NO342271B1 - A new drug delivery system for treatment of cancer - Google Patents

A new drug delivery system for treatment of cancer Download PDF

Info

Publication number
NO342271B1
NO342271B1 NO20161568A NO20161568A NO342271B1 NO 342271 B1 NO342271 B1 NO 342271B1 NO 20161568 A NO20161568 A NO 20161568A NO 20161568 A NO20161568 A NO 20161568A NO 342271 B1 NO342271 B1 NO 342271B1
Authority
NO
Norway
Prior art keywords
nanoparticles
drug delivery
delivery system
nps
gas
Prior art date
Application number
NO20161568A
Other languages
Norwegian (no)
Other versions
NO20161568A1 (en
Inventor
Per Stenstad
Yrr Mørch
Heidi Johnsen
Rune Hansen
Ruth Schmid
Original Assignee
Sintef Tto As
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 Sintef Tto As filed Critical Sintef Tto As
Priority to NO20161568A priority Critical patent/NO342271B1/en
Priority to JP2019517040A priority patent/JP2019529488A/en
Priority to US16/338,152 priority patent/US20200023073A1/en
Priority to PCT/EP2017/074798 priority patent/WO2018060437A1/en
Priority to EP17784222.6A priority patent/EP3518900A1/en
Priority to CA3040016A priority patent/CA3040016A1/en
Priority to CN201780073657.2A priority patent/CN110022860A/en
Publication of NO20161568A1 publication Critical patent/NO20161568A1/en
Publication of NO342271B1 publication Critical patent/NO342271B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention is generally directed to improvements in the treatment of cancer and cancerous tumors. A new drug delivery system is provided, method for producing it and medical uses.The present invention is generally directed to improvements in the treatment of cancer and cancerous tumors. A new drug delivery system is provided, method for producing it and medical uses.

Description

A new drug delivery system for treatment of cancer.
TECHNICAL FIELD OF THE INVENTION
The present invention is generally directed to improvements in the treatment of cancer and cancerous tumors. A new drug delivery system is provided, method for producing it and medical uses.
BACKGROUND OF THE INVENTION
Cancer is a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body. This malignant behavior often causes invasion and metastasis to second locations. Cancer is a major cause of mortality in most industrialized countries. The standard treatments include surgery, chemotherapy, radiation, laser and photodynamic therapy, alone or in combination. In addition, immunotherapy and hormonotherapy have been approved for certain types of cancer. Surgical intervention is used to remove macroscopic tumors and irradiation of the tumor site to treat the remaining microscopic tumors.
Chemotherapy is used to attack any residual or non-resectable disease, at either the surgical site or elsewhere in the body. The success rates of the different treatments are depending on the type and stage of the cancer. Although improved in recent years, the prognosis for many types of cancer patients is still poor.
Chemotherapy can be defined as the treatment of cancer with one or more cytotoxic anti-neoplastic drugs (chemotherapeutic agents) as part of a standardized regimen. The term encompasses a variety of drugs, which are divided into broad categories such as alkylating agents and antimetabolites. Traditional chemotherapeutic agents act by killing cells that divide rapidly, a critical property of most cancer cells. This is achieved by impairing mitosis (cell division) or DNA synthesis.
All though chemotherapy is curative for some cancers (such as for example leukemia), it is still ineffective in some and needless in others.
Chemotherapeutic agents are most often delivered parenterally, depending on the drug and the type of cancer to be treated. With traditional parenteral chemotherapy typically only 0.001-0.01% of the injected dose reaches the tumor. Many current chemotherapy drugs unfortunately also have excessive toxicity to healthy tissues and a limited ability to prevent metastases.
Enormous efforts have been put in finding novel tumor-targeting treatments in recent years. Tumors vasculature is generally more ‘leaky’ but suffers from higher interstitial fluid and oncotic pressure that can impede passage of drug throughout the tumor bulk. Uptake of established chemotherapeutics can be highly variable depending on tumor type and such uptake differences may contribute to the variable nature of the therapeutic effect.
Nanoparticles (NPs) as carriers for anti-cancer drugs offer great potential for such targeted cancer therapy as a certain accumulation in the tumor is observed due to the enhanced permeability and retention effect (EPR effect). Still, the uptake of NPs in tumors is relatively low and the distribution heterogeneous. Thus the nanomedicine field has so far shown limited impact. The indicated EPR effect, on which the nanomedicine field largely relies, has mainly been studied in animal tumor models and there is limited experimental data from patients. The EPR effect shows significant heterogeneity within and between tumor types and there is currently an ongoing debate within the oncological and nanomedicine communities regarding the EPR effect in humans. Novel treatment concepts, enhancing or bypassing the EPR effect are of high clinical interest.
It is known that gas-filled microbubbles (MBs), currently in clinical use as contrast agents for ultrasound (US) imaging, used in combination with therapeutic lowfrequency US can locally increase the vascular permeability, inducing an "artificial EPR effect" by loosening up or making pores through tight junctions for paracellular uptake, increased endocytosis and/or transcellular transport from sonoporation. However, commercially available MBs optimized for US imaging have very thin shells (2-20 nm), are fragile and have short blood circulation time (around 1 min). Their application in a drug delivery system to enhance uptake of chemotherapeutic agents to cancerous tissues and tumors is thus limited.
Accordingly, there is a need for an improved drug delivery system, which can increase the vascular permeability and enhance uptake of therapeutic agents in tumors.
Recent work has also been motivated to address the issues of drug delivery across the blood-brain barrier (BBB), and delivery to solid tumors. Tight vascular endothelial junctions that inhibits the passage of larger molecules to the tissue space characterize the blood brain barrier. Brain delivery of drugs is hindered by the BBB, an interface at brain endothelium that protects the brain and maintains its homeostasis, but also restricts the passage of 98% of small and virtually all large molecular drugs.
Nanoparticles (NPs) can offer numerous benefits in drug delivery due to their high drug loading capacity, incorporation of poorly soluble drugs and novel therapeutics such as peptides and oligonucleotides, functionalization for sustained and controlled release and combination of therapeutics with imaging. In the case of solid tumors, nanoparticles can also benefit from the enhanced permeability and retention effect, whereby NPs are retained in the tumor due to its leaky neovasculature and reduced lymphatic drainage. The BBB, however, is a formidable obstacle for NPs as well, and their brain delivery can benefit from versatile BBB opening techniques.
Thus, there is a need to explore the potential use of nanoparticles in drug delivery to the brain.
The most basic form of ultrasound/microbubble mediated drug delivery is administration of a microbubble formulation together with a systemically administered drug. An example of such an approach has recently entered clinical trials [Kotopoulis et al, Med Phys., 40(7) (2013)], where the commercial US contrast agent Sonovue (Bracco Spa.) is co-administered with Gemcitabine followed by US irradiation for treatment of pancreatic cancer.
In addition to the co-administration approach, several other microbubble technologies are explored for drug delivery [Geers et al, Journal of Controlled Release 164 (2012) 248-255]. Examples are drug-loaded microbubbles, in situ formed microbubbles from nanodroplets and targeted microbubbles. Over the years, however, it has been recognized that all these approaches have fundamental limitations, which have effectively hindered a transition to clinical practice. Perhaps the most limiting is the amount of drug that can be incorporated into microbubble systems. In addition, for attachment and/or incorporation of the drug load into the microbubble systems, chemical modification of the drug may be required, with potential changes to biological activity.
US2013183244 A1, US2014212502 A1, US2011200530 A1, WO2012/094541 A1, EP1701745 A1 describes different drug delivery systems comprising different nanoparticles and microbubbles or vesicles.
Liu, Zhe et al. (2011) discloses USPIO (ultra-small superparamagnetic iron oxide) nanoparticle-embedded PBCA (polybutylcyanoacrylate) microbubbles for MR imaging. Moon, Hyungwon, et al. (2015) describes a theranostic system comprising paclitaxel-loaded human serum albumin nanoparticles. Wang, Jiayu, et al (2016), De Cock, Ine, et al (2016), WO2014/191502, EP2508207, Chaudhari K. R., et al. (2012) and Zhang, Yu et al. (2008) describe different drug delivery systems comprising different types of nanoparticles associated, or not, in different ways with microbubbles. Hansen et al (2013), Mørch, Yrr et al (2015) and Baghirov et al (2016) disclose a delivery system comprising nanoparticle stabilized microbubbles.
However, none of the citations mentioned offers a flexible system of ultrasound triggered drug delivery system comprising gas-filled microbubbles with a plurality of nanoparticles associated to said microbubbles, where at least one therapeutic agent is associated with nanoparticles and at least one free nanoparticle with an associated therapeutic agent, nor demonstrate a surprising therapeutic effect of such a system.
Accordingly, there is a need for novel multifunctional drug delivery systems.
The invention is the first successful demonstration of a novel multifunctional drug delivery system comprising gas-filled microbubbles associated with nanoparticles in treatment of cancerous tumors. The delivery-system is used in combination with ultrasound to facilitate the delivery. Enhanced uptake of nanoparticles in tumors is achieved by applying an acoustic field, such as generated by focused ultrasound.
DEFINITIONS
The term ‘microbubble, (MB)’ is used herein to describe microbubbles with a diameter in the range from 0.5 to 30 microns, typically with a mean diameter between 1 to 6 μm.
The term ‘nanoparticle, (NP)’ is used herein to describe particles with linear dimensions less than 800 nm.
The terms “microbubble associated with nanoparticles” and “nanoparticles associated with microbubbles” are used herein to describe in what way the nanoparticles interact with the microbubble interface. The term “associated with” as used in connection with this include association by any type of chemical bonding, such as covalent bonding, non-covalent bonding, hydrogen bonding, ionic bonding or any other surface-surface interactions.
The term “free nanoparticles” describes nanoparticles that are non-associated with the microbubbles.
The term ‘surfactant’ is used in herein for chemical compounds that lower the surface tension between two liquids, or between a gas and a liquid, e.g. used as a stabiliser in a dispersion of microbubbles.
‘Acoustic field’ is the term used to describe the area where the focused ultrawawes are applied, hence the area of exposure or treatment. The acoustic field generates “thermal and non-thermal mechanisms”. “Non-thermal mechanisms” include cavitation, vibrations and oscillations.
“High intensity focused ultrasound, (HIFU)” or “focused ultrasound, (FUS)” refers to the medical technology that uses an acoustic lens to concentrate multiple intersecting beams of ultrasound on a target. Each individual beam passes through tissue with little effect but at the focal point where the beams converge, the energy can have useful thermal or mechanical effects. HIFU or FUS is typically performed with real-time imaging via ultrasound or MRI to enable treatment targeting and monitoring (including thermal tracking with MRI).
The term ‘cavitation’ is used to describe the process where MB expand and compress upon exposure to in the acoustic field, generated by a ultrasound.
Ultrasound waves propagate through high- and low-pressure cycles, and the pressure differences make the MBs expand during the low-pressure phase and compress during the high-pressure phase. This oscillation can be stable for several cycles (stable cavitation), but it can also end in more or less violent collapse of the MBs (inertial cavitation), depending on the pressure amplitude and frequency. Cavitation-related mechanisms include radiation force, microstreaming, shock waves, free radicals, microjets and strain.
The term “sonoporation”, or “cellular sonication”, is used herein to describe the use of sound (typically ultrasonic frequencies) for modifying the permeability of the cell plasma membrane. Sonoporation employs the acoustic cavitation of microbubbles, thus enhancing the delivery of nanoparticles to tumors. As used herein, the term “drug delivery” is understood to include the delivery of drug molecules, therapeutic agents, diagnostic agents, genes, and radioisotopes.
The term “pharmaceutical composition” used in this text has its conventional meaning, and in particular are in a form suitable for mammalian administration, especially via parenteral injection.
The term “therapeutic agent” is meant to include every active force or substance capable of producing a therapeutic effect. The terms “chemotherapeutic agent” and “anti-cancer drugs” are used interchangeably throughout the description.
The term “diagnostic agent” is used to described a substance used to reveal, pinpoint, and define the localization of a pathological process.
The expression “enhanced permeability and retention (EPR) effect” and 'artificial EPR effect' are used herein to describe the property by which molecules of certain sizes (typically liposomes, nanoparticles, and macromolecular drugs) tend to accumulate in tumor tissue much more than they do in normal tissue.
The term “blood-brain-barrier” as used herein refers to the highly selective permeability barrier that separates the circulating blood from the brain extracellular fluid in the central nervous system (CNS). The blood–brain barrier is formed by brain endothelial cells, which are connected by tight junctions with an extremely high electrical resistivity.
SUMMARY OF INVENTION
The present invention is generally directed to improvement in treatment of cancer and cancerous tumors.
It is described a nanoparticle filled (or loaded) with a therapeutic agent, a gas-filled microbubble, and the combination of the two. A drug delivery system is disclosed which facilitates the delivery of the therapeutic agent to tissue. The system uses ultrasound or acoustic radiation force to induce an acoustic field that covers the diseased area. In the acoustic field, cavitation and/or oscillation can occur. The cavitation or oscillation may cause a possible collapse of the microbubbles. The collapse of gas microbubbles releases the nanoparticles. In the acoustic field radiation forces will act on the microbubbles, and may push them towards the vessel wall before they collapse. Cavitation and collapse can further generate shear stress and jet streams on endothelial cells, which will both, together and independently, improve transport of nanoparticles across the capillary wall.
In a first aspect of the invention, it is disclosed a drug delivery system for use in therapy comprising at least one gas-filled microbubble, a plurality of nanoparticles associated with the microbubble and one or more therapeutic agent associated with one or more of said nanoparticles for ultrasound-mediated delivery of the nanoparticles and/or the therapeutic agent to a tumorous tissue, wherein the system further comprising at least one free nanoparticle and at least one therapeutic agent associated with the at least one free nanoparticle.
The drug delivery system according to the invention is multimodal and multifunctional, and constitutes a novel medical use for treatment of cancer, in particular solid tumors.
In certain embodiments, the nanoparticles may be surface-associated to the microbubble and covering at least a part of the microbubble surface, optionally the at least one of the nanoparticles are polymeric, such as poly(alkylcyanoacrylate) (PACA) nanoparticle. In one preferred embodiment, the PACA-particle is a poly(isohexylcyanoacrylate). According to one embodiment of the invention, the therapeutic agent is loaded within the nanoparticles. Optionally, the nanoparticles may also contain co-stabilizers.
Particularly, the drug delivery system according to the first aspect comprises free nanoparticles and one or more therapeutic agent associated with the free nanoparticle. In certain embodiments, the nanoparticles associated with the microbubble are the same kind of nanoparticles as the free nanoparticles, and both may be filled with at least one therapeutic agent.
According to another embodiment, the surface-associated polymeric nanoparticles stabilizes the microbubble. The stabilizing of microbubbles by the nanoparticles will influence the possible circulation time of the microbubbles in blood.
In certain embodiments, the drug delivery system according to the invention may further optionally comprise at least one or more targeting agents, a pharmaceutically acceptable carrier, and the nanoparticles may further be coated with a hydrophilic polymer such as polyethylene glycol (PEG).
In certain embodiments, the mean diameter of microbubble with surface-associated polymeric nanoparticles is in the range 0,5 to 30 μm.
The therapeutic agent is in certain embodiments a chemotherapeutic agent or a chemopotentiator.
According to further embodiments, the microbubble may be filled with a gas selected from the group consisting of: perfluorocarbon, air, N2, O2, CO2..
In one embodiment, the ultrasound-mediated delivery is mediated by ultrasound, such as focused ultrasound.
In another embodiment, the delivery is mediated by radiation force and/or heating, which can also lead to increased transport of nanoparticles and drugs in extracellular matrix in tumor tissue.
In a further embodiment, the delivery is mediated by a combination of ultrasound and radiation force and/or heating.
In another embodiment, the microbubble is destroyable upon application of focused ultrasound thereto.
The invention also includes a method for preparing a drug delivery system according to the invention, comprising the steps of:
a) Synthesizing the nanoparticles to be loaded with the therapeutic agent and/or contrast agent.
b) Adding nanoparticles to a solution comprising a surface-active substance. c) Mixing the solution with gas to obtain gas-filled bubbles.
According to one embodiment of the method, the microbubbles are stabilized by self-assembly of nanoparticles in the gas-water interface.
In certain embodiments of the method, the solution in c) is mixed with gas for a desired time, such as from about 2 seconds to 60 minutes, preferentially 1 to 10 minutes, and/or desired speed, such as about 500 to 50 000 rpm when ultraturrax mixing is used, preferentially 1 000 to 30 000 rpm to obtain microbubbles of desired size.
According to certain embodiments of the method, the surface-active substance in the solution in step b) is selected from the group consisting of a protein or a lipid or a polymer or a surfactant.
An aspect of the invention includes a gas-filled microbubble associated with nanoparticles for use in treatment of cancer, wherein at least one of the nanoparticles is loaded with a therapeutic agent and delivery of the nanoparticles to tumorous tissue is facilitated by an acoustic field generated by ultrasound.
The nanoparticles according to this aspect may be surface-associated to the microbubble and covering at least a part of the microbubble surface.
According to an embodiment, the surface-associated nanoparticles stabilizes the microbubble.
In one embodiment, the invention is a gas-filled microbubble associated with nanoparticles and free nanoparticles for use in treatment of cancer, wherein at least one of the nanoparticles is loaded with a therapeutic agent and delivery of the nanoparticles to tumorous tissue is facilitated by an acoustic field generated by ultrasound
According to certain embodiments, the acoustic field causes cavitation, i.e. oscillation and/or collapse of the gas-filled microbubbles. The cavitation may improve the transport of nanoparticles across the capillary wall. As such, this novel use enhances the EPR effect. The acoustic field is mediated by ultrasound or other methods known to the skilled person.
The surface-associated nanoparticles used according to this aspect may optionally comprise at least one or more targeting agents.
Another aspect of the invention is a composition for use in treatment of cancer comprising a gas-filled microbubble, a plurality of nanoparticles associated with the microbubble and one or more chemotherapeutic agent associated with one or more of the nanoparticle, wherein the composition further comprising at least one free nanoparticle and at least one therapeutic agent associated with the at least one free nanoparticle.
Particularly it is described that the composition further comprises free nanoparticles, i.e. nanoparticles that are non-associated with the microbubbles.
According to one embodiment of this aspect, the composition comprises a drug delivery system according to the first aspect of the invention.
A last aspect describes a method of treating cancer comprising administering a drug delivery system according to the first aspect of the invention to a patient in need thereof.
The therapeutic agent may be loaded within at least one nanoparticle, optionally, the system according to the first aspect of the invention may also comprise nanoparticles loaded with diagnostic agents.
Further, it is described a method for the treatment of cancer comprising delivering a microbubble with associated nanoparticles to a treatment site of a patient, wherein the at least one nanoparticle is filled with a therapeutic agent. The method includes applying ultrasound energy to the treatment site. The disease is cancer, such as breast cancer or cancer in the brain.
BRIEF DESCRIPTION OF DRAWINGS
Figure 1: Size distribution of PEGylated cabazitaxel-loaded PIHCA NPs as measured by dynamic light scattering. The drug loading is 10.7 wt%.
Figure 2: Histogram showing size distribution of MBs stabilized by PEGylated cabazitaxel-loaded PIHCA NPs as measured by light microcsopy and image analysis.
Figure 3: Electron microscopy image of microbubble with surface-associated nanoparticles
Figure 4. The size and zetapotential of the NPs were approximately 170 nm and -1 mV, respectively. Cellular uptake in the breast cancer cell line was confirmed by CLSM (A). The NPs were imaged by encapsulating a fluorescent dye (red). From quantification by FCM, 90% of the cells had taken up NPs by endocytosis after 3 h incubation (B).
Figure 5. In vivo circulation half-life of the PEGylated NPs was found to be 136 minutes (n=5 animals) (A). An exponential decay on the form of 206160.9e<-0.0051x>fitted the data with R<2>=0.67 and p-values ≤0.0001. The MBs stabilized by the selfassembled NPs had a size of approximately 3 µm, and were found to be suitable for in vivo contrast enhanced US imaging and image guided drug delivery. Contrast enhancement due to inflow and circulation of bubbles in a tumor imaged by ultrasound(B).
Figure 6: The biodistribution of NPs 6 h post injection. An example of organs and tumor from one animal is shown (A). Quantification of accumulation in organs and tumors is shown as mean and standard deviation (n=10 animals, n=5 for brain) (B). Autofluorescence from non-treated organs and tumor is shown from one animal.
Figure 7: 87% of the dose can be found in these organs, tumor and brain. The rest is likely found in urine, stool, skin, muscle and other tissues. The majority of the dose is located in the liver and spleen, and about 1% of the dose is located in the tumor (Corresponds well with the reported 0.7% median)
Figure 8: An example of a CLSM tile scan from an entire tumor section, showing NPs in red (A). The number of pixels with fluorescence from NPs was quantified in tile scans from each animal (B). Similar results were seen when pixel intensities were measured. No effect of stable cavitation was found, whereas the violent collapse of MBs increased the delivery of NPs to tumors, and the uptake increased with increasing MI.
Figure 9: Analysis of sections, uptake of PIHCA NPs.
Figure 10: Except for the highest MI (G7), which caused substantial visual hemorrhage, evaluation of HES stained tumor sections showed that all FUS treatments were considered safe. Example of an overview image (A), and representative images of non-treated and treated tissue are shown (B and C, respectively).
Figure 11: The microdistribution of NPs in the tumors 2 h post treatment was imaged using CLSM. Representative examples from the control group that did not receive any ultrasound treatment (A) and a group that was treated with high pressure (B). Blood vessels are shown in green and nanoparticles in red. An increased delivery of NPs is observed in the treated group (G6) compared to the control group. Distribution of fluorescent dye in tumors with (b) and without (a) applying ultrasound. 250 times more drugs in b) than in a).
Figure 12: Probing the intracellular degradation of poly (alkyl cyanoacrylate) nanoparticles using confocal microscopy. Measuring the drug release intracellularly
Figure 13: Uptake of nanoparticles in cells, in vitro.
Figure 14: Viability of MDA-MB-231 cells (human epithelial, mammary Figure 15: Uptake of MRI contrast agent in brain. This specific agent will normally not pass the BBB. Thus, the results illustrate transient BBB opening.
Figure 16: FUS-mediated BBB disruption and transport of NPs across the BBB. a) BBB opening mediated by FUS in combination with the PIHCA-MB platform. b) transport of PIHCA NPs across the BBB following FUS exposure. Red – PIHCA NPs, Green – blood vessels.
Figure 17: Weight of the animals as a function of time is shown as average and standard deviation for the three different treatment groups. n=4 animals pr group. Day 0 is the day of implantation of tumor cells. Treatments were done at day 21 and 29.
Figure 18: Tumor volume as a function of time is shown as average and standard deviation for the three different groups. Group 1: Control, saline. Group 2:
Microbubbles associated with nanoparticles and the cytostatic drug (cabazitaxel). Group 3: Ultrasound and microbubbles associasted with nanoparticles and the cytostatic drug. n=4 animals pr group. Day 0 is the day of implantation of tumor cells. Treatments were done at day 21 and 29
Figure 19: Tumor volume at day 35 after tumor cell implantation for the three different treatment groups, n=4 animals pr group. Mean and standard deviation is shown
DETAILED DESCRIPTION
The present invention is an innovative drug delivery system allowing for controlled and enhanced delivery of anticancer agents to tumors with the aid of focused US. The drug delivery system according to the invention comprises gas-filled microbubbles associated with nanoparticles, wherein at least one of the nanoparticles is loaded with a therapeutic agent and delivery of the nanoparticles to tumors is facilitated by an acoustic field generated by ultrasound wherein the system further comprising at least one free nanoparticle and at least one therapeutic agent associated with the at least one free nanoparticle. The gas-filled MBs associated with NPs loaded with at least one therapeutic agent may be used in treatment of cancer. In particular, the MBs associated with NPs, according to the invention, are for use in treatment of solid tumors, including tumors in the brain.
In one embodiment, the gas-filled MBs is stabilized by NPs. The NPs stabilize the gas/water interfaces by self-assembly at the MB surface, thus resulting in very stable MBs. One advantage of the nanoparticle-stabilized MBs according to one embodiment of the invention is thus increased shelf-life.
The delivery of nanoparticles and the therapeutic agent to tumor tissue and/or cancer cells are enhanced by applying ultrasound or an acoustic radiation force. The ultrasound or acoustic radiation force induce an acoustic field that covers the diseased area. With ultrasound applied locally at the tumor, small pores in the blood vessel will transiently be formed. The acoustic field generated by ultrasound will cause the bubbles to oscillite and collapse, leading to release of individual NPs. The ultrasound also causes sonoporation, which enhances the vascular permeability.
Drug-loaded NPs may then accumulate in tumor tissue thanks to the enhanced vascular permeability.
In one embodiment of the invention, the delivery system further comprises free nanoparticles, i.e. nanoparticles that are non-associated with the microbubbles, and at least one therapeutic agent associated with the free nanoparticles.
Without being bound by theory, the advantages of this embodiment of the invention is a result of several mechanisms:
- microbubbles in combination with ultrasound create an artificial EPR effect transiently increasing the permeability of blood vessel walls. This enhances the accumulation of freely circulating NPs, i.e. the free NPs that are loaded with at least one therapeutic agent.
- nanoparticles associated with microbubbles will, upon bubble destruction by US, lead to high local deposit of NPs (and hence therapeutic agent), and deeper penetration into tumor tissue
As such, the drug delivery system according to this embodiment of the invention may deposit an even higher concentration of therapeutic agent than MBs associated with NPs alone.
The general principle is that the present invention utilizes nanoparticles (NPs) to deliver drugs. The nanoparticles are typically too large to penetrate healthy blood vessels, but small enough to extravasate the (tumor) blood vessels via the enhanced permeability and retention (EPR) effect or via ultrasound-induced "artificial EPR effect". NPs according to the invention may be loaded with therapeutic agents, such as anti-cancer agents, and/or diagnostic agents such as contrast agents. In one embodiment, the NPs are biodegradable. Contrast agents can optionally be further incorporated into the NPs for monitoring and follow-up of the NPs. Optionally, the nanoparticles may optionally contain co-stabilizers.
The NPs may typically be of a size from about 1-800 nm, such as about 10-500, preferably about 70-150 nm. The NPs may further be coated with a hydrophilic polymer such as polyethylene glycol (PEG) to avoid recognition by immune cells. Coating with PEG may further increase blood circulation time. Molecules targeting specific cells may optionally be attached to the NP surface in order to increase the local deposit of NPs at the disease site. The NPs according to the invention is designed for encapsulation of anti-cancer agents. Further, they may successfully be used for producing stabile MBs as described herein. In certain embodiments, the NPs are polymer-based NP, composed of the widely used biocompatible and biodegradable poly(alkyl cyanoacrylate) (PACA) polymer. As demonstrated herein, the NP according to the invention is especially well suited for BBB penetration. In one particular embodiment, the drug-loaded biodegradable NPs is a polymer-based nanoparticle as described in WO 2014/191502.
Nanotechnology has started a new era in engineering multifunctional NPs to improve diagnosis and therapy of various diseases, incorporating both contrast agents for imaging and drugs for therapy into so called theranostic NPs. In cancer therapy, encapsulating the drugs into NPs, such as described herein, will improve the pharmacokinetics and reduces the systemic exposure due to the leaky capillaries in tumours. The NPs according to the invention have also been shown to have a potential of treating diseases in the central nervous system (CNS) as they can pass through the BBB. The access of molecules to the CNS is strictly controlled by the specialized and tight junction between the endothelial cells forming the blood vessels constituting the BBB.
In one embodiment, the nanoparticles comprised in the system of the invention is a poly(alkyl cyanoacrylate) (PACA) NP. PACA NPs have shown promise as drug carriers both to solid tumors and across the BBB. This is partly due to the flexibility of the system allowing surface functionalization and drug encapsulation in one step. Moreover, the degradation and drug release from these nanoparticles (NPs) can be tuned by choosing different monomers. In one embodiment, the NP is prepared by the method as described in WO 2014/191502.
As described herein, the nanoparticles are used in association with MBs. In certain embodiments, the NPs may stabilize the MBs by self-assembly at the MB gas/liquid interface thus forming a stabilizing shell around the MBs. The result is a very stabile microbubble with improved technical features. In certain embodiments, the MBs are produced by addition of a further stabilizing agent, such as a surface-active agent. The stabilizing agent may be a surface-active agent chosen from the group of serum, proteins, polymers, lipids or surfactants. The MBs may be produced mixing the solution comprising nanoparticles with a gas by using ultra-turrax, shaking, ultrasound, or other means known to the skilled person. In certain embodiments, the NPs will self-assemble in the gas/liquid interface and form a stabilizing shell around the MBs. In certain embodiments, the nanoparticle-stabilized MBs reduce the fragility of the MBs e compared to commercially available MBs.
In order to improve the uptake and distribution of NPs into diseased tissue, the administration of NPs according to the invention is combined with a treatment facilitating the delivery, such as by applying ultrasound to establish an acoustic field. Without being bound by theory, the hypothesis is that ultrasound is able to improve drug delivery by different mechanisms. In an acoustic field, cavitation, which is the oscillation and possible collapse of gas microbubbles, can occur.
Cavitation can then generate shear stresses and jet streams on endothelial cells thereby improving the transport of NPs across the capillary wall. In certain embodiments, the improved extravasation and distribution of NPs in tumours may be achieved by a non-thermal mechanism, however heating and radiation forces may also further enhance the delivery.
In certain embodiments, the present system comprises three elements:
1. NPs containing the therapeutic agents and contrast agents, alone or in combination.
2. Gas-filled MBs stabilized by the drug-loaded NPs
3. Ultrasound technology for ultrasound-mediated drug delivery using the NP-stabilized MBs
This novel multimodal, multifunctional drug delivery system according to the invention have been shown to improve delivery of therapeutic agents to cancer cells by ultrasound-mediated delivery of NPs. Combining these NP-associated MBs with focused ultrasound results in a higher uptake and improved distribution of the NPs in tumors growing, thus resulting in an improved treatment of cancer. As demonstrated in Example 4 and figure 14, the invention results in reduced tumor growth compared to control.
The new NP-associated MBs can also be used to penetrate the BBB, as documented by magnetic resonance imaging and localization of fluorescently labelled NPs in brain tissue (se figure 16 and 17). Thus, the new NP-associated MB platform demonstrates promising clinical potential in treatment of brain cancer.
Ultrasound and MBs can improve the delivery of non-encapsulated drugs, as recently demonstrated in a clinical study combining ultrasound and co-injection of gemcitabine and commercially available MBs to treat pancreatic cancer. The combination of ultrasound and MBs can also facilitates a transient and local opening of the blood-brain barrier, thereby permitting various drugs to enter the brain and thus treat central nervous system (CNS) disorders. The exact mechanism by which ultrasound and MBs causes blood-brain barrier disruptions is not fully understood, but it is speculated that cavitation i.e.; volume oscillations of MBs in an ultrasound field, might be an important factor.
According to one embodiment of the invention, a mixture of individual drug-loaded NPs and NPs associated with MBs , are injected into the blood stream and will quickly be distributed throughout the entire circulation system. These MBs and free NPs are too large to cross the blood vessel wall of healthy tissue. When entering the acoustic field, applied locally at the tumor site, the MBs will undergo large volume oscillations. During this process, the vascular permeability will be transiently increased due to mechanical stimuli from the oscillating MBs forming small pores in the blood vessel wall. US focused to the disease site will also induce bubble collapse, releasing individual NPs from the MB shell for highly targeted treatment. Upon MB destruction, a very high local concentration of drug-loaded NPs is thus obtained.
The acoustic activity of NP-associated MBs is demonstrated both in vitro and in vivo. As such, they have a great potential in therapeutic applications. It is further shown that US can destroy the MBs, as described herein, thus releasing individual NPs and enhancing model-drug uptake in tumor-bearing mice. Further, it is shown that the BBB in rats maybe safely and transiently opened using the novel MBs together with NPs and US. Finally, the effect of MBs associated with NPs is demonstrated in cancer treatment, by the in vivo study described in example 3. The study demonstrates for the first time the applicability of the described drug delivery system in cancer treatment, as the result demonstrate the ability to significantly reduce tumor growth compared to control.
There is a clear need for novel drug-delivery system comprising MBs and NPs with a high drug payload, specifically designed for US-mediated drug delivery applications. Currently, there are no such products on the market. The system according to the invention fills the void and is thus relevant for tumors that are not effectively treated using existing chemotherapeutic technology.
The uniqueness of the invention is its simplicity and versatility, still leading to highly suitable acoustic and biological properties for US-mediated cancer therapy. The advantages of the invention compared to the research systems described today are:
● The invention offers a multifunctionality in one simple formulation, which constitute an innovative and advantageous drug delivery system for clinical applications.
● The invention can be used separately or simultaneously for US-imaging, diagnosis and therapy
● The invention has circulation times significantly longer than commercial MBs.
● The invention comprises a combination of individual free NPs and NP-associated MBs, hence allowing for the targeted delivery of very high drug concentrations to tumor tissue
● The invention integrates NPs incorporating high payloads of drugs and MBs into one single unit (NP-associated MBs). Integrating NPs and MBs into a single unit is found to have the potential to be much more efficient in US-enhanced tumor uptake as compared to co-injection of NPs and MBs. This is probably caused by a higher concentration of NPs locally in the region of sonication where the MBs are destroyed, in contrast to when NPs and MBs are co-injected intravenously and the NPs are diluted systemically in the blood stream.
● The MBs are prepared in a one-step process by self-assembly of NPs at the gas/liquid interphase.
● The NPs are also prepared in a one-step process and without the use of organic solvents. This offers a simple, cost-efficient and easy translation to the clinic and into profitable products.
The MBs are associated with thousands of single drug-loaded NPs, as opposed to MBs currently on the market, which are composed of a solid shell of lipids, proteins or polymers. This offers a flexible, yet tough and stable shell, and the ability to release the individual NPs small enough to reach the tumor target.
The novel drug delivery system according to the invention clearly addresses the need for novel treatment concepts for enhanced delivery of anti-cancer agents. Further, the invention has the potential to improve treatment of solid tumors significantly. Given the typically poor responses seen with small molecules in solid tumors and the low clinical success up to now with nano-drugs based on the EPR effect, the invention may have a major social impact. Lives may be saved and after-costs of acute and remedial therapy can potentially be greatly reduced. Enhanced drug penetration induced by the invention may affect the necessity of debilitating surgeries. In different embodiments, the invention may particularly be used within a few specific areas of high clinical relevance:
- Patients with inoperable cancer
- Patients with primary tumors or metastases in the brain. Here there is a strong need for novel delivery techniques, as most anti-cancer drugs will not reach the tumor due to the tight junctions of the BBB.
According to one embodiment, the MBs can be used for contrast enhanced US imaging. The NPs can contain drugs as well as contrast agents, and may be optionally further functionalized with targeting ligands. The NPs may further be coated with a hydrophilic polymer, such as polyethylene glycol (PEG), to improve their circulation time and biodistribution. Accordingly, the invention discloses a highly versatile system.
The chemotherapeutic agent comprised in the nanoparticles may be selected from the group, but are not limited to, the drug classes: Alkylating agents, antimetabolites, cytotoxic antibiotics, topoisomerase inhibitors, anti-microtubule agents or any other known chemotherapeutic agents known to the skilled person.
The cancer treated with the nanoparticles may be solid tumors or cancerous cells. In a particularly preferred embodiment, the cancer is a breast cancer.
The invention is illustrated by the following non-limiting examples.
EXAMPLES
Example 1
Production of drug-loaded PACA NPs and NP-stabilized microbubbles
Materials and methods:
Synthesis and physico-chemical characterization of drug loaded PACA NPs:
PEG-coated and cabazitaxel-loaded PIHCA NPs were prepared by the miniemulsion method as follows: An oil phase containing 1.50 g of isohexyl cyanoacrylate (monomer), 0.03 g of Miglyol 812 (co-stabilizer, inactive oil) and 0.18 g cabazitaxel (cytotoxic drug) was prepared by thorough mixing in a glass vial. An aqueous phase containing 0.09 g of Brij L23 (23 PEG units, MW 1225) and 0.09 g of Kolliphor HS15 (15 PEG units, MW 960), dissolved in 12 ml of 0.1 M HCl was prepared. An oil-in-water emulsion was prepared by mixing the oil and aqueous phase and immediately sonicating the mixture (Branson digital sonifier 450) on ice for 2 minutes (4x30 sec intervals, 60% amplitude) followed by another 3 minutes (6x30 sec intervals, 30% amplitude). After sonication the solution was rotated at 15 rpm overnight at room temperature before adjusting the pH to 5 using 0.1M NaOH. The polymerization was continued for 5 hours at room temperature while rotated (15 rpm). The dispersion was dialyzed extensively against 1mM HCl (pH 3) at room temperature to remove unreacted PEG (dialysis membrane, MWCO 100,000 Da). The dialysate was replaced 3 times. The particles were stored in the acidic solution at 4°C. The above mentioned method resulted in PEGylated, drug-loaded and nontargeted NP dispersions with concentrations of 75 mg NP/ml after dialysis. When stored in acidic condition, the particle dispersion was stable for several months, with no aggregation observed.
Zetasizer (Dynamic light scattering) was used in order to determine hydrodynamic size, size distribution and surface charge of the PACA nanoparticles. To calculate the amount of encapsulated drug, drug content was extracted from the particles and the extracted amount of cabazitaxel was quantified by using LC-MS/ MS method.
Production and characterization of NP-stabilized MBs:
Gas-filled MBs associated with PACA NPs were produced as follows: A solution containing 2wt% casein (pH 7) was prepared and filtered through 0.22µm syringe filter. The cabazitaxel-loaded PEGylated PIHCA NPs described above were mixed with the casein solution and distilled water to a final concentration of 0.5 wt% casein and 1 wt% NP, with a total volume of 4 ml. The mixture was placed in a sonication batch for 10 minutes (at ambient temperatures) before the solution was saturated with perfluoropropane gas (approximately 10 seconds) and the vial partly sealed with parafilm. Ultraturrax (25,000 rpm) was then immediately applied for 2 minutes to produce perfluoropropane-filled NP-stabilized MBs. The vial was immediately sealed under perfluoropropane atmosphere using septum.
The size and concentration of the resulting NP-stabilized MBs was determined from light microscopy images using a 20× phase contrast objective and cell counter. MBs were counted and the size was calculated by analyzing the images.
Results:
The above mentioned method resulted in PEGylated, drug-loaded and non-targeted NP dispersions with concentrations of 75 mg NP/ml after dialysis. When stored in acidic condition, the particle dispersion was stable for several months, with no aggregation observed.
Dynamic light scattering method showed an NP size of 142 nm (z-average) with a polydispersity index of 0.18 (see Figure 1). The measured zetapotential was -1 mV. The determined drug loading efficiency was 72% and the drug payload was 10.7% (% wt cabazitaxel/wt NP).
The resulting NP-stabilized MBs had an average size of 2.3 µm (see Figure 2) and concentration of 5.62E+08 MBs/ml as measured by light microscopy and image analysis. Fluorescence microscopy (using same type of NPs only encapsulating a fluorescent dye instead of drug) and electron microscopy (Fig 3) was used to confirm that NPs are associated with the MBs forming a stabilizing (mono)layer. When stored at 4°C, the microbubbles were stable for up to several months.
Example 2
Cellular uptake of fluorescent dye ("model drug") encapsulated in nanoparticles (PIHCA) in breast cancer cells.
The aim of this study was to investigate the mechanisms of ultrasound-mediated delivery, to determine whether stable or inertial cavitation is the major mechanism for improved extravasation and enhanced NP delivery. To achieve successful delivery, the NPs have to circulate in blood for sufficient amount of time, extravasate from the vasculature, penetrate the extracellular matrix and deliver their payload to the intracellular targets.
Size and zetapotential of the biocompatible and biodegradable poly(isohexyl cyanoacrylate) NPs were determined by Zetasizer. In vitro cellular uptake was studied in breast cancer cells (MDA-MB-231) using confocal laser scanning microscopy (CLSM) and flow cytometry (FCM) by encapsulating a fluorescent dye.
Figure 4 shows that the size and zetapotential of the NPs were approximately 170 nm and -1 mV, respectively. Cellular uptake in the breast cancer cell line was confirmed by CLSM (see A). The NPs were imaged by encapsulating a fluorescent dye. From quantification by FCM, 90% of the cells had taken up NPs by endocytosis after 3 h incubation (B)..
In vivo circulation half life of NPs was determined by blood sampling from the saphenous vein in mice at 10 min, 30 min, and 1, 2, 4, 6, and 24 h post injection.
Figure 5 shows in vivo circulation half-life of the PEGylated NPs. It was found to be 136 minutes (n=5 animals) (A). An exponential decay on the form of 206160.9e<-0.0051x>fitted the data with R<2>=0.67 and p-values ≤0.0001. The MBs stabilized by the self-assembled NPs had a size of approximately 3 µm, and were found to be suitable for in vivo contrast enhanced US imaging and image guided drug delivery. Contrast enhancement due to inflow and circulation of bubbles in a tumor imaged by ultrasound (see Fig.5, B).
Perfluoropropane MBs were made by vigorous stirring and self-assembly of the NPs at the gas-water interface. Inflow and circulation of microbubbles in tumors was imaged by ultrasound at 18MHz.
Biodistribution of NPs encapsulating a near infrared dye was imaged 6 h post injection.
The biodistribution of NPs was determined by imaging using a near infrared whole animal scanner, and by ex vivo quantification of accumulation in excised organs and tumors. This is presented in figure 6 and 7.
Figure 6 shows the biodistribution of NPs 6 h post injection. An example of organs and tumor from one animal is shown (A). Quantification of accumulation in organs and tumors is shown as mean and standard deviation (n=10 animals, n=5 for brain) (B). Autofluorescence from non-treated organs and tumor is shown from one animal.
Figure 7 shows that 87% of the dose can be found in these organs, tumor and brain. The rest is likely found in urine, stool, skin, muscle and other tissues. The majority of the dose is located in the liver and spleen, and about 1% of the dose is located in the tumor (Corresponds well with the reported 0.7% median)
To study how stable versus inertial cavitation of MBs affected NP uptake in tumor tissue, subcutaneous breast cancer xenografts (MDA-MB-231) were grown in athymic mice. When tumors reached 7-8 mm length, MBs stabilized by NPs were injected intravenously before the tumors were treated with one of six different FUS treatments, using a 1 MHz FUS transducer and MIs ranging from 0.1 to 1. Blood vessels were stained by injecting FITC-labeled tomato lectin. The microdistribution of NPs was imaged by CLSM on frozen tumor sections. The experimental setup and the different treatment groups are indicated below:
• G1: Control group, no ultrasound.
• G2: 0.5 sec treatment, 1.5 sek break, (global PRF=0.5 Hz), 10.000 cycles (10ms) every 100 ms, (local PRF=10Hz), total duty cycle 2.5%, MI 0.1 (A).
• G3: As G2 with 3 additional cycles flash of MI 1 after each treatment (B).
• G4: As G3, but only the flash of MI 1 (C) .
• G5: As G2 but with an MI 0.25.
• G6: As G2 but with an MI 0.5 (D).
• G7: As G2 but with an MI 1.
Results
Results are presented in figure 8, 910 and 11.
Figure 8 demonstrate an example of a CLSM tile scan from an entire tumor section, showing NPs in red (A). The number of pixels with fluorescence from NPs was quantified in tile scans from each animal (B). Similar results were seen when pixel intensities were measured. No effect of stable cavitation was found, whereas the violent collapse of MBs increased the delivery of NPs to tumors, and the uptake increased with increasing MI.
Figure 9 shows analysis of sections and uptake of PIHCA NPs. The results of G1 is compared to G6.
Normalized to mean of G1 (control group):
● Group 1 (n=6 sections from 3 animals) CTRL
● Group 6 (n=6 sections from 3 animals) MI 0.5.
The mean of group 6 is at 2.5
Hematoxylin erythrosine saffron (HES) stained sections were imaged to evaluate safety of the treatment. Figure 10 shows the evaluation of the safety analysis.
Except for the highest MI (G7), which caused substantial visual hemorrhage was analyzed. The evaluation of HES stained tumor sections showed that all FUS treatments were considered safe. Example of an overview image (A), and representative images of non-treated and treated tissue are shown (B and C, respectively).
The micro distribution of NPs was imaged on frozen tumor sections using confocal laser scanning microscopy. This is presented in figure 11, which shows the microdistribution of NPs in the tumors 2 h post treatment as imaged using CLSM. Representative examples from the control group that did not receive any ultrasound treatment (A) and a group that was treated with high pressure (B). Blood vessels are shown in green and nanoparticles in red. An increased delivery of NPs is observed in the treated group (G6) compared to the control group. Distribution of fluorescent dye in tumors with (b) and without (a) applying ultrasound. The image show approximately 250 times more drugs in b) with the use of ultrasound than in a) without ultrasound.
Conclusion
High pressure sonication and thus violent collapse of MBs was found to improve the delivery of NPs to tumors, and increasing uptake was observed with increasing MI. However, hemorrhage was observed at the highest MI used, indicating that high MI in combination with MBs should be used with caution for drug delivery purposes.
The results show that this NP-MB platform is highly useful for controlled drug delivery.
Example 3
Uptake of drug in cells and cytotoxicity of empty and drug-loaded PACA NPs
Measuring the drug release intracellularly is necessary in order to understand the effect on cancer cells after internalization. The inventors used the model drug NR668 (modified Nile Red) encapsulated in poly (butyl cyanoacrylate) (PBCA) and poly (octyl cyanoacrylate) (POCA) to demonstrate that the NPs have different drug release kinetics also after internalization. While ordinary fluorescence imaging gives little information about the degradation, Fluorescence lifetime imaging (FLIM) (as shown in figure 12), Förster resonance energy transfer (FRET), emission specter analysis and time-laps imaging after cell lysis provids valuable information.
Figure 13 demonstrate the cellular uptake of NPs in breast cancer cells.
The cytotoxic effect of empty PBCA NPs, PBCA NPs with encapsulated cabazitaxel as well as free cabazitaxel was studied on breast cancer cells (MDA-MB-231 cells = human epithelial, mammary adenocarcinoma cell line). AlamarBlueR Cell Viability Assay was used to evaluate cell viability. Cells were seeded in density 5000 cells/ 200μl medium for each well. After 3 days old medium was removed from wells and both encapsulated cabazitaxel and free cabazitaxel was diluted in medium and added to the well. Concentration of NPs was ranged from 0,1 ng/ml to 1000 ng/ml. Concentrations of free cabazitaxel was chosen to match the concentrations of cabazitaxel in NPs. Control wells contained cells in growth medium. The particle size was approximately 125 nm for empty NPs and approximately 160 nm for both drug-loaded NPs.
The well plates were incubated for 24, 48 and 72 hours at 37°C and 5% CO2, before the medium was removed from the well followed by 3 times washing with fresh growth medium. Growth medium containing 10% of alamar Blue assay was added into each well and the plates incubated for another 3 hours at 37°C and 5% CO2, and the fluorescence intensity measured by microplate reader (excitation/emission at 550/590 nm).
Results:
The MDA-MB-231 cells responded to treatment with encapsulated cabazitaxel in PBCA and free cabazitaxel at various concentrations in a dose- responsive manner (Fig. 14). The cytotoxic effect of encapsulated cabazitaxel was similar to free cabazitaxel, demonstrating the successful release of drug from the particles.
Similar effects were seen with other PACA NPs (PIHCA and POCA) and with other cell lines (P3 glioma and HeLa cells).
Example 4
FUS-mediated BBB opening
Methods
For FUS-mediated BBB opening, the inventors used a state-of-the-art ultrasound system able to generate FUS at 1.1 MHz and 7.8 MHz during the same experiment, allowing a very precise magnetic resonance imaging (MRI)-guided selection of the area exposed to FUS. FUS exposure at the lower frequency was used to disrupt the BBB. FUS at the higher frequency of 7.8 MHz was employed to enable the effect of the acoustic radiation force. This force is caused by a transfer of momentum between the ultrasound wave and the propagation tissue, and the hypothesis is that it can facilitate NP transport in the extracellular matrix. Experiments were performed on immunodeficient mice with melanoma brain metastases developed four weeks after intracardiac injection of patient-derived human melanoma cells. A NP-MB platform, based on PIHCA NPs forming a shell around perfluorocarbon MBs, was used for FUS-mediated BBB opening. PIHCA NP-MBs were injected immediately before the FUS exposure. BBB opening was assessed using a gadolinium-based contrast agent. After the experiments, the brains were either frozen or fixed in formalin. NP transport across the BBB and distribution in the brain tissue were assessed in cryosections using confocal microscopy (see figure 17) , while histopathological changes and cellular changes caused by FUS were evaluated using formalin-fixed paraffin embedded tissue sections.
Results and Conclusions
Figure 15 shows uptake of the MRI contrast agentdye in brain. This specific agent will normally not pass the BBB. Thus, the results illustrate transient BBB opening
Figure 16 demonstrate FUS-mediated BBB disruption and transport of NPs across the BBB. In a) one can see BBB opening mediated by FUS in combination with the PIHCA-MB platform. In b), transport of PIHCA NPs across the BBB following FUS exposure. Red – PIHCA NPs, Green – blood vessels
Successful BBB opening was verified by MRI (as shown in figure 15). An optimal window for FUS-mediated BBB disruption using our NP-MB platform was found to be around a mechanical index of 0,31. Analysis of cryosections showed that the combination of FUS with our NP-MB platform allowed transport of NPs across the BBB in an ÷opening-dependent manner. Histological evaluation showed some extent of red blood cell extravasation following FUS exposure. The effect of the acoustic radiation force of NP distribution in the brain parenchyma away from blood vessels and the effect of FUS exposure on P-glycoprotein, an efflux transporter that is an integral part of the BBB, are currently being analyzed. Overall, our results indicate that our platform based on PIHCA NPs and MBs can be used to deliver substantial amount of NPs across the BBB, showing its potential in NP-aided drug delivery to the brain.
Example 5
In vivo demonstration of therapeutic effects
In vivo studies of effect of ultrasound-mediated drug delivery of MBs associated with NP loaded with anti-cancer drug in treatment of tumors.
The aim of the study was to investigate the described drug delivery systems ability to treat cancer, i.e. stop abnormal cell growth and shrinkage of tumors, in an in vivo model. The cancer cell used to demonstrate the potential of the invention was breast cancer cells, and the therapeutic agent was cabazitaxel.
• MDA-MB-231 breast cancer cells implanted subcutaneously on nude mice on day 0
• Tumors were allowed to grow until they reached a diameter of 4 mm in the longest direction (some just above and some just below 4)
• 4 animals were included in each group. Group 1: saline. Group 2:
microbubbles associated with NPs loaded with cabazitaxel. Group 3:
Microbubbles associated with NPs loaded with cabazitaxel and ultrasound.
• Injected volume was 200 ul intraveneously, total 2 mg nanoparticles per animal, and approximately 10mg/kg cabazitaxel
• Ultrasound treatment was optimized previously, and an MI of 0.5 was used.
(se ESCDD poster for detaljer om ultralyd-sekvens og oppsett)
• The mice were treated on day 21 and day 29
• Because the imasonic 1MHz transducer stopped working, the second ultrasound treatment had to be done with the FUS equipment. 16 spots (4x4) were scanned to cover the tumor area. The transducer had to be scanned because of the small focus. In each spot, 10000 cycles were given, and the 16 spots were scanned during 3.5 seconds. Total treatment time was increased from 2 minutes with the previous imasonic, to 3.5 minutes with the FUS equipment.
• Tumor growth is measured using calipers
The results of the study are presented in figure 17-19.
Conclusion
The study demonstrate enhanced delivery of therapeutic agent to tumors, and show for the first time therapeutic effect of the drug delivery system according to the invention.
The tumors in the control group (saline) grow at a certain rate, illustrated with the upper (=blue) curve in figure 18. Animals that are treated with microbubbles containing nanoparticles and the cytostatic drug (cabazitaxel) show reduced tumor growth (the curve in the middle= red curve). Animals which are treated with ultrasound in addition to microbubbles and nanoparticles filled with the cytostatic drug show that the tumor growth stops, the tumors shrink, and 2 out of 4 animals are cured at this time point (the lower curve = green curve). Figure 18 and 19 shows the effect achieved with the treatment. The weight of the animals was stable during and after the treatment for all three groups (see figure 17), proving that the treatment was well tolerated.
Figure 17: Weight of the animals as a function of time is shown as average and standard deviation for the three different treatment groups. n=4 animals pr group. Day 0 is the day of implantation of tumor cells. Treatments were done at day 21 and 29.
Figure 18: Tumor volume as a function of time is shown as average and standard deviation for the three different groups. Group 1: Control, saline. Group 2:
Microbubbles associated with nanoparticles and the cytostatic drug (cabazitaxel). Group 3: Ultrasound and microbubbles associasted with nanoparticles and the cytostatic drug. n=4 animals pr group. Day 0 is the day of implantation of tumor cells. Treatments were done at day 21 and 29
Figure 19: Tumor volume at day 35 after tumor cell implantation for the three different treatment groups, n=4 animals pr group. Mean and standard deviation is shown

Claims (24)

1. A drug delivery system for use in therapy comprising a gas-filled microbubble, a plurality of nanoparticles associated with the gas-filled microbubble and at least one therapeutic agent associated with at least one nanoparticle for ultrasound-mediated delivery of the nanoparticles and/or the at least one therapeutic agent to a tumorous tissue, wherein the system further comprising at least one free nanoparticle and at least one therapeutic agent associated with the at least one free nanoparticle.
2. The drug delivery system according to claim 1, wherein the nanoparticles associated with the gas-filled microbubbles are surface-associated to the gasfilled microbubble.
3. The drug delivery system according to any one of the claims 1-2, wherein the at least one therapeutic agent is loaded within the nanoparticles.
4. The drug delivery system according to any one of the claims 1-3, wherein the nanoparticles are polymeric.
5. The drug delivery system according to any one of the claims 1-4, wherein at least one of the nanoparticles is a poly(alkylcyanoacrylate) (PACA) nanoparticle.
6. The drug delivery system according to any one of the claims 1-5, wherein the nanoparticles associated with the gas-filled microbubbles stabilizes the microbubbles.
7. The drug delivery system according to anyone of the claims 1-6, wherein the nanoparticles further comprising at least one targeting agent.
8. The drug delivery system according to anyone of the claims 1-7, further comprising a pharmaceutically acceptable carrier.
9. The drug delivery system according to any one of the claims 1-8, wherein the nanoparticles further are coated with polyethylene glycol (PEG).
10. The drug delivery system according to any one of the claims 1-9, wherein the mean diameter of the gas-filled microbubbles associated with nanoparticles is in the range 0,5 to 30 μm.
11. The drug delivery system according to any one of the claims 1-10, wherein the therapeutic agent is chemotherapeutic agent or a chemopotentiator.
12. The drug delivery system according to any one of the claims 1-11, wherein the gas-filled microbubbles is filled with a gas selected from the group consisting of: air, perfluorocarbon, N2, O2, CO2.
13. The drug delivery system according to any one of the claims 1-12, wherein the ultrasound-mediated delivery is mediated by ultrasound, such as focused ultrasound.
14. The drug delivery system according to any one of the claims 1-13, wherein the microbubbles is destroyable upon application of focused ultrasound thereto.
15. A method for preparing a drug delivery system according to the claims 1-14, comprising the steps of:
a. Synthesizing the nanoparticles to be loaded with the therapeutic agent.
b. Adding nanoparticles to a solution comprising a surface-active substance.
c. Mixing the solution with gas to obtain gas-filled bubbles.
16. A method according to claim 15, wherein the microbubbles is stabilized by self-assembly of nanoparticles in the gas-water interface.
17. A method according to any one of the claims 15-16, wherein the solution with gas is mixed for a desired time and/or desired speed to obtain microbubbles of desired size.
18. A method according to any one of the claims 15-17, wherein the solution in c) is mixed from 2 seconds to 60 minutes, preferentially 1 to 10 minutes.
19. A method according to anyone of the claims 15-18, wherein the solution in c) is mixed at 500 to 50000 rpm, preferentially 1 000 to 30000 rpm
20. A method according to anyone of the claims 15-18, wherein the surfaceactive substance is a serum, a protein or a lipid or a surfactant.
21. The drug delivery system according to any one of the claims 1-14 for use in treatment of cancer, wherein at least one of the nanoparticles is loaded with a therapeutic agent and delivery of the nanoparticles and the therapeutic agent to tumorous tissue is facilitated by an acoustic field, such as by ultrasound.
22. Use according to claim 21, wherein the acoustic field causes cavitation, oscillation and/or collapse of the gas-filled microbubbles.
23. Use according to anyone of the claims 21-22, wherein the cavitation improves the transport of nanoparticles across the capillary wall.
24. A composition for use in treatment of cancer comprising a drug delivery system according to anyone of the claims 1-14.
NO20161568A 2016-09-29 2016-09-29 A new drug delivery system for treatment of cancer NO342271B1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
NO20161568A NO342271B1 (en) 2016-09-29 2016-09-29 A new drug delivery system for treatment of cancer
JP2019517040A JP2019529488A (en) 2016-09-29 2017-09-29 Novel drug delivery system for treating disease
US16/338,152 US20200023073A1 (en) 2016-09-29 2017-09-29 A new drug delivery system for treatment of disease
PCT/EP2017/074798 WO2018060437A1 (en) 2016-09-29 2017-09-29 A new drug delivery system for treatment of disease
EP17784222.6A EP3518900A1 (en) 2016-09-29 2017-09-29 A new drug delivery system for treatment of disease
CA3040016A CA3040016A1 (en) 2016-09-29 2017-09-29 A new drug delivery system for treatment of disease
CN201780073657.2A CN110022860A (en) 2016-09-29 2017-09-29 For treating the new drug delivery system of disease

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
NO20161568A NO342271B1 (en) 2016-09-29 2016-09-29 A new drug delivery system for treatment of cancer

Publications (2)

Publication Number Publication Date
NO20161568A1 NO20161568A1 (en) 2018-03-30
NO342271B1 true NO342271B1 (en) 2018-04-30

Family

ID=62091609

Family Applications (1)

Application Number Title Priority Date Filing Date
NO20161568A NO342271B1 (en) 2016-09-29 2016-09-29 A new drug delivery system for treatment of cancer

Country Status (1)

Country Link
NO (1) NO342271B1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019185685A1 (en) * 2018-03-27 2019-10-03 Sintef Tto As Poly (alkyl cyanoacrylate) nanoparticles for use in treatment of cancer

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1701745A1 (en) * 2003-12-22 2006-09-20 Bracco Research S.A. Gas-filled microvesicle assembly for contrast imaging
US20110200530A1 (en) * 2008-10-07 2011-08-18 Bracco Research S.A. or Bracco Suisse S.A. Targeting construct comprising anti-polymer antibody and contrast/therapeutic agents binding to the same
WO2012094541A2 (en) * 2011-01-05 2012-07-12 The Regents Of The University Of California Acoustically responsive particles with decreased cavitation threshold
EP2508207A1 (en) * 2011-03-31 2012-10-10 BioAlliance Pharma Nanoparticles loaded with chemotherapeutic antitumoral Drug
US20130183244A1 (en) * 2010-09-10 2013-07-18 The Johns Hopkins University Rapid Diffusion of Large Polymeric Nanoparticles in the Mammalian Brain
US20140212502A1 (en) * 2013-01-31 2014-07-31 National Yang-Ming University Nano- and micro-bubbles with ultrasound-triggered release and imaging functionalities
WO2014191502A1 (en) * 2013-05-28 2014-12-04 Sinvent As Process for preparing stealth nanoparticles

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1701745A1 (en) * 2003-12-22 2006-09-20 Bracco Research S.A. Gas-filled microvesicle assembly for contrast imaging
US20110200530A1 (en) * 2008-10-07 2011-08-18 Bracco Research S.A. or Bracco Suisse S.A. Targeting construct comprising anti-polymer antibody and contrast/therapeutic agents binding to the same
US20130183244A1 (en) * 2010-09-10 2013-07-18 The Johns Hopkins University Rapid Diffusion of Large Polymeric Nanoparticles in the Mammalian Brain
WO2012094541A2 (en) * 2011-01-05 2012-07-12 The Regents Of The University Of California Acoustically responsive particles with decreased cavitation threshold
EP2508207A1 (en) * 2011-03-31 2012-10-10 BioAlliance Pharma Nanoparticles loaded with chemotherapeutic antitumoral Drug
US20140212502A1 (en) * 2013-01-31 2014-07-31 National Yang-Ming University Nano- and micro-bubbles with ultrasound-triggered release and imaging functionalities
WO2014191502A1 (en) * 2013-05-28 2014-12-04 Sinvent As Process for preparing stealth nanoparticles

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
Baghirov, Habib, et al. Focused ultrasound-mediated transport of poly(alkyl) cyanoacrylate nanoparticles across the blood-brain barrier in a melanoma brain metastasis model. Cancer Research, (2016) Vol. 76, No. 14, Supp. Supplement. Abstract Number: 2073. Meeting Info: 107th Annual Meeting of the American Association for Cancer Research, AACR 2016. New Orleans, LA, United States. 16 Apr 2016-20 Apr 2016. ISSN: 1538-7445, Dated: 01.01.0001 *
Chaudhari K. R., et al. Targeting effiency and biodistribution of zoledronate conjugated docetaxel loaded PBCA nanoparticles for bone metastasis. Adv. Funct. Mater. 2012, 22, 4101-4114., Dated: 01.01.0001 *
De Cock, Ine, et al. Sonoprinting and the importance of microbuble loading for the ultrasound mediated cellular delivery of nanoparticles. Biomaterials, 1 Mar 2016 Vol. 83, pp. 294-307. ISSN: 0142-9612; E-ISSN: 1878-5905., Dated: 01.01.0001 *
Hansen, Rune; Stenstad, Per; Morch, Yrr. Ultrasound mediated delivery of a novel nanoparticle microbubble platform. Cancer Research, (15 Apr 2013) Vol. 73, No. 8, Supp. SUPPL. 1. Abstract Number: 5618. Meeting Info: 104th Annual Meeting of the American Association for Cancer Research, AACR 2013. Washington, DC, United States. 06 Apr 2013-10 Apr 2013. ISSN: 0008-5472 DIGITAL OBJECT ID: 10.1158/1538-7445.AM2013-5618., Dated: 01.01.0001 *
Liu, Zhe et al. Iron oxide nanoparticle-containing microbubble composites as contrast agents for MR and ultrasound dual-modality imaging, Biomaterials (2011.05.05) vol. 32. no. 26. pp. 6155-6163., Dated: 01.01.0001 *
Moon, Hyungwon, et al. Multifunctional theranostic contrast agent for photoacoustics- and ultrasound-based tumor diagnosis and ultrasound-stimulated local tumor therapy. Journal of Controlled Release, 28 Nov 2015 Vol. 218, pp. 63-71. ISSN: 0168-3659; E-ISSN: 1873-4995, Dated: 01.01.0001 *
Morch, Yrr et al. Nanoparticle-stabilized microbubbles for multimodal imaging and drug delivery. Contrast Media and Molecular Imaging, (September/October 2015) Vol. 10, No. 5, pp. 356-366. ISSN: 1555-4309; E-ISSN: 1555-4317, Dated: 01.01.0001 *
Wang, Jiayu, et al. A novel microbubble capable of ultrasound -triggered release of drug -loaded nanoparticles. Journal of Biomedical Nanotechnology, (March 2016) Vol. 12, No. 3, pp. 516-524. ISSN: 1550-7033; E-ISSN: 1550-7041, Dated: 01.01.0001 *
Zhang, Yu et al.Preparation, characterization and biocompatability of poly(ethylene glycol)-poly(n-butyl cyanoacrylate) nanocapsules with oil core via miniemulsion polymerization. European Polymer Journal 44 (2008) 1654-1661, Dated: 01.01.0001 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019185685A1 (en) * 2018-03-27 2019-10-03 Sintef Tto As Poly (alkyl cyanoacrylate) nanoparticles for use in treatment of cancer
WO2020192950A1 (en) * 2018-03-27 2020-10-01 Sintef Tto As Poly (alkyl cyanoacrylate) nanoparticles for use in treatment of cancer
US11806330B2 (en) 2018-03-27 2023-11-07 Sintef Tto As PACA and cabazitaxel for anti-cancer treatment

Also Published As

Publication number Publication date
NO20161568A1 (en) 2018-03-30

Similar Documents

Publication Publication Date Title
Snipstad et al. Ultrasound improves the delivery and therapeutic effect of nanoparticle-stabilized microbubbles in breast cancer xenografts
Cao et al. Drug release from phase-changeable nanodroplets triggered by low-intensity focused ultrasound
Deng et al. A laser-activated biocompatible theranostic nanoagent for targeted multimodal imaging and photothermal therapy
Zhang et al. Size-modulable nanoprobe for high-performance ultrasound imaging and drug delivery against cancer
Chen et al. Theranostic multilayer capsules for ultrasound imaging and guided drug delivery
Rapoport Drug-loaded perfluorocarbon nanodroplets for ultrasound-mediated drug delivery
Fokong et al. Image-guided, targeted and triggered drug delivery to tumors using polymer-based microbubbles
US20200023073A1 (en) A new drug delivery system for treatment of disease
Zhou et al. Ultrasound-mediated local drug and gene delivery using nanocarriers
Tanbour et al. Drug delivery systems based on polymeric micelles and ultrasound: a review
Mørch et al. Nanoparticle‐stabilized microbubbles for multimodal imaging and drug delivery
Chen et al. Lipid/PLGA hybrid microbubbles as a versatile platform for noninvasive image-guided targeted drug delivery
Husseini et al. Micelles and nanoparticles for ultrasonic drug and gene delivery
Cavalli et al. Micro-and nanobubbles: a versatile non-viral platform for gene delivery
Figueiredo et al. PLGA nanoparticles for ultrasound-mediated gene delivery to solid tumors
Ozcelikkale et al. Multifaceted transport characteristics of nanomedicine: needs for characterization in dynamic environment
US20090117177A1 (en) Echogenic microbubbles and microemulsions for ultrasound-enhanced nanoparticle-mediated delivery of agents
Jangjou et al. The promising shadow of microbubble over medical sciences: from fighting wide scope of prevalence disease to cancer eradication
Kashkooli et al. Ultrasound-mediated nano drug delivery for treating cancer: Fundamental physics to future directions
Ding et al. Polymer-mediated penetration-independent cancer therapy
Sun et al. Ultrasound-responsive peptide nanogels to balance conflicting requirements for deep tumor penetration and prolonged blood circulation
Oh et al. Effect of HIFU treatment on tumor targeting efficacy of docetaxel-loaded Pluronic nanoparticles
Zhou Ultrasound-mediated drug/gene delivery in solid tumor treatment
Shende et al. Role of solid-gas interface of nanobubbles for therapeutic applications
Olsman et al. Acoustic Cluster Therapy (ACT®) enhances accumulation of polymeric micelles in the murine brain

Legal Events

Date Code Title Description
MM1K Lapsed by not paying the annual fees