WO2000002590A1 - Radiation and nanoparticles for enhancement of drug delivery in solid tumors - Google Patents

Radiation and nanoparticles for enhancement of drug delivery in solid tumors Download PDF

Info

Publication number
WO2000002590A1
WO2000002590A1 PCT/US1999/015025 US9915025W WO0002590A1 WO 2000002590 A1 WO2000002590 A1 WO 2000002590A1 US 9915025 W US9915025 W US 9915025W WO 0002590 A1 WO0002590 A1 WO 0002590A1
Authority
WO
WIPO (PCT)
Prior art keywords
radiation
tumor
particles
nanoparticles
microparticles
Prior art date
Application number
PCT/US1999/015025
Other languages
French (fr)
Inventor
Rinat O. Esenaliev
Original Assignee
The Board Of Regents Of The University Of Texas System
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 The Board Of Regents Of The University Of Texas System filed Critical The Board Of Regents Of The University Of Texas System
Priority to AU48545/99A priority Critical patent/AU4854599A/en
Publication of WO2000002590A1 publication Critical patent/WO2000002590A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • 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
    • A61K41/0028Disruption, e.g. by heat or ultrasounds, sonophysical or sonochemical activation, e.g. thermosensitive or heat-sensitive liposomes, disruption of calculi with a medicinal preparation and ultrasounds
    • 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
    • A61K41/0038Radiosensitizing, i.e. administration of pharmaceutical agents that enhance the effect of radiotherapy
    • 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
    • A61K41/0047Sonopheresis, i.e. ultrasonically-enhanced transdermal delivery, electroporation of a pharmacologically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • invention relates to a system utilizing interaction of electromagnetic
  • therapeutic agents have to pass through blood vessel wall, interstitial
  • surfactant have prolonged circulation time and selectively accumulate
  • Passive delivery of particles in tumors referred to as “passive” delivery of particles in tumors.
  • active delivery is based on attachment of long-
  • peptide sequences can be used for targeting anti-cancer drugs in
  • the present invention fulfills this
  • the present invention is directed to a method or system of
  • the particles can be attached to
  • the anti-cancer drug is selected from the group
  • oligonucleotide oligonucleotide, a gene-targeting vector and any other macromolecular
  • the tumor occurs in the organ selected
  • the nanoparticles or microparticles are long-
  • the nanoparticles or portions thereof are directed against tumor vasculature.
  • the nanoparticles or portions thereof are directed against tumor vasculature.
  • microparticles can be metal particles, carbon particles, graphite particles, polymer particles loaded with an absorbing dye, liquid
  • the nanoparticle has a diameter from about 0.1 nm to
  • the radiation is optical pulsed radiation
  • optical signal generated from a laser or non-laser source.
  • optical signal generated from a laser or non-laser source.
  • radiation is in the spectral range from 0.2 ⁇ m to 2 ⁇ m and delivered
  • the radiation is ultrasonic radiation generated
  • the ultrasonic radiation is
  • solid tumor comprising a source of radiation; an electronic system or
  • Figure I A shows the targeted (active) delivery of
  • Figure IB shows the interaction of the particles with laser or ultrasonic radiation.
  • Figure 2A shows a laser system for enhancement of drug
  • tumors resulted from cold cavitation and acoustic streaming induced
  • Figure 3A shows interstitial irradiation of a deeply
  • Figure 3B shows laser irradiation
  • 3C shows irradiation of a tumor with particles in a hollow organ by a n
  • ultrasonic transducer inserted in the organ.
  • Figure 4 shows particle penetration in rat liver tissue.
  • Figure 4 A shows the penetration due to irradiation by nanosecond
  • Nd:YAG laser pulses Nd:YAG laser pulses.
  • Figure 4B shows the penetration due to
  • Figure 5 shows fluorescence spectra of fluorescein
  • FITC-Dextran isothiocyanate-dextran
  • Figure 6A shows a gross picture of muscle tissue
  • Figure 6C shows a carbon particle penetration into the tissue after 3 -
  • Figure 6D shows carbon particle penetration in the
  • Figure 7 shows a fluorescence spectra of FITC-Dextran
  • Figure 8 shows particle penetration due to 10-min .
  • Figure 9 shows fluorescence spectra of FITC-Dextran.
  • Figure 9 A shows penetration in the liver tissue due to 10-min .
  • liver, lung, and kidney tissue due to 10-min. sonication.
  • electromagnetic radiation refers to radiation (wave) with electrical and magnetic component which
  • optical ultraviolet, visible, and infrared
  • microwave light
  • radiofrequency radiation wave
  • Nanoparticles refers generally to particles with a
  • Microparticles generally refers to particles with a
  • nanoparticles refers to
  • cavitation refers to a physical
  • acoustic streaming refers to a physical
  • minimum fluence refers to lowest possible
  • microconvection refers to micron- sized
  • antibody coating refers to a coating of a
  • particle migration refers to movement
  • the purpose of the invention is to disclose means for
  • solid tumors such as (1) a tumor blood vessel wall; (2) the
  • nanoparticles and microparticles selectively accumulated in tumor
  • the particles are coated with polyethylene glycol or
  • microbubbles is initiated by the nano- or microparticles w hen
  • Cavitation is known as both "cold” cavitation, induced b y
  • the anti-cancer drug is selected from the group
  • oligonucleotide oligonucleotide, a gene-targeting vector and any other macromolecular
  • the tumor occurs in the organ selected
  • the nanoparticles or microparticles are long
  • microparticles can be metal particles, carbon particles, graphite
  • the nanoparticle has a diameter from about 0.1 nm to
  • the radiation is optical pulsed radiation
  • optical signal generated from a laser or non-laser source.
  • optical signal generated from a laser or non-laser source.
  • radiation is in the spectral range from 0.2 ⁇ m to 2 ⁇ m and delivered
  • the radiation is ultrasonic radiation generated
  • the ultrasonic radiation is
  • solid tumor comprising a source of radiation; an electronic system or
  • cancer drugs comprising the steps of injecting nanoparticles or
  • microparticles to the tumor intravenously and irradiating the tumor
  • 2 time, ⁇ , is estimated as ⁇ ⁇ d /k ⁇ , where d is the dimension of th e
  • k is a coefficient which is dependent on the shape of the
  • is the thermal diffusivity of the medium.
  • a surfactant polyethylene glycol or polyethylene oxide
  • the long-circulating particles are attached to antibodies
  • the long-circulating particles may be used for this purpose.
  • the long-circulating particles may be used for this purpose.
  • the cavitation-induced microconvection provides delivery of the
  • nanosecond Nd:YAG laser radiation with the wavelength of '1064 nm to
  • the system consists of: ( 1 )
  • ultrasonic transducer (or transducer array) for producing ultrasonic
  • optical fibers are their optical properties. To overcome this problem, optical fibers
  • inserted into needles can be used for irradiation of deeply located
  • An optical fiber inserted into a needle is introduced into a tumor an d
  • laser radiation is used in this case because the tumors are irradiated
  • Figure 3C illustrates an ultrasound system utilizing
  • transducer incorporated in an endoscope for irradiation of tumors in
  • the tumor is directly irradiated by the transducer that
  • 3 A, 3B and 3C provide selective irradiation of solid tumors at lower
  • FITC-Dextran dextran
  • Particle penetration is up to 160 ⁇ m .
  • Magnification is x 460. Particle penetration is up to 60 ⁇ m .
  • dextran molecules is obtained upon Nd:YAG laser irradiation (4-fold in
  • Figure 6A shows a
  • carbon particles have numerous pores filled with air that substantially
  • sonicated tissue is greater 6.7 times than the one from the n on-
  • Figure 9B shows fluorescence spectra from liver, lung, an d
  • the present invention utilizes the interaction of laser or
  • the nanosecond Nd:YAG laser with a pulse energy of 1 J and a beam
  • microbubbles induces strong inward momentum and pre s sure
  • thermochemical reactions and ionization induced by the absorbing nanoparticles may also enhance drug delivery in solid tumors.
  • the particles should have high absorption
  • Potential candidates are carbon, graphite, or metal (gold, silver,
  • Solid particles injected intravenously have low
  • particles used for the drug are used for the drug.
  • liposomes or biodegradable polymer particles can be stained
  • the ultrasonic "cold” cavitation enhances drug delivery
  • the cavitation is induced by tensile (negative) pressure of
  • the ultrasonic wave propagates through the skin
  • interstitium are used as the nuclei decreasing cavitation threshold.
  • Porous particles (such as activated carbon particles) with gas-filled
  • pores can substantially lower cavitation threshold because they
  • acoustic impedance is different from that of surrounding medium.
  • the acoustic streaming may also enhance delivery of therapeutic
  • Pulsed microwave and radio-frequency radiation can be any frequency
  • tumor vasculature induced by laser and ultrasonic radiation can b e
  • Esenaliev, et al. Lasers in the Life Science, 1994, 6(3), pp.153-161. Esenaliev, et al., IEEE Trans. Biomed. Eng., 1989, 36, Nol2,1188-1194.

Abstract

The present invention discloses a method/system utilizing interaction of electromagnetic pulses or ultrasonic radiation with nano- and microparticles for enhancement of drug delivery in solid tumors. The particles can be attached to antibodies directed against antigens in tumor vasculature and selectively delivered to tumor blood vessel walls. Cavitation induced by ultrasonic waves or local heating of the particles by pulsed electromagnetic radiation results in perforation of tumor blood vessels, microconvection in the interstitium, and perforation of cancer cell membrane, and therefore, provides enhanced delivery of macromolecular therapeutic agents from blood into cancer cells with minimal thermal and mechanical damage to normal tissues.

Description

RADIATION AND NANOPARTICLES FOR ENHANCEMENT OF DRUG DELIVERY IN SOLID TUMORS
BACKGROUND OF THE INVENTION
Cross-reference to Related Application
This patent application claims benefit of non-provisional
patent application U.S. Serial number 09/112,491 , filed July 9, 1998.
Field of the Invention
The present invention relates generally to the fields of
immunology and cancer therapy. More specifically, the present
invention relates to a system utilizing interaction of electromagnetic
pulses or ultrasonic radiation with nanoparticles to enhance anti-
cancer drug delivery in solid tumors and uses of such a system. Description of the Related Art
Many promising therapeutic agents have been proposed
for cancer therapy for the past two decades. Their potential is proven
in numerous preclinical studies. However, limited success has b een
achieved in solid tumor therapy. The presence of physiological
barriers to drug delivery in tumors substantially limits efficacy of th e
anti-cancer drugs. To penetrate into cancer cells in a solid tumor,
therapeutic agents have to pass through blood vessel wall, interstitial
space, and cancer cell membrane. Penetration of anti-cancer drug s
through these physiological barriers is poor especially for the most
promising macromolecular therapeutic agents such as monoclonal
antibodies, cytokines, antisense oligonucleotides, and gene-targeting
vectors.
Methods have been reported for delivery of anti-cancer
drugs with low molecular weight in solid tumors. Many of them are
based on selective delivery of particles loaded with the drugs i n
tumors. It has been demonstrated that particles coated with a
surfactant have prolonged circulation time and selectively accumulate
in tumors because of increased leakage of tumor vasculature in
comparison with the normal one. These long-circulating particles
avoid rapid clearance by reticuloendothelial system. This approach is
referred to as "passive" delivery of particles in tumors. The "active" delivery is based on attachment of long-
circulating particles to antibodies directed against antigens in tumor
vasculature. Results of studies on animals bearing tumors derived
from human cancer cells demonstrate feasibility of active delivery of
anti-cancer drugs to tumor vasculature. These antibodies and short
peptide sequences can be used for targeting anti-cancer drugs in
patients .
The prior art is deficient in the lack of effective means of
enhancing the delivery of anti-cancer drugs (especially
macromolecular ones) from tumor blood vessels into cancer cells with
minimal damage to normal tissues. The present invention fulfills this
long-standing need and desire in the art.
SUMMARY OF THE INVENTION
The present invention is directed to a method or system of
utilizing the interaction of electromagnetic pulses or ultrasonic
radiation with nanoparticles and microparticles for enhancement of
drug delivery in solid tumors. The particles can be attached to
antibodies directed against antigens in tumor vasculature an d
selectively delivered to tumor blood vessel walls. Cavitation induced by ultrasonic waves or local heating of the particles by pulsed
electromagnetic radiation results in perforation of tumor blood
vessels, microconvection in the interstitium, and perforation of cancer
cell membrane. This method provides enhanced delivery of
macromolecular therapeutic agents from the blood into cancer cells
with minimal thermal and mechanical damage to normal tissues.
In one embodiment of the present invention, there is
provided a method of enhancing anti-cancer drug delivery in a solid
tumor, comprising the steps of administrating at least one anti-cancer
drug to the tumor; injecting nanoparticles or microparticles to th e
tumor intravenously; and irradiating the tumor with radiation.
Generally, the anti-cancer drug is selected from the group
consisting of a monoclonal antibody, a cytokine, an antisense
oligonucleotide, a gene-targeting vector and any other macromolecular
therapeutic agent. Generally, the tumor occurs in the organ selected
from the group consisting of breast, lung, brain, liver, skin, kidney, GI
organ, prostate, bladder, gynecological organ and any other hollow
organ.
Preferably, the nanoparticles or microparticles are long-
circulating particles with or without antibody coating, wherein the
antibody is directed against tumor vasculature. The nanoparticles or
microparticles can be metal particles, carbon particles, graphite particles, polymer particles loaded with an absorbing dye, liquid
particles loaded with an absorbing dye or porous particles having gas-
filled pores. The nanoparticle has a diameter from about 0.1 nm to
about 7000 nm.
Preferably, the radiation is optical pulsed radiation
generated from a laser or non-laser source. Specifically, the optical
radiation is in the spectral range from 0.2 μm to 2 μm and delivered
through the skin surface or via optical fibers inserted in a needle or
endoscopes to the tumor.
Preferably, the radiation is ultrasonic radiation generated
from an ultrasonic transducer. Specifically, the ultrasonic radiation is
in the frequency range from 20 to 500 kHz and delivered through the
skin surface to the tumor.
In another embodiment of the present invention, there is
provided a system for enhancement of anti-cancer drug delivery in a
solid tumor, comprising a source of radiation; an electronic system or
means for monitoring of the radiation; a system or means for delivery
of the radiation to the tumor; nanoparticles or microparticles
absorbing the radiation; an injection system or means for
administration of the anti-cancer drug and the nanoparticles or
microparticles in tumor blood.
In still another embodiment of the present invention, there is provided a method of treating a solid tumor, comprising the
steps of injecting nanoparticles or microparticles to the tumor
intravenously and irradiating the tumor with radiation.
Other and further aspects, features, and advantages of th e
present invention will be apparent from the following description of
the presently preferred embodiments of the invention given for th e
purpose of disclosure.
BRIEF DESCRIPTION OF THE DRAWINGS
So that the matter in which the above-recited features ,
advantages and objects of the invention, as well as others which will
become clear, are attained and can be understood in detail, more
particular descriptions of the invention briefly summarized above
may be had by reference to certain embodiments thereof which are
illustrated in the appended drawings. These drawings form a part of
the specification. It is to be noted, however, that the appended
drawings illustrate preferred embodiments of the invention an d
therefore are . not to be considered limiting in their scope.
Figure I A shows the targeted (active) delivery of
particles to tumor blood vessels. Figure IB shows the interaction of the particles with laser or ultrasonic radiation.
Figure 2A shows a laser system for enhancement of drug
delivery in solid tumors resulted from hot cavitation and local heating
induced by interaction of laser radiation with particles. Figure 2 B
shows an ultrasound system for enhancement of drug delivery in solid
tumors resulted from cold cavitation and acoustic streaming induced
by interaction of ultrasonic radiation with particles.
Figure 3A shows interstitial irradiation of a deeply
located tumor with particles by laser radiation delivered through a n
optical fiber inserted in a needle. Figure 3B shows laser irradiation
of a tumor with particles in a hollow organ via an endoscope. Figure
3C shows irradiation of a tumor with particles in a hollow organ by a n
ultrasonic transducer inserted in the organ.
Figure 4 shows particle penetration in rat liver tissue.
Figure 4 A shows the penetration due to irradiation by nanosecond
Nd:YAG laser pulses. Figure 4B shows the penetration due to
irradiation by nanosecond Alexandrite laser pulses. Figure 4C shows
the penetration without laser irradiation (control).
Figure 5 shows fluorescence spectra of fluorescein
isothiocyanate-dextran (FITC-Dextran) penetrated into the liver tissue
due to irradiation by nanosecond Nd:YAG laser pulses (upper
spectrum), Alexandrite laser pulses (middle spectrum) and without laser irradiation (lower spectrum).
Figure 6A shows a gross picture of muscle tissue with
carbon (left) and graphite (right) particles penetrated in the
interstitium due to sonication for 10 minutes. Figure 6B shows
carbon particle penetration into the tissue due to 10-min. sonication.
Figure 6C shows a carbon particle penetration into the tissue after 3 -
min. sonication. Figure 6D shows carbon particle penetration in the
tissue without sonication.
Figure 7 shows a fluorescence spectra of FITC-Dextran
penetrated in the muscle tissue due to 10-min. sonication and without
sonication.
Figure 8 shows particle penetration due to 10-min .
sonication in liver (Figure 8A ), kidney (Figure 8B) and lung tissues
(Figure 8C) :
Figure 9 shows fluorescence spectra of FITC-Dextran.
Figure 9 A shows penetration in the liver tissue due to 10-min .
sonication and without sonication. Figure 9B shows penetration in
the liver, lung, and kidney tissue due to 10-min. sonication.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, "electromagnetic radiation (wave)" refers to radiation (wave) with electrical and magnetic component which
includes (but not limited to) optical (ultraviolet, visible, and infrared
light), microwave, and radiofrequency radiation (wave).
As used herein, "ultrasound" or "ultrasonic radiation
(wave)" refers to mechanical ("acoustic" or in other terms of
"pressure") wave in a medium in the frequency range from 20 kHz to
about 1 GHz.
"Nanoparticles" refers generally to particles with a
diameter from 0.1 to several hundred of a nanometer. 1 nanometer =
10"9 meter. "Microparticles" generally refers to particles with a
diameter from 0.1 to several hundred of micrometer. 1 micrometer =
10"6 meter. As used herein, the term "nanoparticles" refers to
particles with a diameter from about 0.1 to about 7000 nm.
As used herein, "cavitation" refers to a physical
phenomenon in a liquid or a liquid-like medium (including tissue)
represented by formation of vapor (or gas) bubbles followed b y
growth, oscillation, and collapse of the bubbles.
As used herein, "acoustic streaming" refers to a physical
phenomenon in a liquid or a liquid-like medium represented by flows
of a part of the liquid medium upon irradiation by an ultrasonic wave.
As used herein, "minimal fluence" refers to lowest possible
fluence of laser radiation (measured in J/cm2) to induce drug delivery enhancement without damage to normal tissues.
As used herein, "microconvection" refers to micron- sized
displacement (flow) in liquid. Generally, convection refers to
movement (flow) of a part of a liquid medium.
As used herein, "antibody coating" refers to a coating of a
particle due to attachment of a number of antibody molecules to the
particle.
As used herein, "particle migration" refers to movement
(travel, displacement) of particles. It is cased by cavitation, streaming ,
or local heating effects.
The purpose of the invention is to disclose means for
enhancement of drug delivery through the physiological barriers of
solid tumors, such as (1) a tumor blood vessel wall; (2) the
interstitium; and (3) a cancer cell membrane. Such methods utilize
interaction of electromagnetic or ultrasonic radiation with
nanoparticles and microparticles selectively accumulated in tumor
blood vessels. The interaction alters the physiological properties of
the barriers of solid tumors due to: (1) cavitation induced b y
ultrasonic, pulsed laser, or microwave radiation near the particles; (2)
transient local heating of the particles by laser or microwave
radiation; and (3) acoustic streaming near the particles. The physical
forces result in rupture of tumor blood vessel wall and cancer cell membrane, and microconvection in the interstitium leading to
enhanced delivery of the macromolecular drugs into cancer cells.
Nanoparticles or microparticles having certain physical
properties and capable of inducing cavitation, local heating, or acoustic
streaming upon irradiation by electromagnetic or ultrasonic radiation
are intravenously injected and selectively accumulated in tumor
vasculature. Passive or active delivery of long-circulating particles
provides a selective accumulation of the particles in tumor
vasculature. The particles are coated with polyethylene glycol or
polyethylene oxide to provide long circulation in blood. Recently
discovered antibodies and short peptide sequences can be used for
active delivery of the particles to tumor blood vessels. Experiments
on animal models demonstrated the feasibility of active delivery of
long-circulating nanoparticles loaded with anti-cancer drugs an d
coated with antibodies that results in their selective accumulation in
tumors .
Cavitation (formation, growth, oscillations, and collapse of
microbubbles) is initiated by the nano- or microparticles w hen
ultrasonic or electromagnetic radiation is applied. A source of
radiation is placed on the surface of skin or inserted in a hollow organ
or in the interstitium so that the irradiation of the particles in the
tumor is provided. Cavitation is known as both "cold" cavitation, induced b y
tensile (negative) pressure of ultrasonic waves and "hot" cavitation,
induced by evaporation upon heating of an absorbing medium b y
electromagnetic radiation. Ultrasonic irradiation results in cavitation
near the particles which serves as cavitation nuclei significantly
reducing cavitation threshold. The material, structure, and dimensions
of the particles and ultrasound frequency are specially selected to
provide cavitation effects at minimal power of the ultrasonic wave.
Pulsed electromagnetic (laser or microwave) irradiation
results in evaporation of blood near the particles strongly absorbing
optical or microwave radiation. The material, structure, an d
dimensions of the particles as well as radiation wavelength and pulse
duration are specially selected to provide cavitation effects at minimal
fluence of laser or microwave radiation. This yields cavitation
confined within the tumor and with minimal mechanical or thermal
damage to normal tissues.
Perforation of tumor blood vessel walls is induced by the
growing, oscillating, and collapsing cavitation bubbles. As a result,
anti-cancer drugs circulating in blood penetrate through the tumor
blood vessel wall into the interstitium. The cavitation bubbles also
induce microconvection in the interstitium. Anti-cancer drugs migrate
in the interstitium due to microconvection. Perforation of cell membrane is induced by the cavitation bubbles, which allows th e
anti-cancer drugs to penetrate from the interstitium into the cancer
cells. Therefore, the death of cancer cells in the tumor is caused b y
chemical or biological effects of anti-cancer drugs; mechanical or
thermal damage to the cancer cells from cavitation bubbles; an d
mechanical or thermal damage as well as chemical and biological
damage to the tumor vasculature from cavitation bubbles and the
drugs .
In one embodiment of the present invention, there is
provided a method of enhancing anti-cancer drug delivery in a solid
tumor, comprising the steps of administrating at least one anti-cancer
drug to the tumor; injecting nanoparticles or microparticles to the
tumor intravenously; and irradiating the tumor with radiation.
Generally, the anti-cancer drug is selected from the group
consisting of a monoclonal antibody, a cytokine, an antisense
oligonucleotide, a gene-targeting vector and any other macromolecular
therapeutic agent. Generally, the tumor occurs in the organ selected
from the group consisting of breast, lung, brain, liver, skin, kidney, GI
organ, prostate, bladder, gynecological organ and any other hollow
organ .
Preferably, the nanoparticles or microparticles are long
circulating particles with or without antibody coating, wherein the antibody is directed against tumor vasculature. The nanoparticles or
microparticles can be metal particles, carbon particles, graphite
particles, polymer particles loaded with an absorbing dye, liquid
particles loaded with an absorbing dye or porous particles having gas-
filled pores. The nanoparticle has a diameter from about 0.1 nm to
about 7000 nm.
Preferably, the radiation is optical pulsed radiation
generated from a laser or non-laser source. Specifically, the optical
radiation is in the spectral range from 0.2 μm to 2 μm and delivered
through the skin surface or via optical fibers inserted in a needle or
endoscopes to the tumor.
Preferably, the radiation is ultrasonic radiation generated
from an ultrasonic transducer. Specifically, the ultrasonic radiation is
in the frequency range from 20 to 500 kHz and delivered through th e
skin surface to the tumor.
In another embodiment of the present invention, there is
provided a system for enhancement of anti-cancer drug delivery in a
solid tumor, comprising a source of radiation; an electronic system or
means for monitoring of the radiation; a system for delivery of th e
radiation to the tumor; nanoparticles or microparticles absorbing the
radiation; an injection system or means for administration of the anti-
cancer drug and the nanoparticles or microparticles in tumor blood. In still another embodiment of the present invention,
there is provided a method of treating a solid tumor without anti-
cancer drugs, comprising the steps of injecting nanoparticles or
microparticles to the tumor intravenously and irradiating the tumor
with radiation.
The following examples are given for the purpose of
illustrating various embodiments of the invention and are not me ant
to limit the present invention in any fashion.
EXAMPLE 1
Short Laser Pulses
It is known that severe local heating of strongly absorbing
particles in transparent optical materials produces vapor
microbubbles upon irradiation by short laser pulses, which results i n
mechanical and thermal damage to the materials. Local heating of a
strongly absorbing particle in a medium can be induced, if laser pulse
duration is shorter than the time of heat diffusion. The heat diffusion
2 time, τ, is estimated as τ ~ d /kχ , where d is the dimension of th e
particle, k is a coefficient which is dependent on the shape of the
particle, and χ is the thermal diffusivity of the medium. For example,
local temperature rise above 20,000°K can be achieved in laser glass containing a metal particle with the radius of 200 nm and absorption
coefficient of about 10 cm" ! upon irradiation with nanosecond laser
2 pulses with the fluence of 20 J/cm .
Local heating of exogenous strongly absorbing
nanoparticles by short (nanosecond) and ultrashort (picosecond) laser
pulses results in explosive evaporation of blood in tumor vasculature
and formation of microbubbles.
EXAMPLE 2
Active and Passive Delivery
To avoid rapid clearance by the reticuloendothelial system
and obtain selective accumulation in tumors, long-circulating particles
coated with a surfactant (polyethylene glycol or polyethylene oxide)
are used. The long-circulating particles are attached to antibodies
directed against antigens in tumor vasculature, which results in
selective delivery of the particles to tumor blood vessel walls (active
delivery) (see Figure IA). Short peptide sequences for organ specific
targeting may be used for this purpose. The long-circulating particles
without antibodies also selectively accumulate in tumors because of
leakage through tumor vasculature (passive delivery). Figure IB
shows the interaction of the particles with pulsed laser or ultrasonic radiation. Cavitation induced by laser or ultrasonic radiation results in
perforation of tumor blood vessels and penetration of the particles
and anti-cancer drug into the interstitium due to microconvection.
The cavitation-induced microconvection provides delivery of the
particles and anti-cancer drug through the interstitium to cancer cell
membranes. Perforation of the cell membranes allows the drug
molecules to penetrate into the cancer cells.
EXAMPLE 3
Systems for Enhancing Drug Delivery in Solid Tumors
A laser system for the enhancement of drug delivery in
solid tumors was developed (see Figure 2A). The system utilizes
nanosecond Nd:YAG laser radiation with the wavelength of '1064 nm to
induce local heating of strongly absorbing particles. The s ystem
consists of: (1) Nd:YAG laser with electronic system for monitoring of
radiation parameters (pulse energy and duration, number of pulses ,
repetition rate, power); (2) a fiber-optic system or conventional optical
system for delivery of laser radiation through the skin to the tumor;
(3) nano- or microparticles strongly absorbing laser radiation an d
attached to antibodies directed against tumor vasculature; and (4) a n injection system for administration of the particles and anti-cancer
drugs in blood.
An ultrasound system for the drug delivery enhancement
utilizing ultrasonic waves capable of inducing cavitation in blood an d
tissues was also developed (see Figure 2B). The system consists of: ( 1 )
ultrasonic transducer (or transducer array) for producing ultrasonic
wave with a frequency in the range from 20 kHz to 10 MHz; (2) a n
electronic system for providing power supply for the ultrasonic
transducer and monitoring of radiation parameters (pressure
amplitude, frequency, pulse duration, number of pulses, repetition
rate, ultrasonic power); (3) a focusing system for directing an d
focusing ultrasonic radiation to tumors; (4) nano- or microparticles
decreasing threshold of cavitation upon irradiation by the ultrasonic
radiation and attached to antibodies directed against tumor
vasculature; and (5) an injection system for administration of th e
particles and anti-cancer drugs in blood.
EXAMPLE 4
Interstitial Irradiation of Tumors
Near infra-red radiation with sufficient fluence can penetrate only up to approximately 2 - 4 cm into tissues depending on
their optical properties. To overcome this problem, optical fibers
inserted into needles can be used for irradiation of deeply located
tumors (so-called interstitial irradiation of tumors). Figure 3 A
illustrates a laser system utilizing the interstitial irradiation of tumors .
An optical fiber inserted into a needle is introduced into a tumor an d
laser radiation is not attenuated by tissues between the tumor an d
skin surface. This provides laser irradiation of the tumor with th e
fluence sufficient to enhance drug delivery.
A laser system utilizing irradiation of tumors in the lung,
stomach, intestine, and other hollow organs through optical fibers or
endoscopes is shown in Figure 3B. Near infra-red, visible, or near-UV
laser radiation is used in this case because the tumors are irradiated
directly and without attenuation in the skin and normal tissues.
Figure 3C illustrates an ultrasound system utilizing
transducer incorporated in an endoscope for irradiation of tumors in
hollow organs. The tumor is directly irradiated by the transducer that
allows more efficient drug delivery. The systems shown in the Figures
3 A, 3B and 3C provide selective irradiation of solid tumors at lower
incident laser pulse energy and ultrasonic power that minimizes
damage to normal tissues. EXAMPLE 5
Laser Irradiation-Induced Particle Penetration in Rat Liver Tissue
Freshly excised ex vivo rat liver was used. Activated
carbon particles with the diameter of 1 μm were used as particles
strongly absorbing Nd:YAG laser radiation with the wavelength of
1064 nm. The particles were placed between two liver slabs with th e
dimensions of 10 x 10 mm. The thickness of each slab was 3 mm. The
slabs were irradiated for 10 minutes by the laser pulses with the
energy of 90 mJ, duration of 15 ns, repetition rate of 10 Hz. The laser
spot diameter was 8 mm yielding incident fluence of 0.18 J/cm2. The
slabs were placed in 1% water solution of fluorescein isothiocyanate-
dextran (FITC-Dextran) to study penetration of macromolecules in the
liver tissue. Molecular weight of the FITC-dextran is 12,000. After
irradiation the slabs were rinsed in pure water to remove the particles
and dextran from the tissue surface. Laser-induced explosive
evaporation of water results in penetration and migration of the
particles and dextran in the interstitium (see Figure 4A). The black
spots represent clusters of the carbon particles. Magnification is x
350. Particle penetration is up to 160 μm .
Particle penetration in the rat liver tissue due to
irradiation by nanosecond Alexandrite laser pulses with the wavelength of 720 nm was also studied (Figure 4B). The liver slabs
were irradiated for 10 minutes by the laser pulses with the energy of
90 mJ, duration of 160 ns, repetition rate of 2 Hz. All other conditions
are the same as in the case of Nd:YAG irradiation. The laser spot
diameter was 4 mm yielding incident fluence of 0.72 J/cm2.
Magnification is x 460. Particle penetration is up to 60 μm .
As in a sample without application of laser irradiation
(control), some particles are visible only on the tissue surface (Figure
4C). However, the particles can not penetrate into the interstitium.
Magnification is x 460.
Fluorescence spectra of FITC-Dextran penetrated in the
liver tissue was further studied (Figure 5). All conditions are the
same as described in the Figure 4. The wavelength for the
fluorescence excitation is 400 nm. Increased penetration of th e
dextran molecules is obtained upon Nd:YAG laser irradiation (4-fold in
comparison with the non-irradiated sample). Dextran penetration
upon Alexandrite laser irradiation is also noticeable (2-fold i n
comparison with the non-irradiated sample).
These data demonstrate that interaction of pulsed laser
radiation with strongly absorbing particles results in penetration of
the particles and macromolecules into the interstitium and migration
of the particles and macromolecules in the interstitium. EXAMPLE 6
Sonication-Induced Particle Penetration in Chicken Muscle Tissue
Particle penetration due to sonication was conducted in
chicken muscle tissue (Figures 6A, 6B and 6C). Figure 6A shows a
gross picture of chicken muscle tissue with carbon (left) and graphite
(right) particles penetrated in the interstitium due to sonication for 1 0
minutes. The activated carbon and graphite particles with the
diameter of 1-2 μm were used as cavitation nuclei. The activated
carbon particles have numerous pores filled with air that substantially
decrease the cavitation threshold and enhances cavitation. Ultrasonic
radiation with the frequency of 50 kHz and incident intensity of 250
mW/cm2 was employed for the experiments. Penetration of the
activated carbon particles is more pronounced. No cavitation w as
visible on the tissue surface where the particles were not applied.
Also shown is that carbon particle penetrates into the
tissue up to 180 μm due to 10 minute sonication (Figure 6B,
Magnification: x 230) and up to 60 μm due to 3 minute sonication
(Figure 6C, Magnification: x 300). Particles are visible only on the
tissue surface without penetrating into the interstitium when no
sonication was applied (Figure 6D, Magnification: x 340).
Fluorescence spectra of FITC-Dextran penetrated in th e muscle tissue due to 10-min. sonication and without sonication w as
also conducted (Figure 7). All conditions are the same as in the Figure
6. The data demonstrate more than 6-fold increase of the penetration
of the macromolecules into the tissue which is caused by ultrasound-
induced cavitation. The cavitation results in microconvection in the
tissue leading to enhanced delivery of macromolecules in th e
interstitium.
EXAMPLE 7
Sonication-Induced Particle Penetration in Rat Tissues
Particle penetration due to 10 minute sonication w a s
studied in freshly excised rat tissues ex vivo (Figures 8A, 8B and 8C,
Magnification: x 460). The penetration is up to 160 μm in the liver, u p
to 30 μm in the kidney, and up to 150 μm in the lung tissue.
Fluorescence spectra of FITC-Dextran penetrated in the r a t
liver tissue due to 10-min. sonication and without sonication was
further conducted (Figure 9A). The intensity of fluorescence from the
sonicated tissue is greater 6.7 times than the one from the n on-
sonicated. Figure 9B shows fluorescence spectra from liver, lung, an d
kidney tissue after 10-min. sonication. The intake of the dextran
molecules is almost the s ame for all the tissues. The ultrasound d ata demonstrate that the interaction of the particles with ultrasonic
radiation induces cavitation in tissues and the cavitation results in
penetration of particles and macromolecules into the interstitium.
Discussion
The present invention utilizes the interaction of laser or
ultrasonic radiation with exogenous intravenously injected particles
for the purpose of enhancing drug delivery in solid tumors. The
interaction enhances penetration of anti-cancer drugs, especially
macromolecular therapeutic agents, from blood into cancer cells
preferably due to cavitation accompanied by disruption of tumor
blood vessel walls and cancer cell membranes as well a s
microconvection in the interstitium.
Local heating of exogenous strongly absorbing
nanoparticles by short (nanosecond) and ultrashort (picosecond) laser
pulses results in explosive evaporation of blood in tumor vasculature
and formation of microbubbles. Optical radiation in the near infra-red
and visible spectral range (so-called "therapeutic window": λ = 600 -
1300 nm) has low attenuation in tissues. Therefore, it can induce local
heating of the strongly absorbing particles in deeply located tumors
without damage to irradiated tissue surface. For example, absorption
and scattering coefficients of breast tissue equal to 0.05-0.08 cm an d 5.0-9.0 cm , respectively, in the near infra-red spectral range. Using
- 1 -2 2 these values, one can estimate that the fluence of 10 - 10 J/cm can
be achieved at the depth of 2-3 cm, if breast tissue is irradiated b y
the nanosecond Nd:YAG laser with a pulse energy of 1 J and a beam
diameter of 1 cm. Since temperature rise is proportional to fluence,
local transient temperature of about 50- 140 C can be obtained near
the particles.
Overheating of water above 100°C near absorbing particles
induces local explosive microevaporation resulting in pressure rise,
bubble formation, and outward momentum. Collapse of the
microbubbles induces strong inward momentum and pre s sure
transients. Both the explosive evaporation and the following bubble
collapse perforate tumor vasculature and therefore increase vascular
permeability, which allows therapeutic agents to penetrate from blood
into the interstitium, migrate in the interstitium, and penetrate into
the cancer cells. Moderate heating without evaporation can induce
local thermal damage or local temperature rise without damage which
may also increase vascular permeability. Pulsed laser heating of
absorbing volumes generates thermoelastic pressure transients in
tissues which may contribute to damaging tumor vasculature or
increasing vascular permeability. Possible photochemical an d
thermochemical reactions and ionization induced by the absorbing nanoparticles may also enhance drug delivery in solid tumors.
Low absorption of optical radiation by tissues and strong
absorption by particles enhances drug delivery in tumors without
damage to normal tissues. Laser ablation of the irradiated tissue
2 surface is not induced, because the incident fluence of about 1 J/cm is
substantially lower than the tissue ablation threshold of 40-45 J/cm
at this wavelength. The particles should have high absorption
4 6 - 1 coefficient ( 10 - 10 cm ), low emissivity, short lifetime of excited
states, capability to bind to antibodies, low acute and chronic toxicity.
Potential candidates are carbon, graphite, or metal (gold, silver,
platinum) particles. Solid particles injected intravenously have low
acute and chronic toxicity. Ideally, particles used for the drug
delivery enhancement should be biodegradable. Insoluble liquid (for
instance, liposomes) or biodegradable polymer particles can be stained
with a strongly absorbing dye and used for the drug delivery
enhancement instead of carbon or metal particles.
The ultrasonic "cold" cavitation enhances drug delivery
from blood into cancer cells as in the case of laser-induced "hot"
cavitation. The cavitation is induced by tensile (negative) pressure of
the ultrasonic, wave. The ultrasonic wave propagates through the skin
and normal tissues with insignificant attenuation and induces
cavitation upon interaction with the particles. It is known th at cavitation threshold is substantially lowered by cavitation nuclei. The
particles selectively delivered in tumor blood vessels and the
interstitium are used as the nuclei decreasing cavitation threshold.
Porous particles (such as activated carbon particles) with gas-filled
pores can substantially lower cavitation threshold because they
already have initial ("seed") bubbles.
Combination of laser-induced "seed" cavitation and the
following bubble growth and collapse caused by ultrasonic waves can
be applied for more efficient drug delivery in solid tumors. In this
case, laser radiation initiates bubble formation producing cavitation
nuclei while ultrasonic radiation increases dimensions of the bubbles,
induces their oscillations and collapse resulting in microconvection.
Ultrasonic waves with moderate amplitude induce acoustic
streaming (microconvection) near particles without cavitation, if their
acoustic impedance is different from that of surrounding medium.
The acoustic streaming may also enhance delivery of therapeutic
agents from blood into tumor cells.
Pulsed microwave and radio-frequency radiation can
induce local heating of absorbing particles (e.g., metal), because of its
deep penetration into tissues and strong absorption in metals.
However, the difference in absorption between metals and tissues is
substantially less than in the case of visible or near infra-red radiation. In addition, generation and delivery of powerful short
microwave or radio-frequency pulses to tumors are more complicated
than the generation and delivery of short optical pulses.
Thermal and mechanical damage to endothelial cells of
tumor vasculature induced by laser and ultrasonic radiation can b e
used for cancer treatment without drugs. Such damage results in the
lack of blood supply to the tumor with the following avalanche d eath
of cancer cells. Particle migration in the interstitium caused b y
interaction with laser or ultrasonic radiation induces thermal an d
mechanical damage to cancer cells that may result in death of cancer
cells. Such an approach eliminates side effects of chemotherapy.
The following publications were referred to herein.
US Patent No. 5,474,765 Thorpe
US Patent No. 5,487,390 Cohen et. al.
US Patent No. 5,543,158 Gref et. al.
US Patent No. 5,651 ,986 Brem et. al.
US Patent No. 5,565,215 Gref et. al.
US Patent No. 5,578,325 Domb et. al.
US Patent No. 5,614,502 Flotte et. al.
US Patent No. 5,658,892 Flotte et. al.
US Patent No. 5,660,827 Thorpe et. al.
US Patent No. 5,718,921 Mathiowitz et. al. US Patent No. 5,713,845 Tankovich
US Patent No. 5,762,918 Thorpe
US Patent No. 4,971 ,991 Imemura et. al.
US Patent No. 5,403,590 Forse
US Patent No. 5,380,411 Schlief
Jain R. K. Science v. 271 , pp. 1079-1080, 1996.
Curti B. D. Cancer Chemotherapy and Biotherapy. 1996, pp. 709-719.
Jain R. K. Sci. Am. v. 271 , pp. 58-65, 1994.
Hopper R. W., et al., J. Appl. Phys. v. 41 (10), pp. 4023-4037, 1970.
Oku N., et al, Biol. Pharm. Bull. 20 (6), pp. 670-673, 1997.
Gref R., et al., Science v. 263, pp. 1600-1603, 1994.
Dvorak H. F., et al., Cancer Cells v. 3 (3), pp. 77-85, 1991.
Thorpe, et al., Breast Cane. Res. and Treatm. 36 (2), 237-251 , 1995.
R. Pasqualini, et al., Nature v. 380, pp. 364-366, 1996.
W. Arap, et al., Science v. 279, pp. 377-380, 1998.
Takahashi T., Crit. Rev. Ther. Drug Carr. Syst. 2(3), pp. 245-274, 1986.
Mitragotri S., et al., Science, v. 269, pp. 850-853, 1995.
Johnson M. E., et al, J. Pharm. Sci. v. 85 (7), pp. 670-679, 1996.
Folkman J., Sci. Am. v. 275, pp. 150-154, 1996.
Esenaliev R. O.; et al., Appl. Phys. B v. 59, pp. 73-81 , 1994.
Esenaliev R. O.; et al, Lasers Surg. Med. v. 13, pp. 470-484, 1993.
Esenaliev, et al., Lasers in the Life Science, 1994, 6(3), pp.153-161. Esenaliev, et al., IEEE Trans. Biomed. Eng., 1989, 36, Nol2,1188-1194.
Any patents or publications mentioned in this specification
are indicative of the levels of those skilled in the art to which the
invention pertains. These patents and publications are herein
incorporated by reference to the same extent as if each individual
publication was specifically and individually indicated to b e
incorporated by reference.
One skilled in the art will readily appreciate that the
present invention is well adapted to carry out the objects and obtain
the ends and advantages mentioned, as well as those inherent therein.
The present examples along with the methods, procedures, treatments ,
molecules, and specific compounds described herein are presently
representative of preferred embodiments, are exemplary, and are not
intended as limitations on the scope of the invention. Changes therein
and other uses will occur to those skilled in the art which are
encompassed within the spirit of the invention as defined by the scope
of the claims.

Claims

WHAT IS CLAIMED IS:
1. A method of enhancing anti-cancer drug delivery in
a solid tumor, comprising the steps of:
administering at least one anti-cancer drug to said tumor;
injecting nanoparticles or microparticles to said tumor
intravenously; and
irradiating said tumor with radiation.
2. The method of claim 1 , wherein said anti-cancer
drug is selected from the group consisting of a monoclonal antibody, a
cytokine, an antisense oligonucleotide, and a gene-targeting vector.
3. The method of claim 1, wherein said tumor is in an
organ selected from the group consisting of breast, lung, brain, liver,
skin, kidney, GI organ, prostate, bladder and gynecological organ.
4. The method of claim 1 , wherein said nanoparticles or
microparticles are long-circulating particles with or without antibody coating.
5. The method of claim 4, wherein said antibody is
directed against tumor vasculature.
6. The method of claim 1 , wherein said nanoparticle h as
a diameter from about 0.1 nm to about 7000 nm.
7. The method of claim 1 , wherein said nanoparticle or
microparticle is selected from the group consisting of a metal particle,
a carbon particle, a graphite particle, a polymer particle, a liquid
particle and a porous particle.
8. The method of claim 1 , wherein said radiation is
optical pulsed radiation generated from a laser or non-laser source.
9. The method of claim 8, wherein said optical radiation
is delivered through skin surface or via optical fibers or endoscopes to said tumor
10. The method of claim 8, wherein said optical radiation
is in the spectral range from 0.2 ╬╝m to 2 ╬╝m .
1 1 . The method of claim 1 , wherein said radiation is
utrasonic radiation generated from an ultrasonic transducer.
12. The method of claim 11 , wherein said ultrasonic
radiation is in the frequency range from 20 to 500 kHz.
13. The method of claim 11 , wherein said ultrasonic
radiation is delivered through skin surface to said tumor.
14. An anti-cancer drug delivery system, comprising:
a source of radiation;
an electronic system for monitoring of said radiation; a means for delivery of said radiation to said tumor;
nanoparticles or microparticles absorbing said radiation;
an injection means for administration of said anti-cancer
drug and said nanoparticles or microparticles in blood.
15. A method of treating a solid tumor, comprising the
steps of:
injecting nano- or microparticles to said tumor
intravenously; and
irradiating said tumor with radiation.
PCT/US1999/015025 1998-07-09 1999-07-01 Radiation and nanoparticles for enhancement of drug delivery in solid tumors WO2000002590A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU48545/99A AU4854599A (en) 1998-07-09 1999-07-01 Radiation and nanoparticles for enhancement of drug delivery in solid tumors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/112,491 1998-07-09
US09/112,491 US6165440A (en) 1997-07-09 1998-07-09 Radiation and nanoparticles for enhancement of drug delivery in solid tumors

Publications (1)

Publication Number Publication Date
WO2000002590A1 true WO2000002590A1 (en) 2000-01-20

Family

ID=22344176

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/015025 WO2000002590A1 (en) 1998-07-09 1999-07-01 Radiation and nanoparticles for enhancement of drug delivery in solid tumors

Country Status (3)

Country Link
US (1) US6165440A (en)
AU (1) AU4854599A (en)
WO (1) WO2000002590A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001058458A1 (en) 2000-02-08 2001-08-16 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
US6573491B1 (en) 1999-05-17 2003-06-03 Rock Mountain Biosystems, Inc. Electromagnetic energy driven separation methods
KR100411434B1 (en) * 2000-12-12 2003-12-18 엘지전자 주식회사 Part heating apparatus using a carbon nanotube
EP1779855A1 (en) * 2005-10-28 2007-05-02 Abdula Kurkayev Nanoparticles of a heterocrystal mineral for use as a medicament and method of producing the same
US20090220423A1 (en) * 2005-10-28 2009-09-03 Abdula Kurkayev Method of activating a photosensitizer
US8802154B2 (en) 2010-08-27 2014-08-12 Sienna Labs, Inc. Thermal treatment of a pilosebaceous unit with nanoparticles
WO2015154547A1 (en) * 2014-04-11 2015-10-15 重庆莱美药业股份有限公司 New use of nano carbon injection suspension
US9212294B2 (en) 2012-10-11 2015-12-15 Nanocomposix, Inc. Silver nanoplate compositions and methods
US9572880B2 (en) 2010-08-27 2017-02-21 Sienna Biopharmaceuticals, Inc. Ultrasound delivery of nanoparticles
WO2022234266A1 (en) * 2021-05-05 2022-11-10 Biocompatibles Uk Limited Distributing microparticles

Families Citing this family (152)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6607829B1 (en) 1997-11-13 2003-08-19 Massachusetts Institute Of Technology Tellurium-containing nanocrystalline materials
US6207392B1 (en) 1997-11-25 2001-03-27 The Regents Of The University Of California Semiconductor nanocrystal probes for biological applications and process for making and using such probes
WO2000006244A2 (en) * 1998-07-30 2000-02-10 Hainfeld James F Loading metal particles into cell membrane vesicles and metal particle use for imaging and therapy
JP2002534218A (en) 1999-01-15 2002-10-15 ライト サイエンシーズ コーポレイション Non-invasive vascular therapy
US7914442B1 (en) 1999-03-01 2011-03-29 Gazdzinski Robert F Endoscopic smart probe and method
US8068897B1 (en) 1999-03-01 2011-11-29 Gazdzinski Robert F Endoscopic smart probe and method
US8636648B2 (en) 1999-03-01 2014-01-28 West View Research, Llc Endoscopic smart probe
US10973397B2 (en) 1999-03-01 2021-04-13 West View Research, Llc Computerized information collection and processing apparatus
US6532387B1 (en) * 1999-03-26 2003-03-11 Kevin S. Marchitto Catheter for delivering electromagnetic energy for enhanced permeation of substances
US8119101B2 (en) 1999-05-10 2012-02-21 The Ohio State University Anti-CD74 immunoconjugates and methods of use
US8383081B2 (en) * 1999-05-10 2013-02-26 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods of use
US7829064B2 (en) * 1999-05-10 2010-11-09 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods
US6530944B2 (en) 2000-02-08 2003-03-11 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
MXPA02008361A (en) 2000-02-28 2004-05-17 Genesegues Inc Nanocapsule encapsulation system and method.
US7137968B1 (en) * 2000-03-13 2006-11-21 Nucryst Pharmaceuticals Corp. Transcutaneous medical device dressings and method of use
US6719987B2 (en) 2000-04-17 2004-04-13 Nucryst Pharmaceuticals Corp. Antimicrobial bioabsorbable materials
US7427416B2 (en) * 2000-07-27 2008-09-23 Nucryst Pharmaceuticals Corp. Methods of treating conditions using metal-containing materials
US20030180379A1 (en) 2000-07-27 2003-09-25 Burrell Robert E. Solutions and aerosols of metal-containing compounds
US20030185901A1 (en) 2000-07-27 2003-10-02 Burrell Robert E. Methods of treating conditions with a metal-containing material
US6989157B2 (en) * 2000-07-27 2006-01-24 Nucryst Pharmaceuticals Corp. Dry powders of metal-containing compounds
US7001617B2 (en) 2001-04-23 2006-02-21 Nueryst Pharmaceuticals Corp. Method of induction of apoptosis and inhibition of matrix metalloproteinases using antimicrobial metals
US20030206966A1 (en) * 2000-07-27 2003-11-06 Burrell Robert E. Methods of inducing apoptosis and modulating metalloproteinases
AU7832201A (en) 2000-07-27 2002-02-13 Nucryst Pharm Corp Treatment of hyperproliferative skin disorders and diseases
US7255881B2 (en) * 2000-07-27 2007-08-14 Nucryst Pharmaceuticals Corp. Metal-containing materials
US7008647B2 (en) * 2001-04-23 2006-03-07 Nucryst Pharmaceuticals Corp. Treatment of acne
US6416492B1 (en) 2000-09-28 2002-07-09 Scimed Life Systems, Inc. Radiation delivery system utilizing intravascular ultrasound
US6487447B1 (en) * 2000-10-17 2002-11-26 Ultra-Sonic Technologies, L.L.C. Method and apparatus for in-vivo transdermal and/or intradermal delivery of drugs by sonoporation
AUPR098300A0 (en) 2000-10-25 2000-11-16 Sirtex Medical Limited Polymer based radionuclide containing microspheres
EP1251707A3 (en) * 2001-04-20 2003-09-24 Matsushita Electric Industrial Co., Ltd. Vacant channel searching method
WO2002085386A2 (en) 2001-04-23 2002-10-31 Nucryst Pharmaceuticals Corp. Medicament containing a metal such as silver, gold, platinum or palladium as an antimicrobial agent and their use to induce apoptosis in cancerous tissue
US7074175B2 (en) 2001-07-25 2006-07-11 Erik Schroeder Handy Thermotherapy via targeted delivery of nanoscale magnetic particles
US6997863B2 (en) * 2001-07-25 2006-02-14 Triton Biosystems, Inc. Thermotherapy via targeted delivery of nanoscale magnetic particles
US7951061B2 (en) * 2001-07-25 2011-05-31 Allan Foreman Devices for targeted delivery of thermotherapy, and methods related thereto
US7731648B2 (en) * 2001-07-25 2010-06-08 Aduro Biotech Magnetic nanoscale particle compositions, and therapeutic methods related thereto
US20040156852A1 (en) * 2003-02-06 2004-08-12 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles
EP1437975B1 (en) * 2001-09-26 2011-08-10 Rice University Optically-absorbing nanoparticles for enhanced tissue repair
US9770517B2 (en) 2002-03-01 2017-09-26 Immunomedics, Inc. Anti-Trop-2 antibody-drug conjugates and uses thereof
US6962685B2 (en) 2002-04-17 2005-11-08 International Business Machines Corporation Synthesis of magnetite nanoparticles and the process of forming Fe-based nanomaterials
US7201925B2 (en) * 2002-04-23 2007-04-10 Nueryst Pharmaceuticals Corp. Treatment of ungual and subungual diseases
US7549985B2 (en) * 2002-06-26 2009-06-23 The Regents Of The University Of Michigan Method and system to create and acoustically manipulate a microbubble
US7367948B2 (en) * 2002-08-29 2008-05-06 The Regents Of The University Of Michigan Acoustic monitoring method and system in laser-induced optical breakdown (LIOB)
AU2003286575A1 (en) * 2002-10-22 2004-05-13 Nucryst Pharmaceuticals Corp. Prophylactic treatment methods
US8420086B2 (en) 2002-12-13 2013-04-16 Immunomedics, Inc. Camptothecin conjugates of anti-CD22 antibodies for treatment of B cell diseases
US7666410B2 (en) 2002-12-20 2010-02-23 Kimberly-Clark Worldwide, Inc. Delivery system for functional compounds
US6780896B2 (en) 2002-12-20 2004-08-24 Kimberly-Clark Worldwide, Inc. Stabilized photoinitiators and applications thereof
US8409618B2 (en) 2002-12-20 2013-04-02 Kimberly-Clark Worldwide, Inc. Odor-reducing quinone compounds
US20040156846A1 (en) * 2003-02-06 2004-08-12 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles using L6 antibodies
US7181266B2 (en) * 2003-03-04 2007-02-20 Massachusetts Institute Of Technology Materials and methods for near-infrared and infrared lymph node mapping
EP1620079A1 (en) * 2003-04-03 2006-02-01 Semafore Pharmaceuticals, Inc. Bone targeting of biodegradable drug-containing nanoparticles
US20040198783A1 (en) * 2003-04-03 2004-10-07 Semafore Pharmaceuticals Inc. Targeted bone marrow protection agents
US7430445B2 (en) * 2003-04-24 2008-09-30 The Board Of Regents Of The University Of Texas System Noninvasive blood analysis by optical probing of the veins under the tongue
US7290197B2 (en) * 2003-06-03 2007-10-30 Quantum Corporation Correcting data using redundancy blocks
US20070122529A1 (en) * 2003-08-21 2007-05-31 Advanced Nutri-Tech Systems Inc. Fruit sponge
US7608240B2 (en) * 2003-12-05 2009-10-27 Board Of Trustees Of The University Of Arkansas Nanotubes for cancer therapy and diagnostics
WO2005070473A1 (en) 2004-01-15 2005-08-04 Koninklijke Philips Electronics N.V. Ultrasound contrast agents for molecular imaging
US9550838B2 (en) 2004-02-13 2017-01-24 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for therapeutic and diagnostic use
US8883160B2 (en) * 2004-02-13 2014-11-11 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for therapeutic and diagnostic use
US7627381B2 (en) * 2004-05-07 2009-12-01 Therm Med, Llc Systems and methods for combined RF-induced hyperthermia and radioimmunotherapy
US20050251233A1 (en) * 2004-05-07 2005-11-10 John Kanzius System and method for RF-induced hyperthermia
US7510555B2 (en) * 2004-05-07 2009-03-31 Therm Med, Llc Enhanced systems and methods for RF-induced hyperthermia
US20070250139A1 (en) * 2004-05-07 2007-10-25 John Kanzius Enhanced systems and methods for RF-induced hyperthermia II
US20050260190A1 (en) * 2004-05-19 2005-11-24 Webber Robert J Therapeutic agent for iNOS generating illness
US7794490B2 (en) * 2004-06-22 2010-09-14 Boston Scientific Scimed, Inc. Implantable medical devices with antimicrobial and biodegradable matrices
US7356368B2 (en) * 2004-07-21 2008-04-08 Boston Scientific Scimed, Inc. Light-activated anti-infective coatings and devices made thereof
EP1819277A4 (en) * 2004-11-12 2010-05-05 Ltd Kpe Nanoparticle mediated ultrasound therapy and diagnostic imaging
EP1824521B1 (en) * 2004-11-19 2015-01-07 Koninklijke Philips N.V. Ultrasound contrast agents for molecular imaging
US7541028B2 (en) 2005-01-04 2009-06-02 Gp Medical, Inc. Nanoparticles for monoclonal antibody delivery
US8323976B2 (en) * 2005-01-19 2012-12-04 International Technology Center Alterations utilizing nanoparticles
US7999161B2 (en) * 2005-01-22 2011-08-16 Alexander Oraevsky Laser-activated nanothermolysis of cells
US10058621B2 (en) 2015-06-25 2018-08-28 Immunomedics, Inc. Combination therapy with anti-HLA-DR antibodies and kinase inhibitors in hematopoietic cancers
US9707302B2 (en) 2013-07-23 2017-07-18 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US8475794B2 (en) 2005-04-06 2013-07-02 Ibc Pharmaceuticals, Inc. Combination therapy with anti-CD74 antibodies provides enhanced toxicity to malignancies, Autoimmune disease and other diseases
US8349332B2 (en) 2005-04-06 2013-01-08 Ibc Pharmaceuticals, Inc. Multiple signaling pathways induced by hexavalent, monospecific and bispecific antibodies for enhanced toxicity to B-cell lymphomas and other diseases
US20090022785A1 (en) * 2005-05-03 2009-01-22 Veterinarmedizinische Universitat Wien Permeable Capsules
JP4911915B2 (en) * 2005-05-09 2012-04-04 トヨタ自動車株式会社 Target decomposition method and decomposition apparatus
US20080184618A1 (en) * 2005-08-03 2008-08-07 Amcol International Virus-Interacting Layered Phyllosilicates and Methods of Use
US20100272769A1 (en) * 2005-08-03 2010-10-28 Amcol International Virus-, Bacteria-, and Fungi-Interacting Layered Phyllosilicates and Methods of Use
US20070031512A1 (en) * 2005-08-03 2007-02-08 Amcol International Corporation Virus-interacting layered phyllosilicates and methods of inactivating viruses
US20070071683A1 (en) * 2005-09-27 2007-03-29 The Regents Of The University Of California Ultrasonic concentration of carrier particles
US20070078290A1 (en) * 2005-09-30 2007-04-05 Esenaliev Rinat O Ultrasound-based treatment methods for therapeutic treatment of skin and subcutaneous tissues
WO2007142674A2 (en) * 2005-10-27 2007-12-13 Massachusetts Institute Of Technology Nanoparticle heating and applications thereof
US20070167798A1 (en) * 2005-11-23 2007-07-19 Cai Anming H Contrast agent augmented ultrasound therapy system with ultrasound imaging guidance for thrombus treatment
MX2008007748A (en) * 2005-12-14 2009-02-10 Mobilestream Oil Inc Microwave-based recovery of hydrocarbons and fossil fuels.
US7947368B2 (en) 2005-12-21 2011-05-24 Boston Scientific Scimed, Inc. Block copolymer particles
US8135450B2 (en) * 2006-01-20 2012-03-13 Esenaliev Rinat O Noninvasive glucose sensing methods and systems
US7927465B2 (en) * 2006-02-02 2011-04-19 Novak John F Method and apparatus for microwave reduction of organic compounds
US20070184085A1 (en) * 2006-02-03 2007-08-09 Boston Scientific Scimed, Inc. Ultrasound activated medical device
WO2008014060A2 (en) * 2006-07-27 2008-01-31 Boston Scientific Limited Microparticle
US7892520B2 (en) * 2006-07-31 2011-02-22 The Hong Kong University Of Science And Technology Solid-state synthesis of iron oxide nanoparticles
CN101553889A (en) 2006-09-05 2009-10-07 哥伦布纳米制品公司 Magnetic particles and methods of making and using the same
WO2008127743A2 (en) * 2007-01-05 2008-10-23 William Marsh Rice University Composition for targeted drug delivery and controlled release
US9493817B2 (en) * 2007-03-05 2016-11-15 Genesis Research Institute, Inc. Decomposition method and decomposition apparatus for nucleic acid polymer
WO2008147807A2 (en) 2007-05-23 2008-12-04 Amcol International Corporation Cholesterol-interacting layered phyllosilicates and methods of reducing hypercholesteremia in a mammal
US8784846B2 (en) * 2007-07-30 2014-07-22 Loma Linda University Medical Center Systems and methods for particle radiation enhanced delivery of therapy
JP2010539245A (en) 2007-09-14 2010-12-16 日東電工株式会社 Drug carrier
US8507556B2 (en) 2007-09-22 2013-08-13 Boise State University Fluorescent particles comprising nanoscale ZnO layer and exhibiting cell-specific toxicity
WO2009039508A2 (en) * 2007-09-22 2009-03-26 Boise State University Preferential killing of cancer cells and activated human t cells using zno nanoparticles
US9107858B2 (en) * 2007-12-05 2015-08-18 Wisconsin Alumni Research Foundation Dendritic cell targeting compositions and uses thereof
US20110052672A1 (en) * 2008-01-16 2011-03-03 Sunil Krishnan Treatments of disease or disorders using nanoparticles for focused hyperthermia to increase therapy efficacy
CN102056563A (en) * 2008-04-09 2011-05-11 康奈尔大学 Nanoparticle-mediated microwave treatment methods
US20110212163A1 (en) * 2008-07-24 2011-09-01 Children's Medical Center Corporation Magnetic heating for drug delivery and other applications
WO2010011326A2 (en) * 2008-07-24 2010-01-28 Children's Medical Center Corporation Heating of polymers-and other materials using radiation for drug delivery and other applications
US20110212027A1 (en) * 2008-07-24 2011-09-01 Children's Medical Center Corporation Radiative heating for drug delivery and other applications
US8961577B2 (en) 2009-04-02 2015-02-24 Julie Ann Reil Correction of female urinary incontinence and skin reduction
US20110245587A1 (en) * 2010-04-05 2011-10-06 Julie Ann Reil Method for correction of female urinary incontinence and skin reduction
US9504824B2 (en) * 2009-06-23 2016-11-29 Board Of Regents, The University Of Texas System Noninvasive therapies in the absence or presence of exogenous particulate agents
US10105317B2 (en) 2009-07-07 2018-10-23 Anpac Bio-Medical Science Co., Ltd. Method of drug delivery
US8565892B2 (en) * 2009-10-31 2013-10-22 Qteris, Inc. Nanoparticle-sized magnetic absorption enhancers having three-dimensional geometries adapted for improved diagnostics and hyperthermic treatment
CA2787054A1 (en) * 2010-01-11 2011-07-14 Center For Molecular Medicine And Immunology Enhanced cytotoxicity of anti-cd74 and anti-hla-dr antibodies with interferon-gamma
US8828246B2 (en) 2010-02-18 2014-09-09 Anpac Bio-Medical Science Co., Ltd. Method of fabricating micro-devices
US8865123B1 (en) 2010-09-16 2014-10-21 Mo-Sci Corporation Strontium phosphate microparticle for radiological imaging and therapy
US9849200B2 (en) 2010-09-16 2017-12-26 Mo-Sci Corporation Strontium phosphate microparticle for radiological imaging and therapy
US9119887B2 (en) 2010-09-16 2015-09-01 Mo-Sci Corporation Low-density magnesium-aluminum-silicate (MAS) microparticles for radiotherapy and/or radioimaging
GB201019434D0 (en) * 2010-11-17 2010-12-29 Isis Innovation Sonosensitive nanoparticles
US8757166B2 (en) 2011-01-24 2014-06-24 Actium BioSystems, LLC System for defining energy field characteristics to illuminate nano-particles used to treat invasive agents
US8968171B2 (en) 2011-01-24 2015-03-03 Endomagnetics Limited System for correlating energy field characteristics with target particle characteristics in the application of an energy field to a living organism for imaging and treatment of invasive agents
US20120190911A1 (en) * 2011-01-24 2012-07-26 Actium BioSystems, LLC Low temperature hyperthermia system for therapeutic treatment of invasive agents
US20120190979A1 (en) 2011-01-24 2012-07-26 Actium BioSystems, LLC System for automatically amending energy field characteristics in the application of an energy field to a living organism for treatment of invasive agents
RU2470377C1 (en) * 2011-05-13 2012-12-20 Федеральное государственное бюджетное учреждение "Ростовский научно-исследовательский онкологический институт" Министерства здравоохранения и социального развития Российской Федерации (ФГБУ "РНИОИ" Минздравсоцразвития России) Method of preventing tumour growth in experiment
US9687668B2 (en) 2011-08-26 2017-06-27 Endomagnetics Limited Treatment of cancer in body cavities and parts that are cavity-like
MX347548B (en) * 2011-09-29 2017-05-02 Koninklijke Philips Nv Ultrasound mediated delivery with critical-organ protection.
EP2760368A4 (en) * 2011-09-30 2015-08-26 Biolase Inc Pressure wave root canal cleaning system
WO2013085893A1 (en) 2011-12-05 2013-06-13 Immunomedics, Inc. Therapeutic use of anti-cd22 antibodies for inducing trogocytosis
US9757458B2 (en) 2011-12-05 2017-09-12 Immunomedics, Inc. Crosslinking of CD22 by epratuzumab triggers BCR signaling and caspase-dependent apoptosis in hematopoietic cancer cells
WO2013166487A1 (en) 2012-05-04 2013-11-07 Yale University Highly penetrative nanocarriers for treatment of cns disease
US9522289B2 (en) 2012-05-08 2016-12-20 The Regents Of The University Of California Selective fat removal using photothermal heating
CN104321108B (en) 2012-05-08 2018-05-01 加利福尼亚大学董事会 Controlled using the fine space-time of NIR light pyrolysis and lipolysis
SI2900277T1 (en) 2012-12-13 2022-05-31 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
US10206918B2 (en) 2012-12-13 2019-02-19 Immunomedics, Inc. Efficacy of anti-HLA-DR antiboddy drug conjugate IMMU-140 (hL243-CL2A-SN-38) in HLA-DR positive cancers
US10744129B2 (en) 2012-12-13 2020-08-18 Immunomedics, Inc. Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting Trop-2
WO2015012904A2 (en) 2012-12-13 2015-01-29 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
US9931417B2 (en) 2012-12-13 2018-04-03 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US9492566B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US10413539B2 (en) 2012-12-13 2019-09-17 Immunomedics, Inc. Therapy for metastatic urothelial cancer with the antibody-drug conjugate, sacituzumab govitecan (IMMU-132)
US10137196B2 (en) 2012-12-13 2018-11-27 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US20140271453A1 (en) 2013-03-14 2014-09-18 Abbott Laboratories Methods for the early detection of lung cancer
US11253606B2 (en) 2013-07-23 2022-02-22 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, Bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US10064940B2 (en) 2013-12-11 2018-09-04 Siva Therapeutics Inc. Multifunctional radiation delivery apparatus and method
WO2016015015A1 (en) * 2014-07-24 2016-01-28 Baylor College Of Medicine Non-invasive radiofrequency field treatment for cancer therapy
SG10201902499VA (en) 2014-09-03 2019-04-29 Genesegues Inc Therapeutic nanoparticles and related compositions, methods and systems
JP6746845B2 (en) 2015-04-22 2020-08-26 イミューノメディクス、インコーポレイテッドImmunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating TROP-2 positive cancer cells
US10022333B2 (en) * 2015-06-10 2018-07-17 Siemens Medical Solutions Usa, Inc. Image-guided delivery of a mixture of bacteria and non-bacteria linked nanoparticles
US10195175B2 (en) 2015-06-25 2019-02-05 Immunomedics, Inc. Synergistic effect of anti-Trop-2 antibody-drug conjugate in combination therapy for triple-negative breast cancer when used with microtubule inhibitors or PARP inhibitors
AU2016323806A1 (en) 2015-09-16 2018-05-10 La Jolla Nanomedical A cellular activity targeted nanoparticle system and methods of producing the nanoparticle system
WO2017139623A1 (en) 2016-02-10 2017-08-17 Immunomedics, Inc. Combination of abcg2 inhibitors with sacituzumab govitecan (immu-132) overcomes resistance to sn-38 in trop-2 expressing cancers
CA3016917A1 (en) 2016-04-27 2017-11-02 Immunomedics, Inc. Efficacy of anti-trop-2-sn-38 antibody drug conjugates for therapy of tumors relapsed/refractory to checkpoint inhibitors
AU2017368332A1 (en) 2016-12-03 2019-06-13 Juno Therapeutics, Inc. Methods for modulation of CAR-T cells
JP7247090B2 (en) 2016-12-16 2023-03-28 ナノスペクトラ バイオサイエンセズ インコーポレイテッド Apparatus and its use in methods for ablation therapy
CA3050332A1 (en) 2017-03-27 2018-10-04 Immunomedics, Inc. Treatment of trop-2 expressing triple negative breast cancer with sacituzumab govitecan and a rad51 inhibitor
CN110352201A (en) 2017-04-03 2019-10-18 免疫医疗公司 The subcutaneous administration of antibody drug conjugate for cancer therapy
WO2019046304A1 (en) 2017-08-28 2019-03-07 Matthias Wagner Microfluidic laser-activated intracellular delivery systems and methods
US11701035B2 (en) 2019-10-28 2023-07-18 Bruce M. Landy Noninvasive blood glucose detector and method using IR

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5599712A (en) * 1993-10-15 1997-02-04 University Of Pittsburgh Protection from ionizing irradiation or chemotherapeutic drug damage by in vivo gene therapy
US5836905A (en) * 1994-06-20 1998-11-17 Lemelson; Jerome H. Apparatus and methods for gene therapy
WO1998053852A1 (en) * 1997-05-30 1998-12-03 Arch Development Corporation P-selectin translocation to vascular epithelial lumen by ionizing radiation

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0629196B2 (en) * 1987-12-01 1994-04-20 甲子郎 梅村 Physiological action enhancer for tumor treatment by ultrasound
DE3741201A1 (en) * 1987-12-02 1989-06-15 Schering Ag ULTRASONIC PROCESS AND METHOD FOR IMPLEMENTING IT
US5487390A (en) * 1990-10-05 1996-01-30 Massachusetts Institute Of Technology Gas-filled polymeric microbubbles for ultrasound imaging
US5474765A (en) * 1992-03-23 1995-12-12 Ut Sw Medical Ctr At Dallas Preparation and use of steroid-polyanionic polymer-based conjugates targeted to vascular endothelial cells
US5403590A (en) * 1992-12-21 1995-04-04 New England Deaconess Hospital Corporation Method of pulsatile drug infusion
US5614502A (en) * 1993-01-15 1997-03-25 The General Hospital Corporation High-pressure impulse transient drug delivery for the treatment of proliferative diseases
US5565215A (en) * 1993-07-23 1996-10-15 Massachusettes Institute Of Technology Biodegradable injectable particles for imaging
US5543158A (en) * 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5626862A (en) * 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5599712A (en) * 1993-10-15 1997-02-04 University Of Pittsburgh Protection from ionizing irradiation or chemotherapeutic drug damage by in vivo gene therapy
US5836905A (en) * 1994-06-20 1998-11-17 Lemelson; Jerome H. Apparatus and methods for gene therapy
WO1998053852A1 (en) * 1997-05-30 1998-12-03 Arch Development Corporation P-selectin translocation to vascular epithelial lumen by ionizing radiation

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6573491B1 (en) 1999-05-17 2003-06-03 Rock Mountain Biosystems, Inc. Electromagnetic energy driven separation methods
EP2343047B1 (en) 2000-02-08 2016-01-06 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
EP1263447A1 (en) * 2000-02-08 2002-12-11 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
JP2003522149A (en) * 2000-02-08 2003-07-22 ライスユニバーシティ Optically active nanoparticles used in therapeutics and diagnostics
EP2343047B2 (en) 2000-02-08 2021-03-17 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
EP2343047A3 (en) * 2000-02-08 2012-12-26 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
WO2001058458A1 (en) 2000-02-08 2001-08-16 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
EP1263447A4 (en) * 2000-02-08 2003-05-07 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
KR100411434B1 (en) * 2000-12-12 2003-12-18 엘지전자 주식회사 Part heating apparatus using a carbon nanotube
WO2007048634A2 (en) 2005-10-28 2007-05-03 Abdula Kurkayev Nanoparticles of a heterocrystal mineral for use as a medicament and method of producing the same
EP1779855A1 (en) * 2005-10-28 2007-05-02 Abdula Kurkayev Nanoparticles of a heterocrystal mineral for use as a medicament and method of producing the same
WO2007048634A3 (en) * 2005-10-28 2007-12-06 Abdula Kurkayev Nanoparticles of a heterocrystal mineral for use as a medicament and method of producing the same
US20090220423A1 (en) * 2005-10-28 2009-09-03 Abdula Kurkayev Method of activating a photosensitizer
US9439964B2 (en) 2010-08-27 2016-09-13 Sienna Biopharmaceuticals, Inc. Thermal treatment of the skin surface with coated metal nanoparticles
US9421259B2 (en) 2010-08-27 2016-08-23 Sienna Biopharmaceuticals, Inc. Hair removal with coated metal nanoparticles
US8895071B1 (en) 2010-08-27 2014-11-25 Sienna Labs, Inc. Thermal treatment of a pilosebaceous unit with coated metal nanoparticles
US8906418B1 (en) 2010-08-27 2014-12-09 Sienna Labs, Inc. Thermal treatment of a pilosebaceous unit with nanoparticles with coatings that facilitate selective removal from the skin surface
US9061056B2 (en) 2010-08-27 2015-06-23 Sienna Labs, Inc. Compositions and methods for targeted thermomodulation
US11826087B2 (en) 2010-08-27 2023-11-28 Coronado Aesthetics, Llc Compositions and methods for thermal skin treatment with metal nanoparticles
US8821941B2 (en) 2010-08-27 2014-09-02 Sienna Labs, Inc. Hair removal with nanoparticles
US8834933B2 (en) 2010-08-27 2014-09-16 Sienna Labs, Inc. Thermal treatment of acne with nanoparticles
US11419937B2 (en) 2010-08-27 2022-08-23 Coronado Aesthetics, Llc Delivery of nanoparticles
US9421261B2 (en) 2010-08-27 2016-08-23 Sienna Biopharmaceuticals, Inc. Thermal treatment of the skin surface with nanoparticles with coatings that facilitate selective removal from the skin surface
US9421260B2 (en) 2010-08-27 2016-08-23 Sienna Biopharmaceuticals, Inc. Thermal treatment of acne with nanoparticles with coatings that facilitate selective removal from the skin surface
US10537640B2 (en) 2010-08-27 2020-01-21 Sienna Biopharmaceuticals, Inc. Ultrasound delivery of nanoparticles
US9427467B2 (en) 2010-08-27 2016-08-30 Sienna Biopharmaceuticals, Inc. Hair removal with metal nanoparticles in surfactant containing solutions
US9433676B2 (en) 2010-08-27 2016-09-06 Sienna Biopharmaceuticals, Inc. Hair removal with nanoparticles with coatings that facilitate selective removal from the skin surface
US9433677B2 (en) 2010-08-27 2016-09-06 Sienna Biopharmaceuticals, Inc. Thermal treatment of a pilosebaceous unit with metal nanoparticles in surfactant containing solutions
US9433678B2 (en) 2010-08-27 2016-09-06 Sienna Biopharmaceuticals, Inc. Thermal treatment of acne with metal nanoparticles in surfactant containing solutions
US8821940B2 (en) 2010-08-27 2014-09-02 Sienna Labs, Inc. Thermal treatment of the skin surface with nanoparticles
US9439965B2 (en) 2010-08-27 2016-09-13 Sienna Biopharmaceuticals, Inc. Thermal treatment of the skin surface with metal nanoparticles in surfactant containing solutions
US9446126B2 (en) 2010-08-27 2016-09-20 Sienna Biopharmaceuticals, Inc. Thermal treatment of acne with coated metal nanoparticles
US8802154B2 (en) 2010-08-27 2014-08-12 Sienna Labs, Inc. Thermal treatment of a pilosebaceous unit with nanoparticles
US9572880B2 (en) 2010-08-27 2017-02-21 Sienna Biopharmaceuticals, Inc. Ultrasound delivery of nanoparticles
US10688126B2 (en) 2012-10-11 2020-06-23 Nanocomposix, Inc. Silver nanoplate compositions and methods
US9526745B2 (en) 2012-10-11 2016-12-27 Nanocomposix, Inc. Silver nanoplate compositions and methods
US9212294B2 (en) 2012-10-11 2015-12-15 Nanocomposix, Inc. Silver nanoplate compositions and methods
US11583553B2 (en) 2012-10-11 2023-02-21 Nanocomposix, Llc Silver nanoplate compositions and methods
US9249334B2 (en) 2012-10-11 2016-02-02 Nanocomposix, Inc. Silver nanoplate compositions and methods
WO2015154547A1 (en) * 2014-04-11 2015-10-15 重庆莱美药业股份有限公司 New use of nano carbon injection suspension
WO2022234266A1 (en) * 2021-05-05 2022-11-10 Biocompatibles Uk Limited Distributing microparticles

Also Published As

Publication number Publication date
US6165440A (en) 2000-12-26
AU4854599A (en) 2000-02-01

Similar Documents

Publication Publication Date Title
US6165440A (en) Radiation and nanoparticles for enhancement of drug delivery in solid tumors
Qin et al. Thermophysical and biological responses of gold nanoparticle laser heating
US6484052B1 (en) Optically generated ultrasound for enhanced drug delivery
Lapotko Plasmonic nanoparticle-generated photothermal bubbles and their biomedical applications
JP6224773B2 (en) Compositions and methods for targeted thermal modulation
Frenkel et al. Delivery of liposomal doxorubicin (Doxil) in a breast cancer tumor model: investigation of potential enhancement by pulsed-high intensity focused ultrasound exposure
US6424863B1 (en) Delivery of pharmaceutical compounds and collection of biomolecules using electromagnetic energy and uses thereof
Larina et al. Enhancement of drug delivery in tumors by using interaction of nanoparticles with ultrasound radiation
US7020516B2 (en) Catheter for delivering electromagnetic energy for enhanced permeation of substances
Thanou et al. MRI-guided focused ultrasound as a new method of drug delivery
Yao et al. Laser-based gene transfection and gene therapy
AU2001287264A1 (en) Catheter for delivering electromagnetic energy for enhanced permeation of substances
Wang et al. Treating tumors with minimally invasive therapy: A review
US10518096B2 (en) Noninvasive therapies in the treatment of pathogenic infections
Wang et al. Advances in low-frequency ultrasound combined with microbubbles in targeted tumor therapy
KR20160119076A (en) Treatment intervals for use of compositions comprising energy absorbing materials for dermatological applications
US20160030726A1 (en) Methods of delivering nanoshells into sebaceous glands
KR20180069106A (en) How to treat skin condition using plasmonic nanoparticles
Hu et al. Controlled ultrasound erosion for transdermal delivery and hepatitis B immunization
JP6334214B2 (en) Drug release method using laser
Zharov et al. Laser combined medical technologies from Russia
Esenaliev Interaction of radiation with microparticles for enhancement of drug delivery in tumors
WO2013109722A1 (en) Theranostic methods and systems for diagnosis and treatment of malaria
Chen et al. Advances in cancer photothermal therapy
KR20160119077A (en) Liquid vehicle for suspension of undelivered particles

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
REF Corresponds to

Ref document number: 10081863

Country of ref document: DE

Date of ref document: 20020627

Format of ref document f/p: P