of digital data bits, where the term "bits" is to be construed broadly to mean symbol, where each symbol conveys at least one or more information bits.
  • the digital data may also be used to modulate a carrier wave, such as with amplitude, phase and/or frequency shift keyed signals that are propagated over a conductive media, or transmitted as electromagnetic waves through a propagation medium.
  • the digital data may be sent as unmodulated baseband data through a "wired" communication channel and/or sent within a predetermined frequency band, different than baseband, by modulating a carrier wave.
  • the computer system 1201 can transmit and receive data, including program code, through the network(s) 1215 and 1216, the network link 1214, and the communication interface 1213.
  • the network link 1214 may provide a connection through a LAN 1215 to a mobile device 1217 such as a personal digital assistant (PDA) laptop computer, or cellular telephone.
  • PDA personal digital assistant
  • kits to facilitate application of the invention.
  • a kit would comprise at least one activatable agent capable of producing a predetermined cellular change, optionally at least one energy modulation agent capable of activating the at least one activatable agent when energized, optionally at least one plasmonics agent that can enhance the CR light such that the CR light activates the at least one activatable agent which produces a change in the medium when activated, and containers suitable for storing the various agents in stable form, and further comprising instructions for administering the at least one activatable agent and/or at least one energy modulation agent to a medium, and for applying an initiation energy from an initiation energy source to activate the activatable agent.
  • the instructions could be in any desired form, including but not limited to, printed on a kit insert, printed on one or more containers, as well as electronically stored instructions provided on an electronic storage medium, such as a computer readable storage medium. Also optionally included is a software package on a computer readable storage medium that permits the user to integrate the
  • a system for imaging or treating a tumor in a human or animal body includes a pharmaceutical carrier, a photoactivatable drug , one or more devices which infuse the tumor with the photoactivatable drug and the pharmaceutical carrier, an x-ray or high energy electron or proton source, and a processor programmed to control a dose of x-rays or electrons to the tumor for production of light inside the tumor by CR to activate the photoactivatable drug.
  • the first system includes a mechanism configured to supply in the biological medium at least one of a plasmonics agent and a photoactivatable drug and an energy modulation agent.
  • the plasmonics agent enhances or modifies energy in a vicinity of itself.
  • the plasmonics agent enhances or modifies the CR such that the enhanced CR produces directly or indirectly the change in the medium.
  • the system includes an initiation energy source configured to apply an initiation energy to the biological medium to activate the at least one activatable agent in the biological medium.
  • the applied initiation energy or the CR interacts with the energy modulation agent to directly or indirectly produce the change in the medium by emitted light (UV and/or visible light) from the CR light or from the energy modulation agent.
  • emitted light UV and/or visible light
  • the energy modulation agent converts the applied initiation energy or the CR light and produces light (UV and/or visible light) at an energy to activate the drug or photoactivatable substance.
  • the plasmonics agent (if present) can enhance the light from the at least one energy modulation agent or the CR light.
  • the applied initiation energy source is an external initiation energy source. .
  • the systems described herein can further permit the at least one activatable agent to include a photoinitiator such as one of benzoin, substituted benzoins, alkyl ester substituted benzoins, Michler's ketone, dialkoxyacetophenones, diethoxyacetophenone, benzophenone, substituted benzophenones, acetophenone, substituted acetophenones, xanthone, substituted xanthones, benzoin methyl ether, benzoin ethyl ether, benzoin isopropyl ether,
  • a photoinitiator such as one of benzoin, substituted benzoins, alkyl ester substituted benzoins, Michler's ketone, dialkoxyacetophenones, diethoxyacetophenone, benzophenone, substituted benzophenones, acetophenone, substituted acetophenones, xanthone, substituted xanthones,
  • diethanolaminebenzophenone camphoquinone
  • peroxyester initiators non-fluorene-carboxylic acid peroxyesters and mixtures thereof.
  • the systems described herein can also include a mechanism configured to provide in the medium plasmonics-agents including metal nanostructures such as for example nanospheres, nanorods, nanocubes, nanopyramids, nanoshells, multi-layer nanoshells, and combinations thereof.
  • metal nanostructures such as for example nanospheres, nanorods, nanocubes, nanopyramids, nanoshells, multi-layer nanoshells, and combinations thereof.
  • the article can include luminescent particles such as for example nanotubes, nanoparticles, chemiluminescent particles, and bioluminescent particles, and mixtures thereof.
  • the article can include nanoparticles of semiconducting or metallic materials.
  • the article can include chemiluminescent particles.
  • the article can include plasmonics-agents including metal nanostructures such as for example nanospheres, nanorods, nanocubes, nanopyramids, nanoshells, multi-layer nanoshells, and combinations thereof.
  • a subject is administered an activatable pharmaceutical agent, optionally along with at least one energy modulation agent capable of converting x-rays into a wavelength that will activate the activatable pharmaceutical agent.
  • the subject is then placed into a source of x-rays or high energy particles which generate inside the subject CR. From the CR light, at least one photoactive drug is activated inside the subject to thereby treat the subject for a cell proliferation disorder.
  • another aspect of the invention includes a method for treating a subject carrying a virus in which the method provides within the subject at least one photoactive drug for treatment of the subject carrying the virus and applies initiation energy from at least one source to a target inside the subject.
  • the at least one photoactive drug is activated directly or indirectly at the target inside the subject by CR light or light from energy modulation agents to thereby treat the subject carrying the virus.
  • Mechanisms included in the invention can involve photoactivation of a drug such as a psoralen or its derivatives or an alkylating agent.
  • Mechanisms included in the invention can involve the formation of highly reactive oxygen species, such as singlet oxygen. Any of these mechanisms can be used in combination or selectively to treat a subject with a cell proliferation disorder, or who is carrying viruses and/or has associated disorders or symptoms thereof.
  • the CR light can be used to activate an alkylating agent (e.g., iodonophthylazide) for its attachment to a virus.
  • the CR light can be used to activate a psoralen (or a derivative or substitute thereof) for treatment of a bacterial infection or other disorders in the patient.
  • one wavelength of the CR light can be used to activate an alkylating agent (e.g., iodonophthylazide) for its attachment to a virus, while another different wavelength of the CR light can be used to activate a psoralen (or a derivative or substitute thereof) for treatment of a bacterial infection or other disorders in the patient.
  • an alkylating agent e.g., iodonophthylazide
  • another different wavelength of the CR light can be used to activate a psoralen (or a derivative or substitute thereof) for treatment of a bacterial infection or other disorders in the patient.
  • one wavelength can be used to activate an alkylating agent or a psoralen, while another wavelength is used for a different purpose such as for example production of singlet oxygen (i.e., highly reactive oxygen species) or for production of sterilizing UV light or to promote cell growth or reduce inflammation, etc.
  • singlet oxygen i.e., highly reactive oxygen species
  • one or more wavelengths of the CR light could be used for treatment a host or arrest of viruses such as Ebola, West Nile, encephalitis, HIV, etc., and/or for the regulation and control of biological responses having varying degrees of apoptosis (the process of programmed cell death PCD) and necrosis (the premature death of cells and living tissue typically from external factors).
  • apoptosis the process of programmed cell death PCD
  • necrosis the premature death of cells and living tissue typically from external factors.
  • factors external to the cell or tissue such as infection, toxins, or trauma that result in the unregulated digestion of cell components.
  • necrosis is a naturally occurring programmed and targeted cause of cellular death. While apoptosis often provides beneficial effects to the organism, necrosis is almost always detrimental and may be fatal.
  • the alkylating agent can be at least one or more of drugs from the iodonophthylazide family, such as 1,5-iodonaphthylazide (INA).
  • INA 1,5-iodonaphthylazide
  • These photoactivatable compounds are non-toxic, hydrophobic compounds that can penetrate into the innermost regions of biological membrane bilayers and selectively accumulate in such inner membrane regions. Upon irradiation with CR light or light from energy modulation agents, generated inside or nearby the membrane region, it is believed that a reactive derivative of the compound is generated that binds to membrane proteins deep in the lipid bilayer.
  • the inactivated agent constitutes a vaccine created inside the subject animal or bird or human with the vaccine specific to the viral or bacterial infection of the animal or bird or human.
  • a photoactive drug such as a psoralen or its derivatives is used separately or in conjunction with at least one alkylating agent.
  • the psoralen is photactivated inside the cell by ultraviolet or visible light generated within the cell or nearby the cell by the CR light or by light from energy modulation agents.
  • the activated psoralen attaches to the virus's genetic contents, prevents its replication, and causes local cell death (one form of treatment).
  • the psoralen- inactivated virus can induce an autoimmune response from the animal or bird or human resulting in the body effectively eliminating untreated viruses in other regions of the body.
  • 1,5-iodonaphthyl azide INA
  • INA is employed as a photoactivatable hydrophobic compound.
  • INA is a nontoxic hydrophobic compound.
  • the structure for 1,5-iodonaphthyl azide (INA) is provided below.
  • the photoactivatable hydrophobic compounds Upon exposure to cells, the photoactivatable hydrophobic compounds can penetrate into the innermost regions of biological membrane bilayers and will accumulate selectively in these regions.
  • ultraviolet light e.g., 320 to 400 nm
  • a reactive derivative is generated that binds to membrane proteins deep in the lipid bilayer.
  • the photoactivatable hydrophobic compounds of the invention can be used for inactivation of viruses, bacteria, parasites and tumor cells using visible light.
  • a photosensitizer when visible light is used a photosensitizer, a chromophore is typically needed unless the photoactive drug is developed to be activated directly by visible light.
  • a photosensitizer chromophore has an absorption maximum in the visible light range and can photosensitize the photoactivatable hydrophobic compounds of the invention.
  • the photosensitizer chromophores have absorption maxima in the range of about 450 to about 525 nm or about 600 to about 700 nm.
  • Suitable photosensitizer chromophores can include one or more of a porphyrin, chlorin, bacteriochlorin, purpurin, phthalocyanine, naphthalocyanine, merocyanines, carbocyanine, texaphyrin, non-tetrapyrrole, or other photosensitizers known in the art.
  • Specific examples of photosensitizer chromophores include fluorescein, eosin, bodipy, nitro-benzo-diazol ( BD), erythrosine, acridine orange, doxorubicin, rhodamine 123, picoerythrin and the like.
  • viruses, bacteria, parasites and tumor cells and other infectious structures and microorganisms can be inactivated by exposure to photoactivatable hydrophobic compounds which were themselves activated by light generated internally within the animal or bird or human subject by CR light or light from energy modulation agents or light from photosensitizer chromophores.
  • the photoactivatable hydrophobic compound is 1,5-iodonaphthyl azide (INA) or a related compound.
  • INA 1,5-iodonaphthyl azide
  • the virus, parasite or tumor cell is contacted with the recently photoactivated hydrophobic compound, which was photoactivated by ultraviolet light generated internally using the energy modulation agents of the invention.
  • the virus, parasite, tumor cell or other infectious structures and microorganisms are contacted with both the photoactivatable hydrophobic compound and a photosensitizer chromophore that absorbs visible light, then visible light generated internally by CR light or light from energy modulation agents or light from photosensitizer chromophores can photoactivate the photoactivatable hydrophobic compound. Accordingly, in one embodiment, exposure to internally generated ultraviolet light directly photoactivate s the photoactivatable hydrophobic compound within viral and cellular membranes. In one embodiment, exposure to internally generated visible light first
  • a reactive derivative of the photoactivatable hydrophobic compound is generated that binds to membrane proteins deep within the lipid bilayer. This process is believed to cause specific inactivation of integral membrane proteins embedded in the membrane, while maintaining the integrity and activity of proteins that protrude outside of the membrane.
  • the invention with internally generated light can provide a method that can inactivate a wide variety of viruses, bacteria, parasites and tumor cells in a way that the inactivated species can be safely used as immunological compositions or vaccines to inhibit the disease they cause.
  • the activated drug agents (generated indirectly from the CR light activating a photoactivatable drug) kill the organism or cell in a specific manner that maintains its structure and conformation.
  • the structure of the inactivated virus/cell is similar to that of the live virus/cell.
  • the immunogenicity of the organism or cell as a whole is maintained and can be safely used to stimulate the immune system of a subject animal or bird or patient.
  • the inactivated viruses, bacteria, cancer cells, or parasites generated inside the animal or bird or human subject can be used for vaccination without causing disease or other negative side effects.
  • the IN A internal treatment procedures generate inactive viruses inside the subject that can be used in a manner similar to aldrithiol inactivated HIV (developed by the AIDS vaccine program SAIC).
  • the INA-internal-inactivation procedures of this invention can be used in conjunction with aldrithiol inactivation procedures to generate inactive HIV that comply with the requirements of the FDA.
  • two mechanistically independent methods of inactivation can be used to provide a prophylactic AIDS or HIV vaccine.
  • prevention or treatment of microbial infections, viral infections, parasitic infections, prion infection or cancer is intended to include the alleviation of or diminishment of at least one symptom typically associated with the infection or cancer.
  • Prevention or treatment also includes alleviation or diminishment of more than one symptom.
  • treatment with the internally inactivated agents of the invention generates immunity in the animal or bird or human towards the agent while prevention by the inactivated agents of the invention substantially eliminates the symptoms associated with the infection or cancer.
  • infections that can be treated by the present internally activated drug agents generated indirectly from the CR light activating a
  • photoactivatable drug include infections by any target infectious organisms and structures that can infect a mammal or other animal or a bird.
  • target infectious organisms and structures include, but are not limited to, any virus, bacterium, fungus, single cell organism, prion conformations or parasite that can infect an animal, including mammals.
  • target microbial organisms include viruses, bacteria, fungi, yeast strains and other single cell organisms.
  • the inactivated agents of the invention can give rise to immunity against both gram-negative and gram-positive bacteria.
  • Exemplary viral infections that can be treated by this invention include infections by any virus that can infect animals (including but not limited to mammals or birds), including enveloped and non-enveloped viruses, DNA and RNA viruses, viroids, and prions.
  • infections or unwanted levels of the following viruses and viral types can be treated internally: human immunodeficiency virus (HIV), simian immunodeficiency virus (SIV), hemorrhagic fever viruses, hepatitis A virus, hepatitis B virus, hepatitis C virus, poxviruses, herpes viruses, adenoviruses, papovaviruses, parvoviruses, reoviruses, orbiviruses, picornaviruses, rotaviruses, alphaviruses, rubiviruses, influenza virus type A and B, flaviviruses, coronaviruses, paramyxoviruses, morbilliviruses, pneumoviruses, rhab
  • HBVs hemorrhagic fever viruses
  • Chikungunya virus Japanese encephalitis virus
  • Monkey pox virus variola virus
  • Congo-Crimean hemorrhagic fever virus Junin virus
  • Omsk hemorrhagic fever virus Venezuelan equine encephalitis virus
  • Dengue fever virus Lassa fever virus
  • Rift valley fever virus Western equine encephalitis virus
  • Eastern equine encephalitis virus Lymphocytic choriomeningitis virus
  • Russian Spring-Summer encephalitis virus White pox, Ebola virus, Machupo virus, Smallpox virus, Yellow fever virus, Hantaan virus, Marburg virus, and Tick-borne encephalitis virus.
  • Aeromonas spp. including, for example, Aeromonas hydrophila, Aeromonas caviae and Aeromonas sobria
  • Bacillus spp. including, for example, Bacillus cereus, Bacillus anthracis and Bacillus thuringiensis
  • Bacteroides spp. including, for example, B. fragilis, B. thetaiotaomicron, B. vulgatus, B. ovatus, B. distasonis, B. uniformis, B. stercoris, B. eggerthii, B. merdae, and B.
  • Campylobacter spp. including, for example, Campylobacter jejuni, Campylobacter laridis, and Campylobacter hyointestinalis
  • Clostridium spp. such as the pathogenic Clostridia including all types of Clostridium botulinum (including those in Groups I, II, III and IV, and including those that produce botulism A, B, C, D, E, F and G), all types of Clostridium tetani, all types of Clostridium difficile, and all types of Clostridium perfringens
  • Ebola spp. e.g. EBOV Zaire
  • Enterobacter spp. including, for example,
  • Enterobacter aerogenes also sometimes referred to as Klebsiella mobilis
  • Enterobacter agglomerans also sometimes referred to as Pantoea agglomerans
  • Enterobacter amnigenus Enterobacter asburiae
  • Enterobacter cancerogenus also sometimes referred to as Enterobacter taylorae and/or Erwinia cancerogena
  • Enterobacter cloacae Enterobacter cowanii
  • Enterobacter dissolvens also sometimes referred to as Erwinia dissolvens
  • Enterobacter hormaechei Enterobacter intermedium
  • Enterobacter intermedius also sometimes referred to as Enterobacter intermedium
  • Enterobacter kobei Enterobacter nimipressuralis (also sometimes referred to as Erwinia nimipressuralis)
  • Enterobacter sakazakii Enterobacter taylorae (also sometimes referred to as Enterobacter cancerogenus)
  • Enterococcus spp Enterococcus spp.
  • VRE Vancomycin Resistant Enterococcus
  • ETEC enterotoxigenic
  • enteropathogenic (EPEC) strains the enterohemorrhagic (EHEC) strain designated E. coli 0157:H7, and the enteroinvasive (EIEC) strains
  • Gastrospirillum spp. including, for example, Gastrospirillum hominis (also sometimes now referred to as Helicobacter heilmannii)
  • Helicobacter spp. including, for example, Helicobacter pylori and Helicobacter hepaticus
  • Klebsiella spp. including, for example, Klebsiella pneumoniae, Klebsiella ozaenae, Klebsiella rhinoscleromatis, Klebsiella oxytoca, Klebsiella planticola, Klebsiella terrigena, and Klebsiella ornithinolytica
  • Salmonella spp. including, for example, S. typhi and S. paratyphi A, B, and C, S. enteritidis, and S. dublin
  • Staphylococcus spp. including, for example, Staphylococcus aureus, methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus saprophyticus and Staphylococcus epidermis
  • Streptococcus ssp including, for example, Shigella sonnei, Shigella boydii, Shigella flexneri, and Shigella dysenteriae
  • Staphylococcus spp. including, for example, Staphylococcus aureus, methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus saprophyticus and Staphylococcus epidermis
  • Streptococcus ssp including, for example, Shigella sonnei, Shigella boydii, Shigella flexneri, and Shigella dysenteriae
  • Staphylococcus spp. including, for example, Staphylococcus aureus
  • Streptococcus pyogenes including Groups A (one species with 40 antigenic types, Streptococcus pyogenes), B, C, D (five species (Streptococcus faecalis, Streptococcus faecium, Streptococcus durans, Streptococcus avium, and Streptococcus bovis)), F, and G, including Streptococcus pneumoniae), Pseudomonas spp.
  • Vibrio cholera Serogroup 01 and Vibrio cholera Serogroup Non-Ol Vibrio parahaemolyticus, Vibrio alginolyticus, Vibrio furnissii, Vibrio carchariae, Vibrio hollisae, Vibrio multiplinnatiensis, Vibrio metschnikovii, Vibrio damsela, Vibrio mimicus, Vibrio vulnificus, and Vibrio fluvialis
  • Yersinia pestis including, for example, Yersinia pestis, Yersinia enterocolitica and Yersinia pseudotuberculosis
  • Neisseria including, for example, Yersinia pestis, Yersinia enterocolitica and Yersinia pseudotuberculosis
  • Neisseria Proteus, Citrobacter, Aerobacter
  • Providencia Serratia
  • Brucella Francisella tularensis
  • Bacillus tularensis Bacillus tularensis
  • Brucella tularensis also sometimes referred to as Pasteurella tularensis
  • Bacillus tularensis Bacillus tularensis
  • Brucella tularensis tularemia
  • rabbit fever deerfly fever
  • Ohara's disease Ohara's disease
  • Francis disease and the like.
  • various bacterial infections or unwanted levels of bacteria that can be treated, prevented or addressed by the present invention include but are not limited to those associated with anthrax (Bacillus anthracis), staph infections (Staphylococcus aureus), typhus (Salmonella typhi), food poisoning (Escherichia coli, such as 0157:H7), bascillary dysentery (Shigella dysenteria), pneumonia (Psuedomonas aerugenosa and/or Pseudomonas cepacia), cholera (Vibrio cholerae), ulcers (Helicobacter pylori), Bacillus cereus, Salmonella, Clostridium perfringens, Campylobacter, Listeria monocytogenes, Vibrio parahaemolyticus, botulism
  • anthrax Bacillus anthrax
  • staph infections Staphylococcus aureus
  • typhus Salmonella ty
  • E. coli serotype 0157:H7 has been implicated in the pathogenesis of diarrhea, hemorrhagic colitis, hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP).
  • the internally inactivated agents of this invention are also active against drug-resistant and multiply-drug resistant strains of bacteria, for example, multiply-resistant strains of Staphylococcus aureus and vancomycin-resistant strains of Enterococcus faecium and Enterococcus faecalis.
  • Fungal infections that can be treated or prevented by this invention include infections by fungi that infect a mammal or a bird, including Histoplasma capsulatum, Coccidioides immitis, Cryptococcus neoformans, Candida ssp. including Candida albicans, Aspergilli ssp. including Aspergillus fumigatus, Sporothrix, Trichophyton ssp., Fusarium ssp., Tricosporon ssp.,
  • Pneumocystis carinii and Trichophyton mentagrophytes.
  • infections or unwanted levels of target fungi can be treated, prevented or addressed by the present inactivated agents.
  • Such fungi also include fungal pathogens that may have potential for use biological weapons, including Coccidioides immitis and Histoplasma capsulatum.
  • Prions that are treatable in the invention are proteins that can access multiple
  • infectious proteins show several remarkable biological activities, including the ability to form multiple infectious prion conformations, also known as strains or variants, encoding unique biological phenotypes, and to establish and overcome prion species (transmission) barriers. See, e.g., Tessier et al., Unraveling infectious structures, strain variants and species barriers for the yeast prion [PSI+], Nat. Struct. Mol. Biol. 2009 Jun; 16(6): 598-605.
  • Solid mammalian tumors include cancers of the head and neck, lung, mesothelioma, mediastinum, esophagus, stomach, pancreas, hepatobiliary system, small intestine, colon, colorectal, rectum, anus, kidney, urethra, bladder, prostate, urethra, penis, testis, gynecological organs, ovaries, breast, endocrine system, skin central nervous system; sarcomas of the soft tissue and bone; and melanoma of cutaneous and intraocular origin.
  • Hematological malignancies include childhood leukemia and lymphomas, Hodgkin's disease, lymphomas of lymphocytic and cutaneous origin, acute and chronic leukemia, plasma cell neoplasm and cancers associated with AIDS.
  • a cancer at any stage of progression can be treated, such as primary, metastatic, and recurrent cancers. Both human and veterinary uses are contemplated.
  • a method for treating a subject with a virus or a bacterium which 1) provides within lymph nodes of the subject at least one photoactive or photoactivatable drug for treatment of the virus or the bacterium, and 2) applies initiation energy from at least one source to the lymph nodes.
  • the method 3) activates by the CR light directly or indirectly the at least one photoactive or photoactivatable drug at the target inside the lymph nodes.
  • the method 4) reacts the activated drug with the virus or bacterium to inactivate the virus or the bacterium to thereby treat the subject.
  • a clinical megavoltage (MV) radiation beam delivers the normal radiation dose to the tumor, while concomitantly emitted Cherenkov light (CL), a byproduct of the radiation beam, simultaneously photo-activates administered psoralen specifically within the treatment zone.
  • CL is a broad-spectrum UV- visible light produced when charged particles exceed the phase velocity of light within a dielectric material.
  • CL is produced throughout irradiated tissue, with intensity proportional to the local absorbed dose produced from secondary electrons generated throughout the beam path (Glaser, A.K., et al., Phys Med Biol, 2014. 59(14): p. 3789- 811).
  • CL intensity per unit radiation dose increases with photon energy (Glaser, A.K., et al., Phys Med Biol, 2015. 60(17): p. 6701-18), suggesting the potential for optimization by using higher energy photon beams and filtering out low-energy photons. This is investigated here through experimental measurements.
  • Figure 6A is a schematic of the experimental setup used to ascertain the relative
  • Figure 6B is a plot of the measured Cherenkov radiation output normalized to account for differences in the total x-ray dose having been transmitted into the quinine sulfate solution for the different filters: with no filter, with a 1 cm thick carbon filter, with a 2 cm thick carbon filter, and with a 10 cm thick polyurethane filter.
  • the results in Figure 6B show that these test filters were effective in providing an x-ray spectrum of x-ray fluxes that preferentially generate more Cherenkov radiation.
  • Figure 6C is a comparison of the UV-Vis Cherenkov light spectrum showing that the light produced has the same or nearly the same spectrum when no filter was used and when the 10 cm thick polyurethane filter was used.
  • both x- ray flux from the above-noted linear accelerator and optionally UV-Vis Cherenkov light from the above-noted phantom were allowed to simultaneously expose proximate wells of B16 melanoma assay, with one cell receiving both x-rays and UV-Vis Cherenkov light and a proximate cell receiving only x-rays.
  • Different concentrations of 4,5',8-trimethyl psoralen (TMP) are applied to different wells containing the B 16 melanoma cells.
  • FIG. 7 A is a plot of cell kill as a function of TMP concentration with and without Cherenkov (i.e. with and without the UV-Vis Cherenkov light) after exposure to 6 MV x-rays at a 2 Gy dose.
  • the cell kill was measured after a 48-hour incubation time. Specifically, a CellTiter-GloTM luminescence (cell viability) measurement versus concentration of TMP was made with or without Cherenkov light.
  • the CellTiter-GloTM luminescence technique involves the introduction of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) into the wells containing the B 16 melanoma cells.
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • Viable cells with active metabolism convert MTT into a purple colored formazan product with an absorbance maximum near 570 nm.
  • the amount of the purple colored formazan product is proportional to the number of living cells. Accordingly, the quantity of formazan (presumed directly proportional to the number of viable cells) is measured by recording changes in absorbance at 570 nm using a plate reading
  • Figure 7B is a plot of the flow cytometry data acquired from B16 melanoma cells indicating a similar effect to the cytotoxicity depicted in Figure 7A.
  • MHC class I molecules present peptides to cytotoxic cells. Only peptides with the right length and sequence bind to the nascent MHC class I molecules in the assay. Accordingly, when cytotoxic products from cell kill of the B 16 melanoma cells are present, these products will be bound to surface containing the MHC class I molecules, which was then evaluated by flow cytometry.
  • the data of Figure 7B shows the results for a 15 MV X-ray exposure after a 72 hour incubation.
  • the data of Figure 7C shows the results for the 15 MV X-ray exposure after the 72 hour incubation with the data presented in terms of cell kill.
  • the relatively high MHC 1 expression (13.7 % vs. 8.01% and 5.46% for the controls with no Cherenkov and with no x-rays) is consistent with an immunogenic response.
  • the luminescence assay measures total cell metabolic activity, which serves as a surrogate measure of cell proliferation and viability (Crouch, S.P., et al., J Immunol Methods, 1993. 160(1): p. 81-8). Flow cell cytometry was used to determine change in Major
  • MHC Histocompatibility Complex
  • 4T1 breast adenocarcinoma and B 16 melanoma cells were thawed from -80oC and plated onto Corning® 100mm culture dish at least 2 days before irradiation.
  • Cells were grown in a 5% C02 maintained incubator in RPMI-1640 with 10% FBS and L-glutamine from GIBCO (Grand Island, NY) at 37oC.
  • RPMI-1640 10% FBS and L-glutamine from GIBCO (Grand Island, NY) at 37oC.
  • GIBCO Gibnd Island, NY
  • TMP trioxsalen
  • RPMI 10% FBS growth medium
  • Flow cytometric analyses were performed on BD LSRFortessaTM Cell Analyzer system and analyzed using FlowJo (Tree Star Inc., version 10.0.7). Cells were first gated on forward and side scatter (FSC/SSC) to exclude small fragments from analysis. Fluorescence of MHC I labelled with Allophycocyanin (APC) was then measured. All samples were analyzed on the same day with equal FSC, SSC, APC detector gain voltages and gating. In preparation for flow-cytometry, cells were trypsinized and centrifuged 48 hours after irradiation, and then re- suspended in Cell Staining Buffer at 100,000 cells per mL as per BioLegend® staining protocol.
  • FSC/SSC forward and side scatter
  • APC Allophycocyanin
  • Isotype cells were prepared from un-irradiated (OGy) controls for auto- fluorescence and non-specific binding control for the antibody.
  • MHC I expression histograms measured as APC fluorescence intensity, were compiled for each treatment condition.
  • the effect of CLAP on overall MHC I expression was investigated through pairs of wells treated with the following conditions: 3Gy with/without psoralen; 6Gy with/without psoralen; and OGy controls.
  • One of each pair of wells received CL and the other did not by virtue of the light block.
  • a CLAP effect would manifest as a difference between cells exposed to CL versus unexposed only when psoralen is present.
  • Two wells were allocated per condition, but the two wells were combined into one sample before analysis. Total number of analyzed events were about 200,000-500,000 per well.
  • ColCountTM (Oxford Optronix, version 5) was used to count the number of surviving colonies. Student's t-test assuming equal variance was performed to compare colony counts with or without CL. Plating efficiency was about 15% at OGy, resulting in -450 colonies per 3,000 cells plated after 1-2 weeks.
  • An ion chamber was placed at a depth of 9cm to measure ionization current (nA), which is proportional to dose rate.
  • An optical fiber was bundled with the ion chamber, directed vertically down and out of the MV beam path. CL read-out was made via optical fiber coupled to LineSpecTM CCD Array
  • the spectrometer and ion chamber read-outs were simultaneously performed while the MV beam was delivered.
  • Spectrometer integration time was set at 800ms per frame with 10 averages, for 8s total acquisition time.
  • Lead radiation shielding protected the CCD from scattered MV beam, and reduced CCD noise.
  • the measured spectrum from the water phantom were normalized by ion chamber reading, then integrated from 350 to 500nm (around quinine sulfate emission peak) to obtain relative CL output per dose.
  • Figures 10A&10B shows the luminescence assay for cell viability for both B16 and 4T1 cells with (blue line) and without (red line) CLAP. All cells were irradiated with 2Gy radiation at 6MV energy, but with varying psoralen concentration as indicated. Lines represent least square fits to data points, with 95% confidence intervals indicated by the shaded regions. Cell-Titer Glo® ATP luminescence assay results are provided at varying concentrations of psoralen (TMP) for (Fig. 10A) 4T1 and (Fig. 10B) B16 cells. All cells were exposed to 2Gy, with half the cells also exposed to CL as illustrated in Figure 8A. A maximum of 20% and 9.5% decrease in viability is noted in presence of Cherenkov for 4T1 and B 16, respectively.
  • TMP psoralen
  • FIGS 11 A&l IB show the MHC I expression results. Flow cytometry for B16 melanoma, demonstrated CLAP causes a substantial increase in MHC I expression over and above that caused by radiation alone.
  • FIG. 1 1 A Histograms of MHC I expression.
  • FIG. 1 IB Median MHC I expression increases for cells receiving CL (Purple) compared to no CL (Green) only in the presence of psoralen. Wilcoxon rank-sum comparisons are shown for each CL/no-CL (green-purple) pair. Statistically significant comparisons (p ⁇ 0.0001) are marked with a star (*). All cells, including the controls, were exposed to ⁇ psoralen, representing the baseline control for comparison.
  • Figure 11 A upper panel the MHC I expression profiles are compared directly between the un-irradiated control (OGy) and cells irradiated with the same 3 Gy treatment field, but with half the cells exposed to CL by virtue of the light block ( Figures 8A&8B).
  • Figure 11 A lower panel shows the same plots but this time for the higher irradiation dose of 6Gy.
  • Figure 1 IB compares the median MHC I of all five conditions after background correction by subtraction of the isotype background MHC I signal. Statistically significant differences between the CL/no-CL pairs are indicated with a star (*), and confirm CLAP enhancement of MHC I expression only occurs when psoralen is present.
  • Figure 13 A shows the relative psoralen absorbance spectrum of 8-MOP at lC ⁇ g/mL compared to Cherenkov emission for 15MV clinical photon beam in water (obtained using
  • Figure 13B shows the potential for optimizing the amount of CL per unit-dose by changing energy and incorporating filters.
  • CL output per MV radiation dose was physically measured from the set-up illustrated in Figure 9. Effects of beam energy and polyurethane (low- Z) filter were demonstrated.
  • Relative Cherenkov output is estimated from cumulative counts from measured spectrum in the range 350-500nm. Adding a specialized low-Z filter to flattening filter free 10MV beam such as 10cm polyurethane increased Cherenkov output per dose than the standard beam (from 97000 to 109000, 13% increase).
  • Figures 10A&10B show increased cytotoxicity of CLAP in both 4T1 and B 16 cell lines as measured by ATP luminescence assay. All other conditions being identical, cells exposed to full CLAP treatment (with Cherenkov) showed lower cell viability compared to cells that were not exposed to CL (radiation only). Interestingly, as exposure to psoralen increases (TMP at 0- ⁇ ) a maximum differential at around 50 ⁇ is observed, after which the differential decreases. The maximum magnitude of difference is 20% and 9.5% for 4T1 and B 16
  • ECP photopheresis
  • Figure 13B demonstrates the possibility to optimize the clinical treatment beam for CLAP by increasing the Cherenkov output per unit dose.
  • Introducing a low-Z filter here a 10cm block of polyurethane
  • CLAP achieves an effect because of the near identical match between the psoralen activation and CL emission spectra ( Figure 13 A) which creates uniquely efficient photo-activation.
  • the peak wavelengths for cytotoxic DNA-DNA crosslinking and DNA-protein crosslinking (specifically RecA, a DNA-repairing protein) have been reported to be 320nm and 300nm respectively for HMT psoralen (Sastry, S.S., et al., J Biol Chem, 1997. 272(6): p. 3715- 23).
  • the CL spectrum spans that range and is more intense at shortwave wavelengths.
  • longwave UVA light (397.9nm) preferentially induces DNA
  • a method for treating a subject with a disorder comprising:
  • applying comprises applying the initiation energy through a filter preferentially removing lower energy x-rays while transmitting higher energy x- rays.
  • applying comprises applying the initiation energy through a low mass filter.
  • applying the initiation energy through a low mass filter comprises applying the initiation energy through a section of carbon-containing material which is between 1 cm and 20 cm thick.
  • applying the initiation energy through a low mass filter comprises applying the initiation energy through a section of carbon-containing material which is between 5 cm and 15 cm thick.
  • applying the initiation energy through a low mass filter comprises applying the initiation energy through a section of carbon-containing material which is between 7 cm and 12 cm thick.
  • applying the initiation energy through a low mass filter comprises applying the initiation energy through a section of carbon-containing material containing at least one of H, F, Si, N, P, and B.
  • applying the initiation energy through a low mass filter comprises applying the initiation energy through a section of carbon-containing material containing in a minority amount at least one of H, F, Si, N, P, and B.
  • activating inside the subject the at least one photoactivatable drug comprises bonding the photoactivatable drug to a cellular structure.
  • the bonding comprises at least one of 1) bonding the photoactivatable drug to at least one of nuclear DNA, mRNA, rRNA, ribosome,
  • photoactivatable drug to lipid bilayers of at least one virus selected from the group consisting of an ebola virus, an encephalitis virus, a West Nile virus, and an HIV virus.
  • activating inside the subject the at least one photoactivatable drug comprises activating an alkylating agent.
  • activating inside the subject the at least one photoactivatable drug comprises activating 1,5-iodonophthylazide.
  • activating inside the subject the at least one photoactivatable drug comprises activating a drug for treating the cell proliferation disorders.
  • activating inside the subject the at least one photoactivatable drug comprises activating a drug for treating at least one of a virus or a bacterium.
  • applying initiation energy comprises applying a filtered set of x-rays to the subject having an energy of at least 0.5 MeV and less than 10 MeV.
  • applying initiation energy comprises applying a filtered set of x-rays to the subject having an energy of at least 1.0 MeV and less than 10 MeV. 22. The method of any one or more of the statements above, wherein applying initiation energy comprises applying a filtered set of x-rays to the subject having an energy of at least 1.5 MeV and less than 10 MeV.
  • applying initiation energy comprises applying a filtered set of x-rays to the subject having an energy of at least 1.0
  • activating inside the subject the at least one photoactivatable drug comprises activating at least one of
  • activating inside the subject the at least one photoactivatable drug comprises inducing an autoimmune response, exciting a DNA strand of a cancer cell, redirecting a metabolic pathway, up-regulating genes, down-regulating genes, secreting cytokines, altering cytokine receptor responses, releasing metabolites, generating a vaccine, or a combination thereof
  • activating inside the subject the at least one photoactivatable drug comprises altering a cellular response or a metabolic rate of the target structure.
  • said energy modulation agent comprises at least one of a biocompatible fluorescing metal nanoparticle, fluorescing metal oxide
  • nanoparticle fluorescing metal coated metal oxide nanoparticle, fluorescing dye molecule, gold nanoparticle, silver nanoparticle, gold-coated silver nanoparticle, a water soluble quantum dot encapsulated by polyamidoamine dendrimers, a luciferase, a fluorophore, a fluorescent material, a phosphorescent material, a biocompatible phosphorescent molecule, and a lanthanide chelate.
  • said energy modulation agent comprises inorganic materials selected from the group consisting of: metal oxides; metal sulfides; doped metal oxides; and mixed metal chalcogenides.
  • said energy modulation agent comprises at least one of Y 2 0 3 , Y 2 0 2 S, NaYF 4 , NaYbF 4 , YAG, YAP, Nd 2 0 3 , LaF 3 , LaCl 3 , La 2 0 3 , Ti0 2 , LuP0 4 , YV0 4 , YbF 3 , YF 3 , Na-doped YbF 3 , ZnS; ZnSe; MgS; CaS, CaW0 4 , CaSi0 2 :Pb, and alkali lead silicate including compositions of Si0 2 , B 2 0 3 , Na 2 0, K 2 0, PbO, MgO, or Ag, and combinations or alloys
  • said energy modulation agent comprises at least one of ZnSeS:Cu, Ag, Ce, Tb; CaS: Ce,Sm; La 2 0 2 S:Tb; Y 2 0 2 S:Tb; Gd 2 0 2 S:Pr, Ce, F; LaP0 4 .
  • said energy modulation agent comprises at least one of ZnS:Ag, ZnS:Cu, Pb, and alloys of the ZnSeS.
  • said energy modulation agent comprises at least one of sodium yttrium fluoride (NaYF ), lanthanum fluoride (LaF 3 ), lanthanum oxysulfide (La 2 0 2 S), yttrium oxysulfide (Y 2 0 2 S), yttrium fluoride (YF 3 ), yttrium gallate, yttrium aluminum garnet (YAG), gadolinium fluoride (GdF 3 ), barium yttrium fluoride (BaYF 5 , BaY 2 F 8 ), gadolinium oxysulfide (Gd 2 0 2 S), calcium tungstate (CaW0 4 ), yttrium oxide:terbium (Yt 2 0 3 Tb), gadolinium oxysulphide:europium (Gd 2 0 2 S:Eu), lanthanum oxysulphide:europium (La 2 0 2 S:
  • said energy modulation agent comprises at least one of KSrP0 4 :Eu" " , Pr + , NaGdF 4 :Eu, Zn 2 SiG 4 :Tb 3" ⁇ Yb + , p-NaGdF 4 co-doped with Ce 3+ and Tb 3+ ions, and Gd 2 0 2 S:Tm or BaYF 5 :Eu 3+ .
  • the plasmonics-active agent comprises metal nanostructures.
  • the metal nanostructures are nanospheres, nanorods, nanocubes, nanopyramids, nanoshells, multi-layer nanoshells and combinations thereof.
  • the initiation energy comprises at least one or more of x-rays, gamma rays, an electron beam, or a proton beam.
  • autoimmune diseases rheumatoid and inflammatory arthritis
  • behavioral and cognitive disorder/conditi on, joint condition Parkinson's disease, retinal injury and other ocular diseases, enlarged prostate, varicose veins, reduction or removal of fat deposits (liposuction), nerve regeneration, sensory regeneration/restoration, wound healing, chronic pain, conditions occurring in bone tissue, conditions occurring in a soft tissue and/or cartilage, and lymph node condition.
  • the at least one photoactivatable drug comprise at least one pharmaceutical agent selected from the group consisting of a psoralen, pyrene cholesteryloleate, acridine, porphyrin, fluorescein, rhodamine, 16-diazorcortisone, ethidium, transition metal complexes of bleomycin, transition metal complexes of deglycobleomycin organoplatinum complexes, alloxazines, vitamin Ks, vitamin L, vitamin metabolite, vitamin precursor, naphthoquinone, naphthalene, naphthol and derivatives thereof having planar molecular conformations, porphorinporphyrin, dye and phenothiazine derivative, coumarin, quinolone, quinone, and anthroquinone.
  • a pharmaceutical agent selected from the group consisting of a psoralen, pyrene cholesteryloleate, acridine, porphyrin, flu
  • the at least one photoactivatable drug comprises one or more of a psoralen, a coumarin, a porphyrin, and iodonophthylazide, or a derivative thereof.
  • the at least one photoactivatable drug comprises at least one pharmaceutical agent selected from the group consisting of 7,8-dimethyl-10-ribityl, isoalloxazine, 7,8, 10-trimethylisoalloxazine, 7,8- dimethylalloxazine, isoalloxazine-adenine dinucleotide, alloxazine mononucleotide, aluminum (III) phthalocyanine tetrasulonate, hematophorphyrin, and phthadocyanine.
  • the at least one photoactivatable drug comprises an alkylating agent and psoralen.
  • a system for treating a subject with a disorder comprising:
  • a drug administrator which provides within the subject at least one photoactivatable drug for treatment of the subject
  • an initiation energy source which provides inside the subject a preferential x-ray flux for generation of Cherenkov radiation (CR) light capable of activating at least one photoactivatable drug
  • the CR light activates inside the subject the at least one photoactivatable drug to thereby treat the disorder.
  • the system of statement 48 further comprising a filter which preferentially removes lower energy x-rays while transmitting higher energy x-rays.
  • the low mass filter comprises a section of carbon-containing material which is between 1 cm and 20 cm thick.
  • the low mass filter comprises a section of carbon-containing material which is between 5 cm and 15 cm thick.
  • the low mass filter comprises a section of carbon-containing material which is between 7 cm and 12 cm thick.
  • the low mass filter comprises a section of carbon-containing material containing at least one of H, F, Si, N, P, and B.
  • the low mass filter comprises a section of carbon-containing material containing in a minority amount at least one of H, F, Si, N, P, and B.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Radiology & Medical Imaging (AREA)
  • Pathology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne un procédé et un système de traitement d'un sujet atteint d'un trouble, consistant à administrer au moins un médicament photo-activable au sujet en vue de son traitement, à appliquer une énergie d'initiation à partir d'au moins une source pour générer, dans le corps du sujet, un flux de rayons X préférentiel permettant la génération d'une lumière de rayonnement Cherenkov (CR) pouvant activer au moins un médicament photo-activable et, à partir de la lumière CR, activer dans le corps du sujet ledit au moins un médicament photo-activable de manière à traiter le trouble.
PCT/US2018/041753 2017-07-12 2018-07-12 Procédés de radiothérapie pour déclencher l'action de médicaments photo-activables WO2019014413A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762531593P 2017-07-12 2017-07-12
US62/531,593 2017-07-12

Publications (1)

Publication Number Publication Date
WO2019014413A1 true WO2019014413A1 (fr) 2019-01-17

Family

ID=65001815

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/041753 WO2019014413A1 (fr) 2017-07-12 2018-07-12 Procédés de radiothérapie pour déclencher l'action de médicaments photo-activables

Country Status (1)

Country Link
WO (1) WO2019014413A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2724480C2 (ru) * 2019-07-18 2020-06-23 Федеральное государственное бюджетное учреждение "Национальный медицинский исследовательский центр радиологии" Министерства здравоохранения Российской Федерации" (ФГБУ "НМИЦ радиологии" Минздрава России) Способ комбинированной лучевой и фотодинамической терапии
WO2020180426A1 (fr) * 2019-03-04 2020-09-10 Immunolight, Llc. Structures d'augmentation d'énergie, émetteurs d'énergie ou collecteurs d'énergie contenant celles-ci et leur utilisation dans des méthodes et des systèmes pour traiter des troubles de la prolifération cellulaire
CN113042076A (zh) * 2021-03-05 2021-06-29 中国科学院深圳先进技术研究院 一种仿过氧化氢酶活性的光催化纳米酶及其制备方法和应用
CN114099975A (zh) * 2021-11-15 2022-03-01 北京京东方技术开发有限公司 一种胃部滞留胶囊、腰带及胃部神经刺激的方法
CN115414354A (zh) * 2022-08-29 2022-12-02 西南医科大学 花椒毒素在制备治疗血小板减少症药物中的应用

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120282185A1 (en) * 2009-12-09 2012-11-08 Isis Innovation Limited Particles for the treatment of cancer in combination with radiotherapy
WO2016176332A1 (fr) * 2015-04-27 2016-11-03 The Regents Of The University Of Colorado Promédicaments d'anthracyne et procédés de production et d'utilisation de ces derniers
US20170007724A1 (en) * 2014-01-31 2017-01-12 Washington University Imaging and treatment of pathophysiologic conditions by cerenkov radiation
WO2017019520A1 (fr) * 2015-07-24 2017-02-02 Memorial Sloan Kettering Cancer Center Compositions et procédés d'imagerie de cerenkov ciblée et activée et agents thérapeutiques
WO2017027874A1 (fr) * 2015-08-13 2017-02-16 Northeastern University Biomatériaux pour une thérapie d'association radiothérapie-chimiothérapie contre le cancer
WO2017075057A1 (fr) * 2015-10-26 2017-05-04 Immunolight, Llc Procédés de radiothérapie pour déclencher des médicaments à photoactivation

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120282185A1 (en) * 2009-12-09 2012-11-08 Isis Innovation Limited Particles for the treatment of cancer in combination with radiotherapy
US20170007724A1 (en) * 2014-01-31 2017-01-12 Washington University Imaging and treatment of pathophysiologic conditions by cerenkov radiation
WO2016176332A1 (fr) * 2015-04-27 2016-11-03 The Regents Of The University Of Colorado Promédicaments d'anthracyne et procédés de production et d'utilisation de ces derniers
WO2017019520A1 (fr) * 2015-07-24 2017-02-02 Memorial Sloan Kettering Cancer Center Compositions et procédés d'imagerie de cerenkov ciblée et activée et agents thérapeutiques
WO2017027874A1 (fr) * 2015-08-13 2017-02-16 Northeastern University Biomatériaux pour une thérapie d'association radiothérapie-chimiothérapie contre le cancer
WO2017075057A1 (fr) * 2015-10-26 2017-05-04 Immunolight, Llc Procédés de radiothérapie pour déclencher des médicaments à photoactivation

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020180426A1 (fr) * 2019-03-04 2020-09-10 Immunolight, Llc. Structures d'augmentation d'énergie, émetteurs d'énergie ou collecteurs d'énergie contenant celles-ci et leur utilisation dans des méthodes et des systèmes pour traiter des troubles de la prolifération cellulaire
RU2724480C2 (ru) * 2019-07-18 2020-06-23 Федеральное государственное бюджетное учреждение "Национальный медицинский исследовательский центр радиологии" Министерства здравоохранения Российской Федерации" (ФГБУ "НМИЦ радиологии" Минздрава России) Способ комбинированной лучевой и фотодинамической терапии
CN113042076A (zh) * 2021-03-05 2021-06-29 中国科学院深圳先进技术研究院 一种仿过氧化氢酶活性的光催化纳米酶及其制备方法和应用
CN113042076B (zh) * 2021-03-05 2022-03-29 中国科学院深圳先进技术研究院 一种仿过氧化氢酶活性的光催化纳米酶及其制备方法和应用
WO2022183811A1 (fr) * 2021-03-05 2022-09-09 中国科学院深圳先进技术研究院 Nano-enzyme photocatalytique présentant une activité de type catalase, son procédé de préparation et son utilisation
CN114099975A (zh) * 2021-11-15 2022-03-01 北京京东方技术开发有限公司 一种胃部滞留胶囊、腰带及胃部神经刺激的方法
CN114099975B (zh) * 2021-11-15 2023-07-14 北京京东方技术开发有限公司 一种胃部滞留胶囊、腰带及胃部神经刺激的方法
CN115414354A (zh) * 2022-08-29 2022-12-02 西南医科大学 花椒毒素在制备治疗血小板减少症药物中的应用

Similar Documents

Publication Publication Date Title
US11786595B2 (en) Methods for radiotherapy to trigger light activation drugs
US11571587B2 (en) Method for treating a disease, disorder, or condition using inhalation to administer an activatable pharmaceutical agent, an energy modulation agent, or both
US11865359B2 (en) Tumor imaging with x-rays and other high energy sources using as contrast agents photon-emitting phosphors having therapeutic properties
US20210353954A1 (en) Insertion devices and systems for production of emitted light internal to a medium and methods for their use
US10596387B2 (en) Tumor imaging with X-rays and other high energy sources using as contrast agents photon-emitting phosphors having therapeutic properties
WO2019014413A1 (fr) Procédés de radiothérapie pour déclencher l'action de médicaments photo-activables
AU2016206832B2 (en) Non-invasive systems and methods for treatment of a host carrying a virus with photoactivatable drugs
TW200914054A (en) Methods and systems for treating cell proliferation disorders using two-photon simultaneous absorption
JP6965252B2 (ja) X線ソラレン活性化がん治療(x−pact)
US20170157418A1 (en) X-ray psoralen activated cancer therapy (x-pact)

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: 18832434

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: 18832434

Country of ref document: EP

Kind code of ref document: A1