WO2019071261A1 - Méthodes de traitement du cancer faisant appel à une thermothérapie et/ou à une immunothérapie améliorée - Google Patents

Méthodes de traitement du cancer faisant appel à une thermothérapie et/ou à une immunothérapie améliorée Download PDF

Info

Publication number
WO2019071261A1
WO2019071261A1 PCT/US2018/054880 US2018054880W WO2019071261A1 WO 2019071261 A1 WO2019071261 A1 WO 2019071261A1 US 2018054880 W US2018054880 W US 2018054880W WO 2019071261 A1 WO2019071261 A1 WO 2019071261A1
Authority
WO
WIPO (PCT)
Prior art keywords
nanoparticles
tumor
medication
cells
patient
Prior art date
Application number
PCT/US2018/054880
Other languages
English (en)
Inventor
Gholam A. Peyman
Original Assignee
Peyman Gholam A
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 Peyman Gholam A filed Critical Peyman Gholam A
Priority to US16/200,195 priority Critical patent/US11433260B2/en
Publication of WO2019071261A1 publication Critical patent/WO2019071261A1/fr
Priority to US16/843,831 priority patent/US11660229B2/en
Priority to US16/988,697 priority patent/US20210002338A1/en
Priority to US17/220,878 priority patent/US20210220480A1/en
Priority to US17/902,814 priority patent/US20230000981A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N7/00Ultrasound therapy
    • A61N7/02Localised ultrasound hyperthermia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61HPHYSICAL THERAPY APPARATUS, e.g. DEVICES FOR LOCATING OR STIMULATING REFLEX POINTS IN THE BODY; ARTIFICIAL RESPIRATION; MASSAGE; BATHING DEVICES FOR SPECIAL THERAPEUTIC OR HYGIENIC PURPOSES OR SPECIFIC PARTS OF THE BODY
    • A61H39/00Devices for locating or stimulating specific reflex points of the body for physical therapy, e.g. acupuncture
    • A61H39/06Devices for heating or cooling such points within cell-life limits
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy
    • A61K9/0009Galenical forms characterised by the drug release technique; Application systems commanded by energy involving or responsive to electricity, magnetism or acoustic waves; Galenical aspects of sonophoresis, iontophoresis, electroporation or electroosmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/167Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0092Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin using ultrasonic, sonic or infrasonic vibrations, e.g. phonophoresis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/22Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for
    • A61B17/22004Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for using mechanical vibrations, e.g. ultrasonic shock waves
    • A61B2017/22005Effects, e.g. on tissue
    • A61B2017/22007Cavitation or pseudocavitation, i.e. creation of gas bubbles generating a secondary shock wave when collapsing
    • A61B2017/22008Cavitation or pseudocavitation, i.e. creation of gas bubbles generating a secondary shock wave when collapsing used or promoted
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/22Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for
    • A61B17/22004Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for using mechanical vibrations, e.g. ultrasonic shock waves
    • A61B17/22012Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for using mechanical vibrations, e.g. ultrasonic shock waves in direct contact with, or very close to, the obstruction or concrement
    • A61B2017/22014Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for using mechanical vibrations, e.g. ultrasonic shock waves in direct contact with, or very close to, the obstruction or concrement the ultrasound transducer being outside patient's body; with an ultrasound transmission member; with a wave guide; with a vibrated guide wire
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/22Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for
    • A61B17/22004Implements for squeezing-off ulcers or the like on the inside of inner organs of the body; Implements for scraping-out cavities of body organs, e.g. bones; Calculus removers; Calculus smashing apparatus; Apparatus for removing obstructions in blood vessels, not otherwise provided for using mechanical vibrations, e.g. ultrasonic shock waves
    • A61B2017/22027Features of transducers
    • A61B2017/22028Features of transducers arrays, e.g. phased arrays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B90/00Instruments, implements or accessories specially adapted for surgery or diagnosis and not covered by any of the groups A61B1/00 - A61B50/00, e.g. for luxation treatment or for protecting wound edges
    • A61B90/36Image-producing devices or illumination devices not otherwise provided for
    • A61B90/37Surgical systems with images on a monitor during operation
    • A61B2090/378Surgical systems with images on a monitor during operation using ultrasound
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N1/00Electrotherapy; Circuits therefor
    • A61N1/18Applying electric currents by contact electrodes
    • A61N1/32Applying electric currents by contact electrodes alternating or intermittent currents
    • A61N1/327Applying electric currents by contact electrodes alternating or intermittent currents for enhancing the absorption properties of tissue, e.g. by electroporation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N1/00Electrotherapy; Circuits therefor
    • A61N1/18Applying electric currents by contact electrodes
    • A61N1/32Applying electric currents by contact electrodes alternating or intermittent currents
    • A61N1/36Applying electric currents by contact electrodes alternating or intermittent currents for stimulation
    • A61N1/36002Cancer treatment, e.g. tumour
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N1/00Electrotherapy; Circuits therefor
    • A61N1/40Applying electric fields by inductive or capacitive coupling ; Applying radio-frequency signals
    • A61N1/403Applying electric fields by inductive or capacitive coupling ; Applying radio-frequency signals for thermotherapy, e.g. hyperthermia
    • A61N1/406Applying electric fields by inductive or capacitive coupling ; Applying radio-frequency signals for thermotherapy, e.g. hyperthermia using implantable thermoseeds or injected particles for localized hyperthermia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N2/00Magnetotherapy
    • A61N2/002Magnetotherapy in combination with another treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N7/00Ultrasound therapy
    • A61N2007/0039Ultrasound therapy using microbubbles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/02Radiation therapy using microwaves
    • A61N5/022Apparatus adapted for a specific treatment
    • A61N5/025Warming the body, e.g. hyperthermia treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N5/0613Apparatus adapted for a specific treatment
    • A61N5/062Photodynamic therapy, i.e. excitation of an agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N5/0613Apparatus adapted for a specific treatment
    • A61N5/0625Warming the body, e.g. hyperthermia treatment

Definitions

  • the invention generally relates to cancer treatment methods using thermotherapy and/or enhanced immunotherapy. More particularly, the invention relates to Wnt-inhibitors and other cellular pathway inhibitors in combination with immunothermotherapy or immunoradiothermotherapy.
  • Wnt signaling is involved in control of stem cell proliferation. Wnt mutation causes developmental defects in many disease processes including cancer.
  • Wnt proteins bind to receptors of the Frizzled and LRP protein families causing accumulation of beta-catenin in the cytoplasm and its translocation in the nucleus forms a complex with transcriptional cofactor (TCF) to activate the transcription of Wnt targeted genes.
  • TCF transcriptional cofactor
  • the Wnt pathway is considered canonical when it is dependent on beta-catenin or non-canonical when it is independent, Wnt signaling is involved in cell fate specification, cell proliferation, cell migration, and insulin sensitivity.
  • Wnt signaling is activated in many cancer stem cells and metastatic cells influencing the immune response to the cancer. Increased Wnt signaling releases a compound from the cancer cell by which the cancer cell evades recognition by the T- lymphocytes. Thus, increased Wnt signaling predicts a poor prognosis in cancer.
  • the cancer cell membrane produces the extracellular vesicles or exosomes that contain Wnt ligands.
  • Wnt ligands When these exosomes are picked up by neighboring cells, the Wnt ligands become liberated in the cytoplasm causing accumulation of ⁇ - catenin and translocated in the nucleus of the host cell, thereby influencing the Wnt related genes in the normal host cells.
  • This mechanism is used by cancer cells when the tumor metastasizes to prepare a niche for the growth of the circulating tumor cells.
  • Wnt activation is reported in adenomatous polyposis coli, Cholangiocarcinoma, Leukemia, leukemia initiating cell, Acute Myeloid leukemia, Chorionic lymphocytic leukemia, acute lymphoblastic leukemia, Melanoma, breast cancer, prostate cancer, glioblastoma, ovarian cancer, esophageal cancer , cervical cancer, etc.
  • Wnt inhibitors are compounds, such as FH535, IWP-2, PNU-74654, IWR-lendo, rWR-exo, Demethoxycurcumin, CCTO36477,KY02111, WAY-316606, SFRP , IWP , LGK974, C59, Antl.4Br/Ant 1.4C1, ivermectin, niclosamide, apicularen and bafilomycin, XAV939, XAV939, G007-LK and G244-LM, NSC668036, SB-216763, gemtuzumab, etc.
  • Glycogen Synthase Kinase-3 (GSK-3) is a serine/threonine protein kinase, which plays a key role in Wnt/ -catenin signaling during embryonic development, inflammation and cancer. Inhibition of GSK-3 inhibits Wnt pathway in cancer.
  • RHO associated protein Kinase is a kinase belonging to the family of serine- threonine Kinase involved in regulating the shape and the cytoskeleton of the cells, it is an important regulator of cell migration, stimulates PTEN phosphatase activity, leading to uncontrolled cell division in cancer.
  • Rock is active in inflammatory processes, cancer, Parkinson's disease, diabetes and many neurodegenerative diseases, such as Alzheimer's disease, and production and stiffen collagen in tumors, such as pancreatic cancer. Therefore, Rock inhibitors inhibit inflammatory processes, reduce TGF- ⁇ formation, block cell migration, and inhibit metastatic spread of the tumors.
  • ras oncogenes and the proteins (Ras) encoded genes have had an important roles in the study of human cancer affecting >50% of colon and >90% of pancreatic cancers.
  • Ras proteins are membrane-bound GTP-binding proteins that serve as molecular switches in mitogenic signal transduction. Prenylation of Ras proteins by intermediates of the isoprenoid analogs of Farnesyl Diphosphate inhibits Farnesyl transferase and Ras activation.
  • Ras-Ral signaling Other relevant pathways influencing the activation of Ras are PK1 and PK8 and P13K, RAF/MEK/ERK, PI3 K/ AKT/mTOR and RalA/B that can be inhibited by gemcitabine, wortmannin, LY294002 and inhibition of Cyclin-dependent kinase 5 (CDK5) and tipifarnib alone or in combination with Gemcitabine or rapamycin and AZD8055 (a TORKi) everolimus preventing Ras-Ral signaling.
  • CDK5 Cyclin-dependent kinase 5
  • tipifarnib alone or in combination with Gemcitabine or rapamycin and AZD8055 (a TORKi) everolimus preventing Ras-Ral signaling.
  • cancer therapy needs a robust immune response of one's body to be successful and prevent recurrences.
  • Antineoplastic medication kills tumor cells, inhibiting their metabolism and cell pathways.
  • the dead tumor cells are lysed by their enzymatic processes that exists inside the tumor cells or the enzymes that are present in any tissue releasing the tumor antigens.
  • the tumor antigen induces an immune response by being picked up by the dendritic cell in the lymph nodes.
  • the dendritic cells present the antigen to Cytotoxic T-Lymphocytes.
  • the cytotoxic T- lymphocytes migrate thorough the circulation, recognize the tumor cells and kill them.
  • the check point inhibitors prevent the tumor cells from escaping recognition by the cytotoxic T-lymphocytes, macrophages and killer cells leukocytes, etc.
  • Neoplastic medications attack cells in an indiscriminating manner, killing the tumor cells, normal cells, and weaken the immune response.
  • the checkpoint inhibitors though extremely beneficial, attack the checkpoint inhibitors of the normal cells and induce autoimmunity.
  • the present invention is directed to cancer treatment methods using thermotherapy and/or enhanced immunotherapy that substantially obviate one or more problems resulting from the limitations and deficiencies of the related art.
  • a cancer treatment method using controlled localized thermotherapy comprises the steps of: (i) administering a plurality of nanoparticles to a patient in need thereof so as to target a tumor in the patient, the administered nanoparticles being coated with an antitumor antibody and a thermosensitive polymer, and the administered nanoparticles containing quenched fluorescein, liposomes, fluorescent dextrans, polymer micelles, or nanoparticles carrying a dye or another dye indicator, such as bubble liposomes containing air pocket or nanoemulsion of PFC carrying fluorescein, a gene and a drug in the
  • thermosensitive polymer coating e.g., poly(N-isopropylacrylamide); (ii) heating the nanoparticles with an energy source to a temperature of about 41 °C to about 43 °C so as to damage one or more tumor cell membranes at a treatment site of the tumor and melt the thermosensitive polymer coating of the nanoparticles, thereby releasing the fluorescein into the circulation of the patient; and (iii) radiating a nail bed of the patient or any other part of the body with ultraviolet radiation so that the released fluorescein fluoresces, thus indicating that the temperature of about 41°C to about 43 °C has been achieved at the treatment site.
  • thermosensitive polymer coating e.g., poly(N-isopropylacrylamide
  • the nanoparticles comprise perfluorocarbon liquid (PFCL) nanoparticles
  • the method further comprising the steps of: (iv) additionally heating the nanoparticles with the energy source to a temperature of about 56°C so as to reach the boiling point of the PFCL nanoparticles, thereby creating a detectable cavitation sound; and (v) recording the detectable cavitation sound with an ultrasonic receiver or microphone so that a control signal is capable being sent to the energy source to indicate that the heating of target tissue in the patient is to be stopped, thereby preventing thermal damage to surrounding healthy cells.
  • PFCL perfluorocarbon liquid
  • the antitumor antibody of the nanoparticles is in the form of a monoclonal antibody.
  • thermosensitive polymer coating of the nanoparticles further comprises at least one medication configured to be released when the thermosensitive polymer is melted, the medication present in an amount to provide the patient a dose lower than a conventional dose of the medication due to the one or more tumor cell membranes being previously compromised by the thermotherapy of the method.
  • thermosensitive polymer coating of the pluralities of the nanoparticles further comprises checkpoint inhibitors alone or in combination with Rock inhibitors configured to be released when the thermosensitive polymer is melted, the checkpoint inhibitors being used in a localized immunotherapy treatment procedure targeting the cells of the tumor without causing damage to the healthy cells of the patient, the Rock inhibitor reducing TGF- ⁇ production after therapy and the subsequent scar formation.
  • the energy source heats the nanoparticles using focused ultrasonic energy in a thermal mode.
  • the nanoparticles are selected from a group consisting of iron oxide gold nanoparticles, gold graphene oxide nanoparticles, gold nanoparticles, silicone nanoparticles, carbon nanoparticles, magnetic nanoparticles, gold nanorods, gold nanoshells, gold nanocages, iron oxide nanotubes, gold nanotubes, carbon nanotubes, and combinations thereof.
  • the nanoparticles are further conjugated with cell penetrating peptides (CPPs) or activatable cell-penetrating peptides (ACPPs) so to enhance cell penetration into the cells of the tumor.
  • CPPs cell penetrating peptides
  • ACPPs activatable cell-penetrating peptides
  • the released fluorescein fluoresces as green light passing through a blue absorbing filter, the blue absorbing filter being operatively connected to a photomultiplier via a fiber optic, the photomultiplier converting the green light into an electrical signal that is delivered to a processor, the processor configured to control the energy source at the desired temperature of about 41°C to about 43 °C for a predetermined period of time.
  • the step of administering a plurality of nanoparticles to a patient further comprises administering a plurality of bubble liposomes filled with the dye or indicator or administering nanoemulsions of perfluorocarbon (PFC) carrying the dye or indicator, the dye or indicator comprising fluorescein.
  • PFC perfluorocarbon
  • the step of heating the antitumor-antibody-coated nanoparticles releases tumor antigens in the circulation of the patient as a result of thermally damaging the tumor cells; and the method further comprising the steps of: (iv) obtaining from the blood of the patient, the tumor antigens to build a new potent vaccine against many additional tumor specific antigens, the vaccine combined with antitumor-antibody-coated nanoparticles conjugated with checkpoint inhibitors, and Rock inhibitors or Wnt inhibitors; (v) administering the vaccine with the antitumor-antibody-coated nanoparticles conjugated with viral-like particles (VLP) and/or oncolytic viruses while simultaneously releasing conjugated checkpoint inhibitors, and Rock inhibitors or Wnt inhibitors, from the antitumor- antibody-coated nanoparticles to prevent new or old tumor cells, metastatic cells, and/or tumor exosomes from being disguised from the T-lymphocytes or the patient' s natural killer (NK
  • the cancer treatment method further comprises the step of: (iv) systemically administering a monoclonal antibody in the form of tocilizumab so as to target interleukin-6 and ornithine phenylacetate, an ammonia scavenger, for the treatment of hepatic encephalopathy.
  • thermotherapy comprises systemically administering tumor-antibody-coated nanoparticles coated with a thermosensitive polymer, cell penetrating peptides (CPPs), and a Wnt inhibitor being conjugated with the thermosensitive polymer coating of the nanoparticles
  • the thermotherapy further comprises heating the tumor-antibody-coated nanoparticles using an energy source at the site of the tumor so as to melt the thermosensitive polymer coating of the nanoparticles and release the Wnt inhibitor to inhibit the Wnt/ -catenin pathway in the cells of the tumor, wherein the heating of the nanoparticles further damages one or more tumor cell membranes and releases antigenic material in vivo that activates and stimulates an immunogenic response of the patient at the site of the tumor; and immunotherapy comprises systemically administering the patient's natural killer (NK) cells/den
  • thermosensitive polymer coating of the nanoparticles further comprises at least one rho-kinase inhibitor configured to be released when the thermosensitive polymer is melted, the rho-kinase inhibitor acting as an anti-inflammatory agent to prevent a cytokine storm resulting from the immunotherapy, and to inhibit Wnt activation in the cells of the tumor.
  • the method further comprises the step of removing cytokines after the immunotherapy by electrophoresis, plasmapheresis, plasma exchange, or ARF6 inhibition so to prevent a cytokine storm, or oral Rho kinase inhibitors.
  • thermosensitive polymer coating of the nanoparticles further comprises metformin, buformin, or phenformin so as to inhibit glucose metabolism in the cells of the tumor.
  • thermosensitive polymer coating of the nanoparticles further comprises ivermectin or niclosamide configured to be released when the thermosensitive polymer is melted, the ivermectin or niclosamide inhibiting the Wnt/ ⁇ - catenin pathway in the cells of the tumor.
  • thermosensitive polymer coating of the nanoparticles further comprises metformin, buformin, or phenformin, and additionally comprises syrosingopine so as to inhibit glucose metabolism in the cells of the tumor.
  • thermosensitive polymer coating of the nanoparticles further comprises an inhibitory gene(s) and a CRISPR/cas9 complex to stimulate or modify tumor genes at the site of the tumor upon release from the
  • thermosensitive polymer coating of the nanoparticles at a temperature of about 41°C to about 43°C.
  • gene modification is done using antibody coated nanoparticles conjugated with CRISPR/cas9 mediated homology-independent targeted integration (HITI) or homology directed repair (HDR).
  • HITI CRISPR/cas9 mediated homology-independent targeted integration
  • HDR homology directed repair
  • the cancer treatment method further comprises the step of orally administering one or more rho-kinase inhibitors acting as anti-inflammatory agents to prevent a cytokine storm resulting from the immunotherapy and/or one or more Wnt/ -catenin pathway inhibitors to inhibit the Wnt/ -catenin pathway in the cells of the tumor.
  • the cancer treatment method further comprises the steps of growing a predetermined quantity of tumor cells of the patient in vitro and applying thermal or non-thermal radiation to the tumor cells so as to damage or kill the genetic component of the tumor cells, while maintaining the antigenicity of the tumor cell proteins; and administering tumor lysates conjugated with antibody coated nanoparticles, VLP, one or more rho kinase inhibitors, and/or Wnt inhibitors to induce an immune response to the tumor cells, circulating tumor cells, and tumor exosomes.
  • a cancer treatment and imaging method using compressive ultrasound comprising the steps of: (i) administering a plurality of nanoparticles to a patient in need thereof so as to target a tumor in the patient, the administered nanoparticles being coated with an antitumor antibody and a thermosensitive polymer, and the administered nanoparticles containing medication and/or gene and quenched fluorescein in the thermosensitive polymer coating, at least some of the nanoparticles attaching to surface antigens of tumor cells of the tumor so as to form a tumor cell/nanoparticle complex; (ii) exciting the nanoparticles in a first compressive non-thermal mode using a ultrasound with a frequency of generally less than 100 kilohertz (kHz) or less than 300 kHz and a power of less than one Watt from a power source that generates a focused compressive ultrasonic wave so as to peel off the thermosensitive polymer coating of the nanop
  • the focused ultrasonic wave generated by the ultrasound source has a frequency between about 10 kilohertz and about 100 kilohertz, and a power of about 1 Watt per cm 2 .
  • the method further comprises the steps of (i) heating the nanoparticles in a second thermal mode using the focused ultrasound source at a frequency of greater than 1 megahertz (MHz) to 50 megahertz (MHz) and a power of greater than 1 Watt/cm 2 to 50 Watts/cm 2 or more that raises the temperature of the tumor
  • the ultrasonic wave generated by the ultrasound source has a frequency between about 150 kilohertz (kHz) and about 300 kilohertz (kHz), and a power of 1 Watt/cm 2 to 50 Watts/cm 2 or more.
  • the nanoparticles comprise piezoelectric nanoparticles, and where the focused compressive ultrasonic wave in the first compressive non-thermal mode is delivered in a pulsed manner so as to generate an electric pulse from the piezoelectric nanoparticles that depolarizes the one or more tumor cell membranes so as to damage the tumor cells.
  • the nanoparticles are coated with one or more antibodies, and the antibody coated nanoparticles contain medication and the medication is selected from the group consisting of Wnt inhibitors, Rock inhibitors, metformin, buformin, syrosingopine, phenformin, anti-VEGFs, checkpoint inhibitors, and combinations thereof.
  • the dye or indicator released is fluorescein.
  • piezoelectric nanoparticles are used to generate a sound wave from an electric pulse (from e.g., a battery) from the exposed nanoparticles, such as in a telephone receiver, and also the piezoelectric nanoparticles may be exposed to an ultrasonic pulse, which is absorbed by the piezoelectric nanoparticles, thus converting the sound wave into an electric pulse.
  • an electric pulse from e.g., a battery
  • the piezoelectric nanoparticles may be exposed to an ultrasonic pulse, which is absorbed by the piezoelectric nanoparticles, thus converting the sound wave into an electric pulse.
  • these principles may be utilized to image a tumor using an external electric pulse and antibody coated piezoelectric nanoparticles to create a sound wave inside the body in the area to be imaged or apply the external ultrasound to antibody coated piezoelectric nanoparticles conjugated with a medication to treat a tumor non-invasively inside the body by depolarizing the tumor cell membrane by the generated electrons from the piezoelectric nanoparticles and make the cell membrane permeable to the medication, e.g., anti-cancer medication, etc. by the internally generated electrical pulses.
  • the antibody coated piezoelectric nanoparticles or piezoelectric quantum dots may be administered intravenously, intra-arterially, topically by injecting submucosally, and/or spraying a nasal mucosa to travel through olfactory nerves to the brain
  • piezoelectric nanoparticles can be rendered biocompatible by coating with zirconate titanate, perovskite-based oxides, barium titanate, polyvinylidene fluoride (PVDF), piezoelectric nanoparticles, nanoparticles coated with chitosan, PEG, biotin, streptavidin, radionuclides, cell penetrating peptides (CPPs), activatable cell penetrating peptides (ACCPs), etc. for cell penetration to deliver drugs or genes, etc.
  • PVDF polyvinylidene fluoride
  • a cancer treatment and imaging method comprising the steps of: (i) systemically administering intravenously antibody coated piezoelectric or pyroelectric nanoparticles to a patient in need thereof so as to target a tumor in the patient, the piezoelectric or pyroelectric nanoparticles being coated with a thermosensitive polymer, and a medication being conjugated with the thermosensitive polymer coating of the piezoelectric or pyroelectric nanoparticles, the piezoelectric or pyroelectric nanoparticles travel through the body attaching to surface antigens of tumor cells of the tumor so as to form a tumor cell/nanoparticle complex; (ii) applying a pulsed electrical current to the piezoelectric or pyroelectric nanoparticles using an electrical source at the site of the tumor so as to create an electroacoustic sound from the piezoelectric or pyroelectric nanoparticles; (iii) recording the electroacoustic
  • the electroacoustic system can be used to scan the entire body, by systemically administering, or intravenously administering, antibody coated piezoelectric or pyroelectric nanoparticles to a patient in need thereof so as to target a tumor in the patient, a metastatic lesion, circulating tumor cells, and their exosomes, the piezoelectric or pyroelectric nanoparticles being coated with a thermosensitive polymer, and a medication being conjugated with the thermosensitive polymer coating/ CPP of the piezoelectric or pyroelectric nanoparticles, the piezoelectric or pyroelectric nanoparticles travel through the body attaching to surface antigens of tumor cells of the tumor so as to form a tumor cell/nanoparticle complex.
  • a pulsed electrical current is applied from moveable electrodes placed stepwise from the top of the head to the bottom of feet of the patient to excite the piezoelectric or pyroelectric nanoparticles attached to the tumor cells while using an electrical pulse at the site so as to create an electroacoustic sound from the piezoelectric or pyroelectric nanoparticles attached to the tumor cells indicating the presence of the tumor in that area, and reconstructing a full-body electroacoustic tomogram of the patient.
  • the electrical pulses can vary from 1-30 pulses per second (30 Hz) up to 1 kHz, or up to 1 MHz or more, or the electrical pulses can be longer than one second to induce various electrical responses in the tumor to which the antibody coated piezoelectric nanoparticles are attached.
  • the lesion discovered by electroacoustic tomogram is treated with focused ultrasound and antibody coated piezoelectric nanoparticles conjugated with either stimulating genes or inhibitory genes, or genes are replaced using CRISPR cas9 mediated Homology-Independent Targeted Integration (HITI) or Homology Directed Repair (HDR).
  • HITI Homology-Independent Targeted Integration
  • HDR Homology Directed Repair
  • the genetic composition of the tumor can be modified using electroacoustic tomography and administration of nanoparticle conjugated gene(s) using CRISPR cas9 mediated Homology-Independent Targeted Integration (HITI) or Homology Directed Repair (HDR).
  • CRISPR cas9 mediated Homology-Independent Targeted Integration
  • HDR Homology Directed Repair
  • the disease being treated may be a genetic disease, such as Alzheimer's disease, retinal degeneration, Parkinson's disease, muscular dystrophy, diabetes, lung and other genetic diseases of the skin, kidney, or spinal cord, etc.
  • a genetic disease such as Alzheimer's disease, retinal degeneration, Parkinson's disease, muscular dystrophy, diabetes, lung and other genetic diseases of the skin, kidney, or spinal cord, etc.
  • the method further comprises the step of (v) increasing the permeability of one or more tumor cell membranes of the tumor using the pulsed ultrasound, thereby initiating an electric pulse from the piezoelectric nanoparticles, e.g.
  • quartz or perovskites coating with zirconate titanate, perovskite-based, oxides, barium titanate, polyvinylidene fluoride (PVDF), piezoelectric nanoparticles, nanoparticles coated with chitosan, PEG, biotin, streptavidin, radionuclides, CPPs, ACCPs to depolarize the tumor cells attached to the nanoparticles, and minimally facilitating the entry of the medication into the depolarized tumor cells of the tumor.
  • PVDF polyvinylidene fluoride
  • the method further comprises the step of (v) increasing the permeability of one or more tumor cell membranes of the tumor using the pulsed focused ultrasound, thereby initiating an electric pulse from the piezoelectric nanoparticles, e.g., quartz or perovskites, conjugated with a medication, CPP, gene or VLP exposed to the ultrasound to depolarize the tumor cells, while increasing the temperature of the tissue with the focused ultrasonic waves and simultaneously measuring the tissue temperature with a second harmonic wave resulting from back scattered ultrasound generated from the heated tissue recorded with a transducer located on the patient's skin, connected to an imaging system, recording the tissue temperature and enhancing penetration of the piezoelectric nanoparticles inside the cell that release the medication, etc.
  • the piezoelectric nanoparticles e.g., quartz or perovskites, conjugated with a medication, CPP, gene or VLP exposed to the ultrasound to depolarize the tumor cells
  • This imaging unit is in turn, connected via software to the initial focused ultrasound producing unit, controlling the intensity of the pulsed ultrasound keeping it at less than 100 kHz intensity and the power at less than 1 W/cm 2 power to peel off and release medication from the nanoparticles, e.g. from antibody coated quartz or perovskites nanoparticles conjugated with medication (e.g., similar to the manner in which an ultrasonic watch or instrument cleaner removes the dirt and cleaning the watch or instrument) and to simultaneously depolarize the tumor cells attached to the piezoelectric nanoparticles, thus facilitating the entry of the medication into the depolarized tumor cell membranes, and as needed, to heat them under the control of power of the focused ultrasound connected to the thermal imaging system via a processor.
  • the initial focused ultrasound producing unit controlling the intensity of the pulsed ultrasound keeping it at less than 100 kHz intensity and the power at less than 1 W/cm 2 power to peel off and release medication from the nanoparticles, e.g. from antibody coated quartz or perovskites nanoparticles conjugated with
  • the method further comprises the step of (v) heating the piezoelectric or pyroelectric nanoparticles using a ultrasound source operating in a thermal mode so as to raise the temperature of the tumor cell/nanoparticle complex controllably to a temperature of about 41°C to about 43 °C, thereby damaging one or more tumor cell membranes at the tumor site and melting the thermosensitive polymer coating of the nanoparticles to release the medication at the tumor site.
  • the electrical source comprises a battery device with an anode lead positioned on a first side of the body (neck) of the patient and a cathode being located on a second side of the neck of the patient, the pulsed electrical current passing through the neck of the patient from the anode to the cathode of the battery device, and where the pulsed electrical current passes through a tumor pretreated with intravenous or intra- arterial injection of the tumor supplying the artery with antibody coated piezoelectric nanoparticles conjugated with medication attached to the tumor cells, where an electrical pulse creates an ultrasonic wave from the piezoelectric nanoparticles that can be recorded by an ultrasonic transducer located on the skin, imaged and localize the tumor precisely, then the lesion is treated non-invasively with a focused ultrasound beam through the skin and simultaneously heating up the tissue to a temperature of 39-40 degrees C to damage the tumor cells with the thermal energy and depolarizing
  • a cancer treatment method using focused ultrasound comprising the steps of: (i) administering a plurality of piezoelectric or pyroelectric nanoparticles to a patient in need thereof so as to target a tumor in the patient, the administered piezoelectric or pyroelectric nanoparticles being coated with an antitumor antibody and a thermosensitive polymer, and the administered piezoelectric or pyroelectric nanoparticles containing medication, a gene, a checkpoint inhibitor, and/or viral-like particles (VLP) , 1L-2, ACPP, toxin(s), TLK 7/8 to stimulate a cellular immune response and quenched fluorescein in the thermosensitive polymer coating, at least some of the piezoelectric or pyroelectric nanoparticles attaching to surface antigens of tumor cells of the tumor so as to form a tumor cell/nanoparticle complex; and (ii) stimulating the piezoelectric or pyr
  • thermosensitive polymer coating of the piezoelectric or pyroelectric nanoparticles upon the heating of the nanoparticles to a temperature of about 41°C to about 43°C.
  • the antibody coated piezoelectric or pyroelectric nanoparticles are further conjugated with cell penetrating peptides (CPPs) or activatable cell-penetrating peptides (ACPPs) so to enhance cell penetration into the cells of the tumor prior to treatment to release the medication inside the tumor cells during nonthermal therapy with focused ultrasound at pulses of less than 100 kHz and a power of less than 1 Watt/cm 2 .
  • CPPs cell penetrating peptides
  • ACPPs activatable cell-penetrating peptides
  • the nanoparticles are coated with one or more antibodies, and the antibody coated nanoparticles contain medication and the medication is selected from the group consisting of Wnt inhibitors, Rock inhibitors, GSK inhibitors, metformin, buformin, syrosingopine, phenformin, anti-VEGFs, checkpoint inhibitors, and combinations thereof further conjugated with cell penetrating peptides (CPPs) or activatable cell-penetrating peptides (ACPPs) so to enhance cell penetration into the cells of the tumor prior to treatment to release the medication inside the tumor cells during non-thermal therapy with focused ultrasound at pulses of less than 100 kHz and a power of less than 1 Watt/cm 2 .
  • CPPs cell penetrating peptides
  • ACPPs activatable cell-penetrating peptides
  • a cancer treatment method using immunotherapy comprising the steps of: (i) administering a plurality of nanoparticles to a patient in need thereof so as to target a tumor in the patient, the administered nanoparticles being coated with an antitumor antibody and a thermosensitive polymer conjugated with cell penetrating peptides (CPPs), and the administered nanoparticles containing a checkpoint inhibitor and an immune stimulator in the thermosensitive polymer coating, at least some of the nanoparticles attaching to surface antigens of tumor cells of the tumor so as to form a tumor
  • CPPs cell penetrating peptides
  • the cell/nanoparticle complex and at least others of the nanoparticles attaching to circulating tumor cells and/or tumor exosomes with receptors the same as, or similar to those of the tumor cells; and (ii) exciting the nanoparticles using a non- thermal energy source so as to peel off the thermosensitive polymer coating of the nanoparticles by vibrating the nanoparticles, thereby releasing the checkpoint inhibitor and the immune stimulator at the tumor site, proximate to the circulating tumor cells and/or proximate to the tumor exosomes so as to provide a multiple modality, localized immunotherapy treatment targeting the cells of the tumor, the circulating tumor cells, and/or the tumor exosomes by enhancing an immune response of the patient without causing damage to the healthy cells of the patient, while synergistically preventing the cells of the tumor, the circulating tumor cells, and/or the tumor exosomes from evading the immune system of the patient.
  • the administered nanoparticles further contain one or more Rock inhibitors in the thermosensitive polymer coating; and the exciting of the nanoparticles using the non-thermal energy source further releases the one or more Rock inhibitors from the thermosensitive polymer coating, the one or more Rock inhibitors reducing TGF- ⁇ production after the localized immunotherapy treatment therapy, and reducing subsequent scar formation.
  • the administered nanoparticles further contain one or more Wnt inhibitors in the thermosensitive polymer coating; and the exciting of the nanoparticles using the non-thermal energy source further releases the one or more Wnt inhibitors from the thermosensitive polymer coating, the one or more Wnt inhibitors inhibiting Wnt activation in the tumor cells of the tumor, the circulating tumor cells, and/or the tumor exosomes, and thus inhibiting tumor cellular proliferation.
  • the circulating tumor cells and/or the tumor exosomes are carrying a checkpoint protein that disguises the circulating tumor cells and/or the tumor exosomes as healthy cells of the patient; and the checkpoint inhibitor released from the thermosensitive polymer coating of the nanoparticles blocks the checkpoint protein of the circulating tumor cells and/or the tumor exosomes so that the circulating tumor cells and/or the tumor exosomes are recognized and killed by the T-cells and killer cells of the patient.
  • tumor antigens are released in the circulation of the patient; and wherein the method further comprising the steps of: (iii) obtaining from the blood of the patient, the tumor antigens to build a new potent vaccine against many additional tumor specific antigens, the vaccine combined with antitumor-antibody-coated nanoparticles conjugated with checkpoint inhibitors, cell penetrating peptides (CPPs), and Rock inhibitors or Wnt inhibitors; and (iv) administering the vaccine with the antitumor-antibody-coated nanoparticles conjugated with viral-like particles (VLP), activatable cell-penetrating peptides (ACPPs), and/or oncolytic viruses while simultaneously releasing conjugated checkpoint inhibitors, and Rock inhibitors or Wnt inhibitors, from the antitumor-antibody-coated nanoparticles to
  • thermosensitive polymer coating of the nanoparticle is selected from the group consisting of bee venom, scorpion venom, viral-like particles (VLPs), and combinations thereof; and the immune stimulator provides a signaling function for attracting T-cells and killer cells of the patient that attack the tumor cells of the tumor, the circulating tumor cells, and/or the tumor exosomes.
  • VLPs viral-like particles
  • the administered nanoparticles further contain oncolytic viruses in the thermosensitive polymer coating; and the exciting of the
  • nanoparticles using the non-thermal energy source further releases the oncolytic viruses from the thermosensitive polymer coating, the oncolytic viruses preferentially infecting and killing the tumor cells of the tumor, the circulating tumor cells, and/or the tumor exosomes.
  • the non-thermal energy source comprises a low power of about one Watt/cm 2 low frequency focused ultrasound source having a frequency between about 40 kilohertz and about 10 megahertz or a magnetic energy source generating an alternating magnetic field.
  • the multiple modality, localized immunotherapy treatment with the checkpoint inhibitor and the immune stimulator eliminates the need for CAR-T cellular therapy.
  • the exciting of the nanoparticles using the nonthermal energy source further increases the cell membrane permeability of the tumor cells of the tumor and/or the circulating tumor cells so as to enhance a penetration of a medication into the tumor cells of the tumor, the circulating tumor cells, and/or tumor exosomes.
  • a cancer treatment method using controlled localized thermotherapy, photodynamic therapy, and immunotherapy comprising the steps of: (i) administering a plurality of nanoparticles to a patient in need thereof so as to target a tumor in the patient, the administered nanoparticles being coated with an antitumor antibody and a thermosensitive polymer conjugated with cell penetrating peptides (CPPs), and the administered nanoparticles containing a photosensitizer and a checkpoint inhibitor and/or an immune stimulator in the thermosensitive polymer coating; and (ii) heating the nanoparticles with an energy source so as to induce a photodynamic effect and melt the thermosensitive polymer coating of the nanoparticles, thereby releasing the photosensitizer and the checkpoint inhibitor and/or the immune stimulator at a site of the tumor, circulating tumor cells, and/or tumor exosomes so as to provide a multiple modality, localized thermotherapy
  • photodynamic therapy, and immunotherapy treatment targeting the cells of the tumor, the circulating tumor cells, and/or the tumor exosomes by enhancing an immune response of the patient without causing damage to the healthy cells of the patient, while synergistically preventing the cells of the tumor, the circulating tumor cells, and/or the tumor exosomes from evading the immune system of the patient.
  • the energy source comprises an external or internal fiber optic device emitting laser light that provides the photodynamic therapy to the patient by delivering the laser light to the cells of the tumor, the circulating tumor cells, and/or the tumor exosomes to which a photosensitizer has been applied, the photodynamic therapy treating the patient by killing the cells of the tumor, the circulating tumor cells, and/or the tumor exosomes.
  • FIG. 1 illustrates a schematic diagram of a cancer treatment and evaluation system, according to an embodiment of the invention.
  • FIG. 2 illustrates a schematic diagram of a cancer treatment and imaging system, according to another embodiment of the invention.
  • FIG. 3 illustrates a schematic diagram of a cancer treatment system, according to yet another embodiment of the invention, wherein a thyroid tumor is being treated;
  • FIG. 4 illustrates a schematic diagram of a cancer treatment and imaging system, according to still another embodiment of the invention, wherein a thyroid tumor is being treated and imaged; and
  • FIG. 5 illustrates a schematic diagram of a cancer treatment and imaging system, according to yet another embodiment of the invention, wherein a thyroid tumor is being treated and imaged.
  • the controlled localized thermotherapy damages the tumor cells releasing their antigens augmenting the immune response without weakening the natural humoral or cellular response to the tumor cells.
  • the use of controlled thermotherapy at 41-43 degrees C at the tumor site using antibody coated nanoparticles weakens the tumor cells that preferentially are sensitive to heat compared to the normal cells and makes them susceptible to antitumor medication and/or immunotherapy reducing their ability to become chemo-resistant.
  • the use of controlled thermotherapy at 41-43 degrees C at the tumor site using antibody coated nanoparticles weakens the tumor cells that preferentially are sensitive to heat compared to the normal cells and makes them less likely to change their genetic composition to become less resistant to heat since the thermal effect is instantaneous when the temperature reaches these levels, or is maintained at the temperatures of these levels (41-43C) for a period of 5-10 minutes under the control of thermal delivery system.
  • the thermal energy being delivered at a frequency of generally 2 MHz to 50 MHz or 2 MHz to 100 MHz, and a power of more than one Watt.
  • the nanoparticles absorb preferentially either electromagnetic radiation or ultrasound, or respond to an alternating magnetic field, or a combination of them that can be heated to a desired degree of temperature under the control of a processor that records the change of the temperature in the body for a period of time, or increases it to higher detectable level of 56 C degrees, and imaged with a thermoacoustic imaging system for control of the temperature.
  • the antibody coated nanoparticles are chosen to last in the tumor cells and the treatment can be repeated locally (e.g., with breast cancer, ovarian cancer, cancer of brain tumors, lung cancer, prostate cancer) as desired without overheating the body, and the tumor can be imaged post treatment to examine the effect of the therapy on the tumor.
  • the pluralities of antibody coated nanoparticles for example, formed from magnetic and paramagnetic fullerene, carry thermosensitive polymers, such as chitosan hyaluronic acids which melts at a 41-43 degree C temperature and can release antineoplastic medication, gene modifying RNAi, CRISPR cas 9, etc., or gene modification is done through CRISPR cas9 mediated Homology -Independent Targeted Integration (HITI) or Homology Directed Repair (HDR).
  • HITI Homology -Independent Targeted Integration
  • HDR Homology Directed Repair
  • the pluralities of nanoparticles may also carry and release checkpoint inhibitors and/or Rock inhibitors from the thermosensitive polymer coating so as to attack the tumor cells with multiple modalities, and obtain post treatment biomarkers from the circulation for creating an appropriate vaccine of each patient's tumor for the future administration of the vaccine combined with antibody coated
  • nanoparticles/checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil
  • a Wnt inhibitor such as niclosamide
  • the pluralities of antibody coated nanoparticles carry with their thermosensitive polymeric coating one or multiple medications, such as cobimetinib, an MEKl and MEK2 inhibitor, BRAF and MEK inhibitors, sorafenib, vemurafenib, dabrafenib, pembrolizumab, a check point inhibitor, ipilimumab, nivolumab, olaparib, niraparib, pazopanib, dacarbazine, temozolomide, imatinib, carmustine, cisplatin, carboplatin, and paclitaxel, or a combination thereof for treatment of brain cancers, ovarian cancer, melanoma, lung cancer, in combination with controlled thermotherapy using an internal or external energy source at temperature of 41-43 C degree for 1-10 minutes as predetermined by the physician.
  • cobimetinib an MEKl and MEK2 inhibitor
  • BRAF and MEK inhibitors sora
  • the pluralities of antibody coated nanoparticles including piezoelectric nanoparticles or bubble liposomes carrying fluorescein which contain air pockets or perfluorocarbon (PFC) nanoemulsions carry with their thermosensitive polymeric coating one or multiple antineoplastic medications, check point inhibitors etc.
  • thermoacoustic imaging system for control of the temperature under the control of a processor controlling thermal energy, along with low- intensity (1-3 V/cm), low frequency 1-50-kHz or intermediate frequency 100-200 kHz non-thermal, and 300 kHz to 1 MHz or more to create thermal energy, electric fields achieving thermotherapy,
  • electroacoustic imaging using an array of ultrasound transducers, focused ultrasound, or a moving transducer etc. that can move and record the produced sounds from the piezoelectric nanoparticles transmitted to a processor or computer to 2-3D electroacoustic images or live video or reconstructed by a computer software, such as in focused ultrasound, and dielectric effect on the cellular components of the tumors, such as brain tumors, lung cancer, ovarian cancer, breast cancer, prostate cancer, and other cancers, which can be repeated numerous times post therapy to eliminate the cancer.
  • ACPPs activatable cell-penetrating peptides
  • the antibody coated pluralities of nanoparticles with activatable cell-penetrating peptides are selected from magnetic, paramagnetic, non-magnetic sphere, rod, nanocarbon, nanowire, nanorod, nanowire, magnetic nanoshells, nanocages, gold nanoshells or silica-gold nanoshells, silica iron oxide nanoshells, gold coated ferric oxide, quantum dots , magnetic and paramagnetic fullerene, encapsulate ferromagnetic nanoclusters, dendrimers, micelles of perfluorocarbon liquids, liposomes, liposomes in combination with the nanoparticle, nanoshells carrying quenched fluorescein, fluorescent dextrans/dye or liposomes or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide carrying a dye indicator in thermosensitive polymers that are administered to a patient to seek the potential tumor cells and attach to them exposed to the external energy source to release the fluor
  • the energy delivery system is a focused ultrasound which heats up the tissue located at the focal point of the focused ultrasound using a frequency of generally about 2 MHz to 50MHz or 2 MHz to 100 MHz and a power of more than one Watt and an antibody coated ultrasound contrast agent such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells nanocage magnetic, gold, silicone, carbon, piezoelectric nanotubes, perfluorocarbon (PFC) nanoemulsions, the mixture of magnetic nanoparticles, piezoelectric nanotransducers, boron nitride nanotubes, perovskites, and quenched fluorescein, fluorescent dextrans, or another dye indicator with the thermosensitive polymer.
  • the temperature is measured non-invasively by the estimation of the temperature depending on acoustic harmonic from 26-46 C degree generated by nonlinear ultrasound wave.
  • the thermal energy delivery system is a focused ultrasound which is capable of either producing non-thermal focused ultrasound at a single frequency of, for example, less than 300MHz and less than 1 Watt of power, in a non-thermal mode by regulating the duration of the software of the system to deliver focused ultrasonic pulses of a few microsecond duration or repeated in an interval that permits dissipation of thermal energy before the second pulse arrives while the same system can deliver focused ultrasound, for example, less than 1 MHz at a power of less than 1 Watt.
  • only the non-thermal mode such as generally at a frequency of about 300 kHz and less than one Watt of power, is used with pluralities of antibody coated ultrasound contrast agents, such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells, nanocages; magnetic, gold, silicone, carbon nanotubes, perfluorocarbon (PFC)
  • polymeric coating conjugated with quenched fluorescein or another dye or indicator such as polymeric materials, such as poly(Y-2-(2-(2-methoxyethoxy)-ethoxy)ethoxy-s
  • only the thermal mode of the focused ultrasound is used with pluralities of antibody coated ultrasound contrast agents, such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells, nanocages; magnetic, gold, silicone, carbon nanotubes, perfluorocarbon (PFC) nanoemulsions, piezoelectric nanotubes, or piezoelectric boron nitride nanotubes, nanogels, liposome and releases an antineoplastic medication/dye, a gene, and a polymeric coating conjugated with quenched fluorescein or another dye or indicator, such as polymeric materials, such as poly(Y-2-(2-(2-methoxyethoxy)-ethoxy-s-caprolactone)- £>-poly(y-octyloxy-s-caprolactone) and a dye, have demonstrated
  • the non-thermal mode and thermal mode of the focused ultrasound is used sequentially with pluralities of antibody coated ultrasound contrast agents, such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells, nanocages, magnetic, gold, silicone, carbon nanotubes, perfluorocarbon (PFC) nanoemulsions, piezoelectric nanotubes, or piezoelectric boron nitride nanotubes, nanogels, liposome and releases antineoplastic medication/dye, a gene, and a polymeric coating conjugated with quenched fluorescein another dye or indicator, such as polymeric materials, such as poly(y-2-(2-(2- methoxyethoxy)-ethoxy-s-caprolactone)- ?-poly(Y-octyloxy-s-caprolactone) and
  • the non-thermal mode and thermal mode of the focused ultrasound is used sequentially with another source of energy radiation or x-ray at lower doses than normally recommended to achieve a complementary effect on the tumors using pluralities of antibody coated ultrasound contrast agents, such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells, nanocages; magnetic, gold, silicone, carbon nanotubes, perfluorocarbon (PFC) nanoemulsions, piezoelectric nanotubes, or piezoelectric boron nitride nanotubes, nanogels, liposome and releases antineoplastic medication/dye, a gene, and a polymeric coating conjugated with quenched fluorescein another dye or indicator, such as polymeric materials, such as poly(Y-2-(2-(2-methoxyethoxy)-ethoxy-s-caprol
  • the thermal energy delivery system is a focused ultrasound which vibrates the tissue located at the focal point of the focused compressive ultrasound (e.g., 1-40 kHz frequency) using an antibody coated ultrasound contrast agent, such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells, nanocages; magnetic, gold, silicone, carbon nanotubes, perfluorocarbon (PFC) nanoemulsions, piezoelectric nanotubes, or piezoelectric boron nitride nanotubes, and releases antineoplastic medication, a gene, and a polymeric coating conjugated with quenched fluorescein another dye or indicator, such as polymeric materials, such as poly(Y-2-(2-(2-methoxyethoxy)-ethoxy-s-caprolactone)- £>-poly(Y-octyloxy-
  • an antibody coated ultrasound contrast agent such as gold
  • the energy delivery system is a focused ultrasound in a compressive non-thermal mode (e.g., at 1-100 kHz), which is visualized in the tissue by ultrasonic imaging only, the top of the cone-shaped image of the tissue is seen where the tip of the focused cone exerts the compressive or pressure force without producing significant thermal effect, or in combination with another imaging modality such as MRI or CT-scan or PET-scan that defines the focal point of the focused compressive ultrasound in relationship with the other body' s structures, using an antibody coated ultrasound contrast agent such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells nanocages, magnetic, gold, silicone, carbon nanotubes, piezoelectric nanotubes, the mixture of magnetic nanoparticles, perfluorocarbon (PFC) nanoemulsions, piezoelectric nanotransducers, boron n
  • GNPs gold nano
  • the energy delivery system is a focused ultrasound in a thermal mode which is visualized in the tissue by ultrasonic imaging only by seeing the top of the cone shaped image of the tissue where the tip of the focused cone produces a thermal effect seen as an increase in thermal coagulation and whitening of the tissue effect or in
  • an antibody coated ultrasound contrast agent such as gold nanoparticles (GNPs), magnetic or paramagnetic nanoparticles, gold, silicone, carbon nanotubes perfluorocarbon (PFC) nanoemulsions, piezoelectric particles or nanotubes, the mixture of magnetic nanoparticles, piezoelectric nanotransducers, boron nitride nanotubes, and quenched fluorescein, fluorescent dextrans or another dye or indicator with chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye or polymeric materials, such as poly(y-2-(2-(2-methoxyethoxy)- ethoxy)ethoxy-s-caprolactone)-£>-poly(y-octyloxy-
  • the patient is exposed to external or internal thermal energy to heat the antibody coated nanoparticles using laser, electromagnetic radiation, visible or infrared light, microwave, radiofrequency, a focused ultrasound, or an alternating magnetic field to heat and damage the tumor cells and increase tumor biomarker in the circulation after the thermotherapy having an important diagnostic value (i.e. indicating presence of a tumor by increased biomarkers) and therapeutic value for vaccine production after harvesting the circulating biomarkers and vaccine production for the future management of the tumor recurrences in the patient to reactivate the immune response and kill the tumor cells.
  • an important diagnostic value i.e. indicating presence of a tumor by increased biomarkers
  • therapeutic value for vaccine production after harvesting the circulating biomarkers and vaccine production for the future management of the tumor recurrences in the patient to reactivate the immune response and kill the tumor cells.
  • the standard ultrasonic transducer produces a divergent wave of ultrasound such that its tissue penetration depends on its frequency and the tissue property. The higher the frequency of the ultrasound, the lower is its transmission into the tissue. However, the ultrasound wave can be made to focus at a certain distance from its transducer head by using a concave ultrasonic transducer using a single array or multiple arrays.
  • the physical effects of the focused ultrasonic waves depend on the power and intensity of the ultrasound wave.
  • the focused ultrasound has a more compressive, low power 0.3 to less than 1 Watt and none, to minimal significant thermal effect (i.e., non-thermal or compressive mode LIFU), while the high power of 10-100 Watts/cm 2 , etc.
  • thermal mode HIFU thermal mode HIFU
  • the second or third harmonic ultrasound back scatter is measured indicating the temperature at the focal point of the focused ultrasound in the tissue.
  • the focused ultrasound can be imaged simultaneously with a non-focused ultrasound in order to image the lesion.
  • the energy source heats the antibody coated nanoparticles conjugated with medication, etc. using a focused low power .compressive or pressure mode ultrasound, and the nanoparticles with cell penetrating agents or activatable cell penetrating agents and quenched fluorescein or another dye or indicator with polymeric materials, such as P°ly(y-2-(2-(2-methoxyethoxy)-ethoxy)ethoxy-s-caprolactone)- ?-poly(Y-octyloxy-s- caprolactone) and a dye have demonstrated significant transition temperatures, allowing improved drug release at low hyperthermia (40 °C), are injected in the patient's circulation, lymphatic vessels, inside a body cavity, and the focused ultrasound is applied in the compressive low power non-thermal mode l-40KHz, where the dye is merely peeled off the nanoparticles or the polymeric carrier by its focused vibrational force.
  • a focused low power non-thermal mode ultrasound such as P°ly(y-2-(
  • the energy source which uses focused compressive or pressure mode ultrasound with low frequencies 1-140 kHz, and ⁇ 0.3 W is used with antibody coated nanoparticles that are conjugated with a medication, a gene, etc., and contain at least one radioactive agent conjugated with cell penetrating agents or activatable cell penetrating agents and quenched fluorescein, fluorescent dextrans, liposomes filled with fluorescein or another dye or indicator, or bubble liposomes carrying fluorescein which contain air pockets or
  • nanoemulsions of PFC are injected in the patient's circulation, lymphatic vessels, inside a body cavity, or cerebrospinal fluid, and the focused low power ultrasound is applied in the compressive or pressure mode (i.e., a non-thermal mode) where the dye is merely peeled off the nanoparticles by the focused vibrational force of the ultrasound while the tissue is imaged by the ultrasound or computer-assisted ultrasound imaging.
  • the compressive or pressure mode i.e., a non-thermal mode
  • the antibody coated nanoparticles are injected in the body, and are exposed to thermal energy, such as electromagnetic radiation.
  • the nanoparticles absorb the energy and expand producing a sound wave that is known as photoacoustic sound which is recorded by a transducer and converted to an electrical signal transmitted to a processor, and then converted to ID, 2D, or 3D computerized images.
  • antibody coated piezoelectric nanoparticles pyroelectric nanoparticles, or perfluorocarbon (PFC) nanoemulsions are injected inside the body, and the nanoparticles attach to the surface antigen of the normal cells or tumor cells and, when the nanoparticles are exposed to a pulse of electrical current, a sound is created by piezoelectric nanoparticles inside the body (called an electroacoustic sound) that can be recorded with a transducer, using an array of the ultrasound receivers or a single moving transducer that can move and record the produces sounds from the piezoelectric nanoparticle transmitted to a processor or computer to 2-3D electroacoustic images or live video, and then the signal is amplified and forwarded to a processor in order to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram of the structure.
  • PFC perfluorocarbon
  • antibody coated piezoelectric nanoparticles pyroelectric nanoparticles, or perfluorocarbon (PFC) nanoemulsions are injected inside the body, and the nanoparticles attach to the surface antigen of the normal cells or tumor cells and, when exposed to a pulse of electrical current, a sound is created by piezoelectric nanoparticles inside the body (call an electroacoustic sound) that can be recorded with a transducer, and then the signal is amplified and forwarded to a processor in order to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram of the structure using a multi- view Hilbert transformation method to recover the unipolar initial pressure for full-ring electroacoustic computed tomography.
  • PFC perfluorocarbon
  • the antibody coated perfluorocarbon (PFC) nanoemulsions, or piezoelectric or pyroelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, liposomes, liposomes filled with fluorescein or polymer micelles or nanoparticles usually poly(N-isopropylacrylamide) carrying a dye quenched with fluorescein, fluorescent dextrans or another dye or indicator, which is able to be released when the nanoparticles are exposed to electromagnetic radiation, microwave, radiofrequency, ultrasound, or an alternating magnetic field to raise the temperature of piezoelectric or pyroelectric
  • thermosensitive polymers such as chitosan, liposomes, liposomes filled with fluorescein or polymer micelles or nanoparticles usually poly(N-isopropylacrylamide) carrying a dye quenched with fluorescein, fluorescent dextrans or another dye or indicator, which is able to be released when the nanoparticles are exposed to electromagnetic radiation, microwave, radiofrequency,
  • the piezoelectric or pyroelectric nanoparticles are injected inside the body, and attach to the surface antigens of normal cells or the surface antigens of tumor cells and, when exposed to a pulse of electrical current with an adjustable signal frequency and voltage, an acoustic response is produced by the electrical stimulation of piezoelectric nanoparticles inside the body (called an electroacoustic sound) that can be recorded using an array of the ultrasound receivers or a moving transducer that can move and record the sounds from the piezoelectric nanoparticles transmitted to a processor or computer to 2-3D electroacoustic images or live video or the signal is amplified and forwarded to a processor in order to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram, where the system is equipped with a high frequency linear transducer array for mapping the microvascular network of, e.g., the brain, lung, breast, genitourinary tract, digestive tract, and studying its
  • thermal and electrical energy is used to influence the permeability of the tumor cell membrane so as to utilize the effect or thermotherapy to enhance the effect of chemotherapy or immune therapy, gene therapy, gene modification using CRISPR-cas9 in benign and malignant cells, such as brain and spinal cord tumors, breast cancer, lung cancer, prostate and ovarian cancer and melanoma, glioblastoma, retinoblastoma, meduloblastoma, gastrointestinal and genitourinary tumor of sarcoma, etc.
  • Nanoparticles are used having pyroelectric or piezoelectric characteristics, and the antibody coated nanoparticle tube complex is imaged with an electroacoustic computed tomography imaging system.
  • the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, perfluorocarbon (PFC) nanoemulsions, and piezoelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N- isopropyl acrylamide) carrying a dye or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC and conjugated with a medication and/or gene, with cell penetrating agents or activatable cell penetrating agents, and with quenched fluorescein or another dye or indicator, then injected in the patient' s circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N- isopropyl acrylamide) carrying
  • the piezoelectric or pyroelectric nanoparticles When the nanoparticles are exposed to electromagnetic radiation, microwaves, radiofrequency, ultrasound, or an alternating magnetic field the temperature of the piezoelectric or pyroelectric nanoparticles and the tumor cell complex is raised to 41-43 degrees C to release the quenched fluorescein or another dye or indicator.
  • the piezoelectric or pyroelectric nanoparticles When injected into the body, the piezoelectric or pyroelectric nanoparticles attach to the surface antigens of normal cells or the surface antigens of tumor cells and, when exposed to a pulse of electrical current with an adjustable signal frequency and voltage, an acoustic response is produced by electrical stimulation of the piezoelectric or pyroelectric
  • nanoparticles inside the body i.e., called an electroacoustic sound
  • an electroacoustic sound that can be recorded by an array of transducers or a single moving transducer, and then the signal is amplified and forwarded to a processor in order to be converted to a ID, 2D, or 3D or live real time video of a stable lesion or pulsating lesion as an electroacoustic computed tomogram using the movement of the piezoelectric nanoparticles to be evaluated by the doppler of the blood vessels for blood flow measurement.
  • focused compressive ultrasound at low power , 1-40 kHz is used with pluralities of antibody coated nanoparticles conjugated with a medication and/or gene, and conjugated with cell penetrating agents or activatable cell penetrating agents and quenched fluorescein liposomes, liposome-filled with fluorescein or, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye or an indicator injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • the dye, gene, and/or medication is merely peeled off the nanoparticles by the focused vibrational force of the focused low power ultrasound in a non-thermal mode ( LIFU), and this minimally-invasive procedure can be repeated many times as needed without damaging the normal tissue for localized controlled gene or drug delivery in a precise location using the ultrasonic imaging technology.
  • LIFU non-thermal mode
  • the energy source vibrates the pluralities of antibody/ medication coated ferroelectric nanoparticles, piezoelectric nanotubes, or pyroelectric nanoparticles conjugated with a medication and/or gene, and quenched fluorescein, liposomes filled with fluorescein or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying another dye or indicator etc., or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC using pulses of electrical current with an adjustable signal frequency and voltage that create an acoustic response by the electrically stimulated ferroelectric nanoparticles, piezoelectric nanotubes, or pyroelectric nanoparticles inside the body, which are recorded by one or multiple transducers located in different parts of the body.
  • the signal is then amplified and forwarded to a processor in order to be converted to a ID, 2D, or 3D image by electroacoustic computed tomography.
  • the lesion or lesions to which the piezoelectric nanoparticles and other nanoparticles are attached are recognized by electroacoustic computed tomography, and are radiated with pulses of nonthermal focused ultrasound low intensity focused ultrasound ( LIFU), thereby releasing the dye, medication, gene etc.
  • LIFU nonthermal focused ultrasound low intensity focused ultrasound
  • the non-thermal focused high power ultrasound energy source vibrates the antibody/medication coated magnetic nanoparticles conjugated with a medication and/or gene, and quenched fluorescein or another dye or indicator etc. using an alternating magnetic field without producing a significant thermal response, thus releasing the dye, medication, gene, etc. by vibrational force created in the magnetic nanoparticle, and the release of dye/ medication is verified by the release of the fluorescein in the circulation.
  • the antibody/medication coated pluralities of gold, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc. and conjugated with a medication, and with cell penetrating agents or an activatable cell penetrating agent and quenched fluorescein, and are injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc. and conjugated with a medication, and with cell penetrating agents or an
  • the piezoelectric nanoparticle are exposed to a pulse of electrical current thereby creating an electroacoustic sound from the piezoelectric nanoparticles that can be recorded with one or multiple transducers. Then, the signal is amplified and forwarded to a processor so as to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram.
  • Focused high power ultrasound energy is used in a thermal mode at a frequency of generally about 2 MHz to 50 MHz or 100 MHz and more, and at a power of more than one Watt, to raise the temperature of nanoparticles/tumor cells complex to 41-43 degrees C, thus releasing the fluorescein and the medication, which is alternated with focused low power ultrasound in a compressive mode with a frequency of 1-140 kHz and a power of less than 1 Watt, or more than 1 Watt, under the control of a processor controlling the thermal energy intensity and duration of the ultrasound, while imaging the lesion with the ultrasound and electroacoustic computerized tomography or in combination with another imaging modality such as MRI or CT-scan or PET-scan that defines the focal point of the focused low power compressive ultrasound in relationship with the other body's structures.
  • another imaging modality such as MRI or CT-scan or PET-scan that defines the focal point of the focused low power compressive ultrasound in relationship with the other body's structures.
  • the antibody coated pluralities of gold, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles are coated thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc., and conjugated with a medication and/or gene, and conjugated with cell penetrating agents or activatable cell penetrating agents and quenched fluorescein injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc., and conjugated with a medication and/or gene, and conjugated with cell pe
  • the piezoelectric nanoparticles are exposed to pulses of electrical current with an adjustable signal frequency and voltage, thereby creating an electroacoustic sound from the piezoelectric nanoparticles that can be recorded with one or multiple array or a single moving transducers.
  • the signal is then amplified and forwarded to a processor to be converted to a ID, 2D stereoscopic image, or 3D image or video as an electroacoustic computed tomogram, using focused high power ultrasound energy in a thermal mode simultaneously to raise the temperature of nanoparticles/tumor cells complex to 41-43 degrees C, or more thereby releasing the fluorescein and the medication using focused ultrasound energy in the thermal mode under the control of a processor controlling the thermal energy intensity and duration of the ultrasound while imaging the lesion with the ultrasound electroacoustic computed tomography or using harmonic back scatter ultrasonic waves that can be seen simultaneously during the thermal heating of the tissue.
  • a robotic arm may be used to heat the tissue or the tumor at the focal point of the focused ultrasound, thus producing a 2 to 3 dimensional image of the ultrasound, while the thermal energy is controlled automatically by the software based on the thermal or temperature of the image to the ultrasonic system to generate more or less energy while the focal point under the observation of the 2-3 D imaging system heats up the entire tumor in a 50 micron by 50 micron size focal point, thus releasing the medication, gene, etc. from the antibody coated nanoparticles/medication injected in the circulation throughout the tumor.
  • non-thermal low power ultrasound (LIFU) and therapy is combined with MRI, thereby eliminating cavity formation of the high power high-intensity focused ultrasound (HIFU) and potential scar and bubble formation with HIFU in the path of the ultrasound, and reducing the time of the ultrasound surgery with HIFU and its complications.
  • LIFU high-intensity focused ultrasound
  • the antibody coated pluralities of gold, iron oxide, silica, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and piezoelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc.
  • the signal is then amplified and forwarded to a processor so as to be converted to a ID, 2D, or 3D image or video as an electroacoustic computed tomogram, using simultaneously focused high power ultrasound energy in a thermal mode (HIFU) to raise the temperature of nanoparticles/tumor cells complex to 41-43 degrees C, or more thereby releasing the fluorescein and the medication using focused ultrasound energy in the thermal mode under the control of a processor controlling the thermal energy intensity and duration of the ultrasound, while imaging the lesion with the ultrasound and electroacoustic computerized tomography or videography.
  • HIFU thermal mode
  • the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator etc.
  • bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC conjugated with a medication, and conjugated with cell penetrating agents or activatable cell penetrating agents and quenched fluorescein, and injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • the dye is released when the nanoparticles are exposed to low power focused ultrasound energy in a non-thermal compressive mode (LIFU)1-140 kHz and 0.1 Watt to vibrate the piezoelectric, pyroelectric nanoparticles/tumor cells complex that are injected inside the body and are attached to the surface antigen of normal cells or of tumor cells, and when exposed to pulses of electrical current with an adjustable signal frequency and voltage, an electroacoustic sound is created inside the body from the piezoelectric nanoparticles that can be recorded with a transducer.
  • LIFU non-thermal compressive mode
  • the signal is then amplified and forwarded to a processor to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram and ultrasound image of the structure, and the dye/ medication is released under observation of the lesion as video-electroacoustic imaging.
  • antibody/medication coated gold, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles are conjugated with a medication and checkpoint inhibitors and immune stimulators such as VLP, TLR3,TLR7, TLR8 or CD40 and quenched fluorescein or another dye or indicator, etc. so that the medications are released using high power focused ultrasound energy in a thermal mode alternating with focused ultrasound in a non-thermal compressive mode at 1-140 kHz to one MHz or more, and less than 1 Watt power (LIFU) while antibody coated
  • a medication and checkpoint inhibitors and immune stimulators such as VLP, TLR3,TLR7, TLR8 or CD40 and quenched fluorescein or another dye or indicator, etc.
  • nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), and the tumor cells are recognized by the T-cells, which together with killer cells phagocytose them, and enhance the immune response to the tumor and its exosomes and the circulating cells.
  • the T-cells destroy the tumor cells, circulating tumor cells, and their exosomes as a result of the VLPs and checkpoint inhibitors that are attached to the tumor cells, circulating tumor cells, and their exosomes.
  • the VLPs make the tumor cells, circulating tumor cells, and their exosomes visible to the T-cells, which destroy them.
  • antibody/medication coated gold, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles that contain at least one radioactive alpha, or beta emitter conjugated with medication and checkpoint inhibitors , VLP , TLR3,TLR7, TLR8 or CD40 etc and quenched fluorescein or another dye or indicator, etc. are exposed to pulses of electrical current with an adjustable signal frequency and voltage so that a electroacoustic sound is created inside the body from the piezoelectric nanoparticles that can be recorded with a transducer.
  • the signal is then amplified and forwarded to a processor so as to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram and ultrasound to release the dye/medication, gene using focused ultrasound energy in a thermal mode at a temperature of 41-43 degrees C alternating with focused ultrasound in a compressive mode at low power 1-140 or more kHz under the control of a processor controlling the thermal energy intensity and duration of the ultrasound while imaging the lesion with the ultrasound by software controlling the thermal energy delivery based on the temperature imaging system, while antibody coated
  • nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which carry PD-L1 (to disguise themselves), and the tumor cells are recognized by the T-cells, which together with killer cells phagocytose them, and enhance the immune response to the tumor and its exosomes and the circulating cells and eliminate them.
  • antibody/medication coated gold, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles are conjugated with medication and checkpoint inhibitors, VLP , TLR3,TLR7, TLR8 or CD40 etc. and quenched fluorescein to be released at a temperature of 41-43 degrees C using focused ultrasound energy in a thermal mode alternating with focused low power ultrasound in a compressive mode at 1-140 kHz or more as many times as needed after initial treatment to damage the remaining tumor cells or recurrences with simultaneous vaccination while imaging the lesion with the ultrasound/electroacoustic tomography and initiate an immune response to the tumor cells circulating cells and their exosomes.
  • the energy source heats the antibody/medication coated pyroelectric or piezoelectric nanoparticles conjugated with medication and checkpoint inhibitors , VLP etc and quenched fluorescein or another dye or indicator, etc. to be released at temperature of 41-43 degrees C for the desired time using focused ultrasonic energy in a thermal mode under the control of a processor controlling the intensity of the focused ultrasound for a desired duration, while antibody coated nanoparticles/checkpoint inhibitors and VLP , TLR3,TLR7, TLR8 or CD40 attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), and the tumor cells are recognized by the T-cells, which together with killer cells phagocytose them, and enhance the immune response to the tumor and its exosomes and the circulating cells.
  • a checkpoint protein such as PD-L1 (to disguise themselves
  • the focused high power ultrasound energy source at 0.5-3MHz heats the antibody/medication coated gold, iron oxide, nanocage, nanotube, nanoshell, piezoelectric nanoparticles using focused ultrasound so as to heat the tumor cell nanoparticle complex conjugated with quenched fluorescein or another dye or indicator etc., and VLP, CD40 or TLR3,TLR7 and apply a pulse of electric current to induce a sound from the piezoelectric nanoparticles, image the lesion and to release fluorescein and medication at 41- 43 degrees C, while maintaining the intensity, duration of the ultrasound energy or reducing its intensity to a desired level, and for a desired time, obtaining blood samples to evaluate increased biomarkers in the circulation and produce a vaccine with viral-like particles (VLPs) and TLR3,TLR7, TLR8 or CD40 and administering them with checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc. and Rock inhibitors, such as Fasudil
  • TLR3,TLR7, TLR8 or CD40 attach to the potentially localized or residual tumor or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), and the tumor cells are recognized by the T-cells, which together with killer cells phagocytose them, and enhance the immune response to the tumor and its exosomes and the circulating cells.
  • a checkpoint protein such as PD-L1 (to disguise themselves)
  • the focused high power ultrasound energy source heats the antibody/medication coated gold, iron oxide, nanocage, nanotube, nanoshell, or piezoelectric nanoparticles using focused ultrasound so as to heat the tumor cell nanoparticle complex conjugated with quenched fluorescein or another dye or indicator etc., and VLP, CD40 or TLR3,TLR7 and a pulse of electric current is applied to induce a sound from the
  • piezoelectric nanoparticles image the lesion and to release fluorescein and medication at 41- 43 degrees C, while maintaining the intensity, duration of the ultrasound energy or reducing its intensity to a desired level, and for a desired time, obtaining blood samples to evaluate increased biomarkers in the circulation and produce a vaccine with viral-like particles (VLPs), or oncolytic viruses, T-Vec and administering them with thermotherapy and releasing them at the temperature or 41-43 degrees C that also damages or kills the VLP and oncolytic viruses such as T-Vec while leaving their antigenic foreign proteins in the tumor site to enhance an immune response, while releasing checkpoint inhibitors, such as PD-1, PD- 11, CTLA-4, Jagged 1 inhibitor 15D11, while antibody coated nanoparticles/checkpoint inhibitors and VLP , CD40 or TLR3,TLR7 attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), and the tumor cells are recognized by
  • antibody coated iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles are conjugated with medication, checkpoint inhibitors, and quenched fluorescein or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC to be released using focused ultrasonic in a compressive mode at 1-40 kHz where the medications are from the group consisting of Wnt inhibitors, rock inhibitors, metformin, anti-VEGFs, Doxorubicin, cyclophosphamide, antibiotics, anti-inflammatory, mammalian target of rapamycin, etc.
  • glucose metabolism activates the Ras signaling that regulates the cell proliferation, along or independently from the Wnt activation and glucose triggers activation of MEK and ERK and inhibition of glucose metabolism at the tumor cell level with metformin, etc. delivered with antibody coated nanoparticles of iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles conjugated with a medication selected from the group consisting of Wnt inhibitors, Rock inhibitors, metformin, buformin, and syrosingopine, phenformin, anti-VEGFs, and checkpoint inhibitors, VLP, IL-2, or bee toxins etc., enzymes such as Matrix
  • metalloproteinases as immune stimulators and quenched fluorescein or another dye or indicator, etc. are released using focused ultrasonic energy in a compressive mode or a thermal mode at 50-150-kHz where the medications/dye are released affecting the membranes of the cancer cells and their metabolism more significantly than the normal surrounding normal cells while affecting the membranes of the cancer cells and their metabolism more significantly than the normal surrounding normal cells and release of VLP, CD40 or TLR3,TLR7 etc. as immune stimulators and checkpoint inhibitors enhance the immune response to the tumor locally and also their invisible metastatic lesions and their exosomes carrying the same tumor receptors and PD-L1 on their membranes.
  • glucose metabolism activates the Ras signaling that regulates the cell proliferation, along or independently from the Wnt activation and glucose triggers activation of MEK and ERK and inhibition of glucose metabolism at the tumor cell level with metformin, etc. delivered with antibody coated nanoparticles of iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric .
  • a medication selected from the group consisting
  • thermosensitive polymer coating of the nanoparticles further comprises buformin or phenformin so as to inhibit glucose metabolism in the cells of the tumor, and inhibit glucose metabolism at the tumor cell level with buformin or phenformin etc. delivered with antibody coated nanoparticles of iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles conjugated with medication selected from the group consisting of Wnt inhibitors, Rock inhibitors, buformin, phenformin, anti-VEGFs and checkpoint inhibitors, and quenched fluorescein or another dye or indicator, etc.
  • antibody/medication coated gold, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and piezoelectric nanoparticles are conjugated with medication and checkpoint inhibitors, VLP, or immune stimulators and quenched fluorescein or another dye or indicator, etc. to be released using focused ultrasound in a thermal mode alternating with focused compressive ultrasound in a non-thermal mode.
  • the functionalized piezoelectric nanoparticles are injected in a patient suspected of a malignancy under a pulse of electrical current creating an
  • electroacoustic sound from the piezoelectric nanoparticles that can be recorded with one or multiple transducers.
  • the signal is then amplified and forwarded to a processor so as to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram verifying the presence or absence of a suspected tumor or to differentiate a benign lesion from a malignant lesion to which the pluralities of nanoparticles are attached and produce the electroacoustic computerized tomography.
  • the antibody piezoelectric nanoparticles are injected to reveal the existent of a lesion or tumor under a pulse ofelectrical current that produces an electroacoustic sound wave that can be captured and amplified, and converted to a signal producing an image as an electroacoustic computed image.
  • the antibody/medication piezoelectric nanoparticles, pyroelectric, or perovskites nanoparticles are re-injected to reveal the existent of a lesion or tumor after the tumor has been treated previously under pulses of electrical current with an adjustable signal frequency and voltage that produces an electroacoustic sound wave from the piezoelectric nanoparticles or pyroelectric or perovskites that can be captured, amplified, and converted to a signal producing an image or as an electroacoustic computed tomogram indicating either persistence of the tumor cells, recurrences, or metastatic lesion or the tumor while electric pulses of 30-100 Hz and more paralyses the tumor cells by depolarizing the membrane potential and making their cell membrane accessible to medication.
  • the antibody piezoelectric nanoparticles, pyroelectric or perovskites, Boron tubes, etc., iron oxide nanoparticles or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of perfluorocarbon (PFCL) liquid are conjugated with monoclonal antibodies, checkpoint inhibitors, VLP, and IL-2, etc., Wnt inhibitors, Rock inhibitors, genes, or gene inhibitors, and are injected to reveal the existent of a lesion or tumor after the tumor has been treated previously under pulses of electrical current with an adjustable signal frequency and voltage that produces an electroacoustic sound wave from the piezoelectric nanoparticles or pyroelectric or perovskites that can be captured, amplified, and converted to a signal producing an image or as an electroacoustic computed tomogram indicating either persistence of the tumor cells, recurrences, or a metastatic lesion or tumor, and simultaneously treated with non-thermal or thermal focused low power ultrasound,
  • PFCL perflu
  • the antibody coated pluralities of gold, iron oxide, silica, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carry a dye quenched with fluorescein or another dye or indicator, etc. conjugated with a medication and/or gene, and conjugated with Wnt inhibitors or rock inhibitors and with cell penetrating agents or activatable cell penetrating agents and quenched fluorescein or another dye or indicator etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carry a dye quenched with fluorescein or another dye or indicator, etc. conjugated with a medication and/or gene, and conjugated with Wnt inhibitors or rock inhibitors
  • the piezoelectric nanoparticles are exposed to a pulse of electrical current, thereby creating an electroacoustic sound from the piezoelectric nanoparticles that can be recorded with a transducer.
  • the transducer signal is then amplified and forwarded to a processor so as to be converted to a ID, 2D, or 3D image using focused low power ultrasound energy in a non-thermal mode, thus releasing the fluorescein and the medication and/or gene etc.
  • a processor controlling the thermal ultrasonic intensity and duration of the ultrasound, while imaging the lesion with the ultrasound along with electroacoustic 2-D or 3- D images as an electroacoustic computed tomogram or in combination with another imaging modality such as MRI or CT-scan or PET-scan that defines the focal point of the focused compressive ultrasound in relationship with the other body's structures.
  • iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric antibody/medication coated nanoparticles contain at least one radioactive agent conjugated with medication and checkpoint inhibitors Botulinum toxin, VLP, etc.
  • Rock inhibitors such as Fasudil, netarsudil, and quenched fluorescein or another dye or indicator, etc.
  • iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric antibody coated nanoparticles are conjugated with a Wnt inhibitor, Rock inhibitors, such as Fasudil, etc., antineoplastic medication, checkpoint inhibitors, immune stimulators, and quenched fluorescein or another dye or indicator, etc.
  • the repeated ultrasound pulses generate an electric pulse from the piezoelectric nanoparticles that damages the tumor cell by depolarizing their cell membrane and initiate an immune response to the tumor cells, circulating cells, and their exosomes.
  • iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and piezoelectric antibody/medication coated nanoparticles are conjugated with a Rock inhibitor, such as Fasudil, and its derivatives etc., antineoplastic medication, checkpoint inhibitors, immune stimulators, e.g., VLP, and quenched fluorescein or another dye or indicator, etc. to be released using high power focused ultrasound in a thermal mode alternating with a low power focused compressive ultrasound mode and initiate an immune response to the tumor cells, circulating cells, and their exosomes.
  • a Rock inhibitor such as Fasudil, and its derivatives etc.
  • antineoplastic medication such as Fasudil, and its derivatives etc.
  • checkpoint inhibitors such as antineoplastic medication
  • immune stimulators e.g., VLP, and quenched fluorescein or another dye or indicator, etc.
  • iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and/or piezoelectric antibody coated nanoparticles are conjugated with a rock inhibitor, antineoplastic medication, RNAi, siRNA, and checkpoint inhibitors, Rock inhibitors, such as Fasudil, etc., and quenched fluorescein or another dye or indicator, etc., and are exposed to pulses of electrical current with an adjustable signal frequency and voltage so that an electroacoustic sound is created inside the body from the piezoelectric nanoparticles that can be recorded with a transducer.
  • the transducer signal is then amplified and forwarded to a processor in order to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram, and to release the medication using focused high power ultrasound in a thermal mode (e.g., at a frequency of 50-1 MHz) alternating with a focused compressive low power ultrasound mode and the repeated low power ultrasound pulses generate an electric pulse from the piezoelectric nanoparticles that damages the tumor cell by depolarizing their cell membrane.
  • a thermal mode e.g., at a frequency of 50-1 MHz
  • pluralities of iron oxide, gold, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and piezoelectric antibody coated nanoparticles are conjugated with rock inhibitors, an antineoplastic medication, genes, CRISPR-cas9, checkpoint inhibitors, Rock inhibitors such as Fasudil, etc. and quenched fluorescein or another dye or indicator, etc., and then exposed to pulses of electrical current with an adjustable signal frequency and voltage so that a electroacoustic sound is created inside the body from the piezoelectric nanoparticles that can be recorded with a transducer.
  • the transducer signal is amplified and forwarded to a processor so as to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram, and the gene, CRISPR-cas9, or medication is alone or in combination, released using focused low power compressive ultrasound to strip the coatings off the nanoparticles by vibrational forces of the non-thermal mode of ultrasound and to modify the gene using CRISPR-cas9 mediated Homology - Independent Targeted Integration (HITI) or Homology Directed Repair (HDR).
  • HITI Homology - Independent Targeted Integration
  • HDR Homology Directed Repair
  • iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and piezoelectric antibody coated nanoparticles conjugated with a rock inhibitor, antineoplastic medication, CRISPR-cas9, a corrective gene, and/or quenched fluorescein or another dye or indicator, etc. are exposed to pulses of electrical current with an adjustable signal frequency and voltage so that an electroacoustic sound is created inside the body from the piezoelectric nanoparticles that can be recorded with a transducer.
  • the transducer signal is then amplified and forwarded to a processor to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram, and the nanoparticle coating is released using focused low power ultrasound in a compressive ultrasound mode so as to modify the gene using CRISPR-cas9 mediated Homology -Independent Targeted Integration (HITI) or Homology Directed Repair (HDR) while imaging the lesion.
  • HITI Homology -Independent Targeted Integration
  • HDR Homology Directed Repair
  • pluralities of nanoparticles, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and/or piezoelectric antibody coated nanoparticles are conjugated with a rock inhibitor, antineoplastic medication, CRISPR-cas9 with a gene, and with cell penetrating agents or activatable cell penetrating agents, and quenched fluorescein or another dye or indicator, etc.
  • the transducer of a focused low power compressive, or thermal high power ultrasound is positioned on the surface of the body to aim the focal point of the ultrasound at specific part of the lesion now made visible by electroacoustic imaging technology, such as its center or the borders of the lesion, or the focal point can move so that the entire lesion is treated with low power compressive or low thermal focused ultrasound providing thermal energy to heat antibody/medication coated nanoparticles and its thermosensitive polymers carrying a dye or gene, checkpoint inhibitors, such as Fasudil, and its derivatives etc., VLP or oncolytic viruses, such as T-Vec , monoclonal antibodies, IL-2, bee toxins, immune stimulators along with checkpoint inhibitors, medication are released and damage already heated tumor cells at a temperature of 41-43 degrees C and release dye and medication from the nanoparticles indicating the temperature of 41-43 degrees C has been achieved which can be controlled by the processor or computer controlling energy delivery, under direct imaging the tumor ,or the lesion or modify
  • the transducer of a low power focused compressive nonthermal , or high power thermal ultrasound is positioned on the surface of the body to aim the focal point of the ultrasound at a specific part of the lesion now made visible by
  • electroacoustic imaging technology such as its center or the borders of the lesion or the focal point of the none-thermal focused ultrasound connected to a movable arm can be moved on the body surface under the control of a processor or computer that show on it monitor both the location of the lesion and the images of the focused low power ultrasound simultaneously and a software controls the areas that are being treated in 3-D manner or as a live video so that the entire lesion is treated with compressive or low thermal focused, low power ultrasound providing minimal thermal energy to heat antibody/medication coated nanoparticles and its polymeric coating carrying a dye, medication or gene, checkpoint inhibitors, Rock inhibitors, such as Fasudil, etc., VLP or oncolytic viruses, such as T-Vec , or monoclonal antibodies, bee or scorpion toxin, metalloproteinases as immune stimulators, antineoplastic medication are released and damage already heated tumor cells at temperature or 41-43 degrees C and release dye and medication from the nanoparticles indicating the temperature of 41-43 degrees C has been achieved which
  • HITI Homology -Independent Targeted Integration
  • HDR Homology Directed Repair
  • the antibody coated nanoparticles, nanospheres, liposomes, nanowires, nanorods, nanocages, nanoshells, magnetic, ferromagnetic, piezoelectric, pyroelectric, nanotubes, zinc oxide, barium titanate, iron oxide, gold, gold silica, strontium titanate, perovskites, and polyvinylidene fluoride, boron nitride nanotubes or a combination thereof are coated with a thermosensitive polymer, such as chitosan, polysaccharides, (e.g., glycol chitosan, poly-L-Lysine (PLL), polyethylene imine (PEI), polylactic, polyglycolic, polyaspartic acid or copolymers), a cationic polymer, such as poly lysine and polyethylene imine, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye and conjugated
  • LIFU focused nonthermal ultrasound
  • CRISPR-cas9 mediated Homology -Independent Targeted Integration (HITI) or Homology Directed Repair (HDR) under observation by an electroacoustic computed tomogram and) and induce an immune response by released of VLP, etc.
  • stimulators and checkpoint inhibitors enhance the immune response to the tumor locally and also their invisible metastatic lesions elsewhere and initiate an immune response to the tumor cells, circulating cells, and their exosomes and eliminate them.
  • the nanoparticles are selected from a group consisting of iron oxide gold nanoparticles, gold graphene oxide nanoparticles, gold nanoparticles, silicone nanoparticles, carbon nanoparticles, magnetic nanoparticles, gold nanorods, gold nanotubes, gold nanoshells, gold nanocages, iron oxide, gold, boron nitride, and carbon nanotubes and combinations thereof that are under observation by an electroacoustic computed tomogram.
  • the nanoparticles are selected from a group consisting of iron oxide gold nanoparticles, gold graphene oxide nanoparticles, gold nanoparticles, silicone nanoparticles, carbon nanoparticles, magnetic nanoparticles, gold nanorods, gold nanotubes, gold nanoshells, gold nanocages, iron oxide, gold, boron nitride, and carbon nanotubes and combinations thereof conjugated with the photosensitizer verteporfin, and the
  • antibody/medication coated nanoparticles contain at least one radioactive compound conjugated with medication and checkpoint inhibitors, Rock inhibitors, such as Fasudil, etc., and quenched fluorescein or another dye or indicator etc. to be released using focused ultrasonic in a thermal mode alternating with a non-thermal focused compressive or pressure ultrasound mode.
  • the pluralities of antibody coated nanoparticles, nanospheres, liposomes, nanowires, nanorods, nanocages, nanoshells, nanotubes; magnetic, ferromagnetic, piezoelectric, pyroelectric nanotubes; zinc oxide, barium titanate, iron oxide, gold, gold silica, strontium titanate, perovskites, and polyvinylidene fluoride boron nitride nanotubes or a combination thereof are coated with a thermosensitive polymer, such as chitosan, polysaccharides, e.g., glycol chitosan, poly-L-Lysine (PLL), polyethylene imine (PEI), polylactic, polyglycolic, polyaspartic acid or copolymers, a cationic polymer such as polylysine and polyethylene imine, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye
  • the pluralities of antibody/medication coated nanoparticles, nanospheres, liposomes, nanowires, nanorods, nanocages, nanoshells, nanotubes; magnetic, ferromagnetic, piezoelectric, or pyroelectric nanotubes; zinc oxide, barium titanate, iron oxide, gold, gold silica, strontium titanate, perovskites, and polyvinylidene fluoride boron nitride nanotubes or a combination thereof are coated with polymer, such as chitosan, polysaccharides, e.g., glycol chitosan, poly-L-Lysine (PLL), polyethylene imine (PEI), polylactic, polyglycolic, polyaspartic acid or copolymers, a cationic polymer such as polylysine and polyethylene imine, polyethylene glycol, biotin streptavidin are injected with cell penetrating agents or activatable cell penetrating agents and
  • polymer such as
  • lymphatic vessels inside a body cavity, in cerebrospinal fluid, in the eye, in the bladder, uterus in the peritoneal cavity and stimulated with pulses of electrical current with an adjustable signal frequency and voltage at predetermined intervals at different places in the body while listening to production of a sound wave which has the same frequency as the frequency of the pulses of electrical current with the adjustable signal frequency and voltage applied to the tissue indicating the presence of a suspected lesion by the production of a sound wave from the piezoelectric nanoparticle/tumor or exosomes and counting the circulating cells or the exosomes as they pass inside the electrical current to confirm non-invasively the presence of the extracellular vesicle, circulating tumor cells, etc., before or after treatment to find out soon within a week if the medication or the therapy has had an effect on the tumor and its circulating exosomes or metastatic cell numbers measured before and after the treatment or recognizing existing live cells inside a tumor after the therapy, such as in breast cancer, glioblast
  • polymers such as chi
  • the medication or the therapy has had an effect on the tumor and its circulating exosomes or metastatic cell numbers measured before and after the treatment or recognizing existing live cells inside a tumor after the therapy, such as in breast cancer, glioblastoma, uterine cancer, ovarian cancer, prostate cancer, lung cancer, etc.
  • a polymer such as
  • the abovementioned electroacoustic computed tomography may be made as a tube, half tube, partial tube like MRI, or CT-scan or PET-scan units in which the patient can lay down and the table moves the patient in or out of the system while multiple rows or electrical connection is made like an electrocardiogram recording electrode, but in this case, they represent anode or cathodes arrays of the system so that the entire patient body may be scanned or imaged using a computer controlled recording, and electroacoustic tomography is done in a less expensive and time consuming way than is needed for an MRI with its specific room or CT-scan or PET-scan, but without exposing the patient to radiation.
  • thermotherapy with focused low power ultrasound or alternating magnetic field is combined with the antibody/medication coated piezoelectric, pyroelectric nano tubes, nanocages, nanoshells may be combined with other
  • the dendrimer poly(amidoamine) can be functionalized to be biocompatible and cell penetrating.
  • Other dendrimers are poly(amidoamine-organosilicon) (PAMAMOS), poly(propyleneimine) (PPIO), tecto, multilingual, chiral, hybrid, amphiphilic, micellar, multiple antigen peptide, and Frechet-type dendrimers.
  • Dendrimers may be used as carriers of inhibitory genes to correct muted gene or elimination of the mutated gene(s) using CRISPR-cas9 mediated Homology -Independent Targeted Integration (HITI) or Homology Directed Repair (HDR) under observation with electroacoustic tomography to precisely locate the lesion for gene delivery.
  • HITI Homology -Independent Targeted Integration
  • HDR Homology Directed Repair
  • thermotherapy with focused low power ultrasound or alternating magnetic field is combined with the pluralities of antibody/medication coated nanoparticles, nanospheres, liposomes, nanowires, nanorods, nanocages, nanoshells, magnetic, non-magnetic, paramagnetic, pyroelectric, piezoelectric, nanotubes; magnetic, ferromagnetic, piezoelectric, pyroelectric, nanotubes; zinc oxide, barium titanate, iron oxide, gold, gold silica, strontium titanate, perovskites, and polyvinylidene fluoride boron nitride nanotubes and/or combinations thereof are coated with polymers, such as chitosan, polysaccharides, e.g., glycol, chitosan, poly-L-Lysine (PLL), polyethylene imine (PEI), polylactic, polyglycolic, polyaspartic acid or copolymers, phospholipid microbubbles containing a harmless gas
  • cell penetrating agents or activatable cell penetrating agents are injected with cell penetrating agents or activatable cell penetrating agents in the patient's circulation, lymphatic vessels, inside a body cavity, in cerebrospinal fluid, in the eye, in the bladder, in the uterus, in the peritoneal cavity etc.
  • Blood may also be obtained to confirm an increase in biomarkers in the blood and use the biomarkers to build an anti-tumor vaccine with or without VLP, IL-2 , toxins and administering them with checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide while antibody coated nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), are are recognized by the T-cells, which together with killer cells phagocytose them and enhance immune response to the tumor and its exosomes and the circulating cells, and the Rock inhibitor reduces TGF- ⁇ production after therapy and the subsequent scar formation.
  • checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fa
  • the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric, and/or piezoelectric nanoparticles, etc. are coated thermosensitive polymers, such as chitosan, quenched with fluorescein or another dye or indicator/medication etc. and conjugated with an antineoplastic medication, gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • the functionalized nanoparticles are exposed to electromagnetic radiation, microwaves, or radiofrequency radiation, or focused high power ultrasound 44 (see FIG.
  • an acoustic response is produced by electrical stimulation of piezoelectric nanoparticles inside the body 38 that is called electroacoustic sounds or signals which can be captured with a transducer (e.g., ultrasound transducer 40 in FIG. 2), or microphone, converted to an electrical signal and is forwarded to a processor to be converted to a ID, 2D, or 3D image 42 as an electroacoustic computed tomogram while the electrical pulse generated in the piezoelectric nanoparticles drives the medication, gene, in the tumor cells locally to damage the tumor cells by multiple modes of the therapy applied non-invasively under observation.
  • a transducer e.g., ultrasound transducer 40 in FIG. 2
  • a processor e.g., ultrasound transducer 40 in FIG. 2D, or 3D image 42 as an electroacoustic computed tomogram while the electrical pulse generated in the piezoelectric nanoparticles drives the medication, gene, in the tumor cells locally to damage the tumor cells by multiple modes of the therapy applied non-invasively
  • an electrical source in a patient with a thyroid tumor 50 (see FIG. 3), comprises a battery device 46 with an anode 48 being positioned on a first side of the body of the patient and a cathode 52 being located on a second side of the body of the patient, the pulsed electrical current (as diagrammatically indicated by the current lines in FIG. 3) passing through the body (e.g., the neck) of the patient from the anode 48 to the cathode 52 of the battery device 46, and where the pulsed electrical current passes through the tumor 50 which has been pretreated with antibody coated piezoelectric nanoparticles conjugated with medication, and attached to the tumor cells.
  • the pulsed electrical current as diagrammatically indicated by the current lines in FIG. 3
  • the thyroid gland 56 of the patient which is disposed around the trachea 54, comprises the right lobe 58, the left lobe 62, and the isthmus 60 connecting the right and left lobes 58, 62.
  • Thyroid cartilage 64 is disposed above the thyroid gland 56 in FIG. 3.
  • the battery device 46 is operatively coupled to a controller with software for generating the pulsed electrical current passing through the body of the patient between the anode 48 and the cathode 52.
  • FIG. 4 it can be seen that the pulsed electrical current (as
  • FIG. 4 diagrammatically indicated by the current lines in FIG. 4) generated by the battery device 46 creates an ultrasonic wave 74 from the piezoelectric nanoparticles that can be recorded by an ultrasonic transducer 68 located on the skin.
  • the ultrasonic transducer 68 is connected to a processor and monitor 70, which allows an image 72 of the thyroid tumor 66 in FIG. 4 to be reconstructed from the ultrasonic wave 74 received at the transducer 68.
  • FIG. 5 it can be seen that a thyroid tumor 75 may be treated non- invasively with a focused ultrasound beam 84 generated by an ultrasound array transducer 82.
  • the focused ultrasound beam 84 passes through the skin and simultaneously heats up the tumor tissue and attached piezoelectric nanoparticles to a temperature of 39-40 degrees C to damage the tumor cells of the tumor 75 with the thermal energy and to depolarize the tumor cell membranes by converting the sound waves to an electric pulse to depolarize the tumor cells exposed to the ultrasound, making the cells permeable to the medication/gene used to treat the tumor 75.
  • FIG. 5 it can be seen that the heating of the tumor 75 by the focused ultrasound beam 84 creates harmonic backscatter ultrasonic waves 86 from the piezoelectric nanoparticles that can be recorded by an ultrasonic transducer 68 located on the skin.
  • FIG. 5 similar to FIG.
  • the ultrasonic transducer 68 is connected to a processor and monitor 70, which allows an image 76 of the thyroid tumor 75 to be reconstructed from the harmonic backscatter ultrasonic waves 86 received at the transducer 68.
  • the processor and monitor 70 connected to the transducer 68 are operatively coupled to another processor 78 that executes software for controlling the energy output of the focused ultrasound delivered by the ultrasound array transducer 82. That is, the processor 78 is operatively coupled to the ultrasound power source 80 so as to enable the energy output of the focused ultrasound delivered by ultrasound array transducer 82 to be selectively varied based upon feedback from the harmonic backscatter ultrasonic waves 86 received by the ultrasonic transducer 68. In this manner, in the system of FIG.
  • the temperature at the tumor site is able to be selectively controlled by varying the energy output of the focused ultrasound delivered by ultrasound array transducer 82.
  • the ultrasound when the focused ultrasound is applied to the tumor 75 with piezoelectric nanoparticles attached to the tumor 75, the ultrasound activates the piezoelectric nanoparticles to produce electrons to depolarize the tumor cells, and the focused ultrasound also creates second harmonic sound waves 86 that are recorded by the transducer 68 indicating the temperature at the tumor site.
  • the imaging system 70 in FIG. 5 is connected to the processor 78 which, in turn, is connected to the ultrasound power source 80 so as to control the temperature of the tumor site at 41-43 degrees C or more, as needed.
  • the tumor cells are depolarized and heated simultaneously (i.e., combining cell thermotherapy with cell depolarization) and medication conjugated with the piezoelectric nanoparticles is released at the tumor site so as to damage the tumor cells.
  • the tumor cells have already been rendered generally defenseless by virtue of their depolarization, the medication is able to easily pass through the damaged tumor cell membranes, thus entering the cytoplasms of the damaged tumor cells and destroying the tumor cells.
  • the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles, etc. are coated thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator/medication etc., and conjugated with an antineoplastic medication, gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye quenched with fluorescein or another dye or indicator/medication etc.
  • the functionalized nanoparticles are exposed to microwaves, radiofrequency radiation, and/or electrical current generated by a battery where low electrical current from a battery passes from one side of the skin (i.e., the anode) through the body and a lesion, or tumor to the cathode electrode positioned on the opposite side of the skin on the body to raise the temperature of the piezoelectric or pyroelectric nanoparticles that are injected inside the body to be attached to the surface antigens of the normal cells or of tumor cells and create nanoparticle/tumor cell complexes to 41-43 degrees C, and when exposed to pulses of electrical current with an adjustable signal frequency and voltage, an acoustic response is produced by electrical stimulation of the piezoelectric nanoparticles inside the body that is called electroacoustic sounds or signals which can be captured with a transducer, or microphone, and converted to an electrical signal and is forwarded to a processor to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram
  • the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and piezoelectric nanoparticles, etc. are coated with thermosensitive polymers, such as chitosan, quenched with fluorescein, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye or another dye or indicator/medication etc. conjugated with an antineoplastic medication, gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • thermosensitive polymers such as chitosan, quenched with fluorescein, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye or another dye or indicator/medication etc. conjugated with an antine
  • the functionalized nanoparticles are exposed to microwaves, radiofrequency radiation, or focused ultrasound in a thermal or non-thermal mode or non-focused ultrasound, and/or electrical current generated by a battery where low electrical current from a battery passes from one side of the skin (i.e., the anode) through the body and a lesion or tumor to the cathode electrode positioned on the opposite side of the skin on the body to raise the temperature of the piezoelectric or pyroelectric nanoparticles that are injected inside the body to be attached to the surface antigens of the normal cells or of tumor cells and heat the nanoparticle/tumor cells complexes to 41-43 degrees C, and when exposed to pulses of electrical current with an adjustable signal frequency and voltage, an acoustic response is produced by electrical stimulation of the piezoelectric nanoparticles inside the body that is called electroacoustic sounds or signals which can be captured with a transducer or microphone, and converted to an electrical signal and forwarded to a processor to be converted to a ID, 2D
  • thermotherapy with focused high power ultrasound or alternating magnetic field, etc. is combined with the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles, etc. are coated with thermosensitive polymers, such as chitosan, with quenched fluorescein, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye another dye or indicator/medication, etc.
  • thermosensitive polymers such as chitosan, with quenched fluorescein, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye another dye or indicator/medication, etc.
  • antineoplastic medication gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • the functionalized nanoparticles are exposed to an alternating magnetic field and/or electrical current generated by a battery where low electrical current from a battery passes from one side of the skin (i.e., the anode) through the body and a lesion or tumor to the cathode electrode positioned on the opposite side of the skin on the body to raise the temperature of the piezoelectric or pyroelectric nanoparticles that are injected inside the body to be attached to the surface antigens of the normal cells or of tumor cells and heat the nanoparticle/tumor cell complexes to 41-43 degrees C, and when exposed to pulses of electrical current with an adjustable signal frequency and voltage, an acoustic response is produced by electrical stimulation of piezoelectric nanoparticles inside the body that we call electroacoustic sounds or signals which can be captured with a transducer, or microphone, and converted to an electrical signal and forwarded to a processor to be converted to a ID, 2D, or 3D as an electroacoustic computed tomogram while the electrical pulse generated
  • thermotherapy with focused low power ultrasound or alternating magnetic field, etc. is combined with the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles, etc. are coated with thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye with quenched fluorescein or another dye or indicator/medication, etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye with quenched fluorescein or another dye or indicator/medication, etc.
  • antineoplastic medication gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • the functionalized nanoparticles are exposed to a divergent ultrasound and electrical current generated by a battery where low electrical current from a battery passes from one side of the skin (i.e., the anode) through the body and a lesion, or tumor to the cathode electrode positioned on the opposite side of the skin on the body to raise the temperature of the piezoelectric and/or pyroelectric nanoparticles that are injected inside the body to be attached to the surface antigens of the normal cells or of tumor cells and create nanoparticle/tumor cell complexes to 41-43 degrees C, and when exposed to pulses of electrical current with an adjustable signal frequency and voltage, an acoustic response is produced by electrical stimulation of the piezoelectric nanoparticles inside the body that is called electroacoustic sounds or signals which can be captured with a transducer, or microphone, and converted to an electrical signal and forwarded to a processor to be converted to a ID, 2D, or 3D as an electroacoustic computed tomogram while the electrical pulse
  • thermotherapy with focused low power ultrasound or alternating magnetic field, etc. is combined with the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles, etc. are coated thermosensitive polymers, such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye with quenched fluorescein or another dye or indicator/medication etc.
  • thermosensitive polymers such as chitosan, liposomes, or polymer micelles or nanoparticles usually poly(N-isopropyl acrylamide) carrying a dye with quenched fluorescein or another dye or indicator/medication etc.
  • antineoplastic medication gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • the functionalized nanoparticles are exposed to a non-focused ultrasound and electrical current generated by a battery where low electrical current from a battery passes from one side of the skin (i.e., the anode) through the body and a lesion or tumor to the cathode electrode positioned on the opposite side of the skin on the body to raise the temperature of the piezoelectric or pyroelectric nanoparticles that are injected inside the body to be attached to the surface antigens of the normal cells or of tumor cells and create nanoparticle/tumor cells complexes to 41-43 degrees, and when exposed to pulses of electrical current with an adjustable signal frequency and voltage, an acoustic response is produced by electrical stimulation of the piezoelectric nanoparticle inside the body that is called electroacoustic sounds or signals which can be captured with a transducer, or microphone, and converted to an electrical signal and forwarded to a processor to be converted to a ID, 2D, or 3D image as an electroacoustic computed tomogram while the electrical pulse generated in the
  • thermotherapy with focused low power ultrasound or alternating magnetic field, etc. is combined with the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles, etc. are coated with thermosensitive polymers, such as chitosan, with quenched fluorescein or another dye or indicator/medication etc.
  • the functionalized nanoparticles are exposed to a focused low power ultrasound in a non-thermal mode (LIFU) or high power thermal mode (HIFU) generating an electrical current by the focused ultrasound from the piezoelectric nanoparticles and/or pyroelectric functionalized
  • LIFU non-thermal mode
  • HIFU high power thermal mode
  • nanoparticles thus creating an electrical signal from the piezoelectric nanoparticles that paralyses the cell permitting the functionalized nanoparticles coated with cell penetrating agents to enter the cell cytoplasm in the cells and release the medication, gene, CRISPR-cas9 inside the lesion or tumor by the focused ultrasound to raise the temperature of the piezoelectric or pyroelectric nanoparticles that are injected inside the body to be attached to the surface antigens of the normal cells or of tumor cells and create nanoparticle/tumor cell complexes to 41-43 degrees C and a ID, 2D, or 3D image as an acoustic computed tomogram while the nanoparticles drive the medication, gene, in the tumor cells locally by depolarizing their cell membrane potential and to damage the tumor cells by multiple modes of the therapy applied non-invasively under observation.
  • thermotherapy with focused high power ultrasound or alternating magnetic field, etc. is combined with the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, with quenched fluorescein or another dye or indicator/ medication etc. conjugated with an antineoplastic medication, gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • thermosensitive polymers such as chitosan
  • an acoustic response is produced by electrical stimulation of piezoelectric nanoparticles inside the body that is called an electroacoustic sound or signal which can be recorded with a transducer, or microphone, and converted to an electrical signal
  • thermotherapy with focused low power ultrasound or alternating magnetic field (at a frequency of less than 100 kHz to 300 kHz) etc. is combined with the antibody coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles are coated with thermosensitive polymers, such as chitosan, with quenched fluorescein or another dye or indicator/medication, etc. conjugated with an antineoplastic medication, gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • thermosensitive polymers such as chitosan
  • an acoustic response is produced by electrical stimulation of piezoelectric nanoparticle inside the body that is called an electroacoustic sound or signal which can be recorded with a transducer, or microphone, converted to an electrical signal and forwarded to a processor to be converted to a ID,
  • the pluralities of nanoparticles absorb the energy and expand, thereby creating a photoacoustic ultrasound that can be recorded by a photoacoustic or thermoacoustic imaging system indicating the degree of the temperature achieved, and the increased tumor biomarker in the circulation after the thermotherapy having an important diagnostic value (i.e., presence of a tumor by increased biomarkers) and therapeutic value for vaccine production after harvesting the circulating biomarkers and vaccine production with antibody coated nanoparticle with viral-like particles (VLP) or oncolytic viruses, such as T-Vec, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors and administering them with checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • VLP viral-like particles
  • T-Vec monoclonal antibodies
  • IL-2 bee toxins or other immune stimulators
  • checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor
  • the photoacoustic or thermoacoustic unit is in communication with the thermal energy delivery system via a processor that can adjust thermal energy to achieve a desired temperature at the tumor site.
  • the thermal output of the thermal energy delivery system is a focused ultrasound which heats up the tissue located at the focal point of the focused ultrasound using an antibody/medication coated ultrasound contrast agent, such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells nanocage magnetic, gold, silicone, carbon, mixture of magnetic Nps measuring the temperature non-invasively by estimation of the temperature depending on acoustic harmonic, and back scatter from 26-46 C degrees generated by the nonlinear ultrasound wave.
  • GNPs gold nanoparticles
  • GNRs gold nanorods
  • GGS gold nanoshells
  • GNS graphene oxides
  • PPy polypyrrole
  • nanoshells nanocage magnetic gold, silicone, carbon, mixture of magnetic Nps measuring the temperature non-invasively by estimation of the temperature depending on acoustic harmonic, and back scatter from 26-46 C degrees generated by the nonlinear ultrasound
  • the thermal output of the thermal energy delivery system is an electromagnetic radiation which heats up the tissue located at the focal point, or is another visible or infrared laser, or other electromagnetic radiation using an antibody/medication coated agent such as gold nanoparticles with activatable cell-penetrating peptides (ACPPs), gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells nanocage magnetic, gold, silicone, carbon, mixture of magnetic Nps to heat up the nanoparticles measuring the temperature non- invasively using a photoacoustic imaging system in a temperature range of 37-60 C degrees.
  • ACPPs gold nanoparticles with activatable cell-penetrating peptides
  • GNPs gold nanoparticles
  • GNRs gold nanorods
  • GNS gold nanoshells
  • Gs graphene oxides
  • PPy polypyrrole
  • the thermal output of the thermal energy delivery system 10 is a focused high power ultrasound (HIFU) which heats up the tissue/tumor 12 located deep inside the body 14 at the focal point of ultrasound using an antibody/medication coated agent, such as iron oxide gold nanoparticles (GNPs) with activatable cell-penetrating peptides (ACPPs), gold graphene oxides (GOs), gold, silicone, carbon, mixture of magnetic nanoparticles (NPs), gold nanorods (GNRs), gold nanoshells (GNS) or a nanocage filled with a fluorescein dye or located inside a liposome containing quenched fluorescein or another dye or indicator, etc.
  • GNPs iron oxide gold nanoparticles
  • ACPPs activatable cell-penetrating peptides
  • GOs gold graphene oxides
  • NPs gold, silicone, carbon
  • GNRs gold nanorods
  • GNS gold nanoshells
  • a green light of 410 to 420 nm passing through a blue absorbing filter 22 and reached through a fiber optic 24 and a photomultiplier that converts the light to an electrical signal then to a processor 26 that, in turn, is connected to the thermal delivery system indicating the temperature of 41-43 degrees C is achieved, the operator can via software maintain the energy delivery for the desired time (e.g., 1-10 minutes or more, if needed) or continue the energy delivery until the next level of 43-56 degrees C when a focused ultrasound is used and its terminal is connected to the same processor to continue delivering energy to reach the temperature of 56 degrees C creating a popping or cavitation sound at the boiling point of the PFCL Nps when the ultrasonic receiver or the microphone 16 records a cavitation sound to be reported, via a receiver, to the processor 26 connected to either the ultrasonic unit or the AMF unit to stop producing the thermal energy to the tissue.
  • the desired time e.g., 1-10 minutes or more, if needed
  • a patient with a malignant tumor or benign lesion is administered antibody coated magnetic nanoparticles conjugated with Rock inhibitors, or Wnt inhibitors, or GSK 269962 inhibitors conjugated with a thermosensitive polymer, such as PEG, or chitosan or other polymers or nanoparticles carrying the slow release medication from porous silicon, or polylactic acid, or polyglycolic acid, along with other medications or genes with CRISPR-cas9 that may be released under an alternating magnetic field (AMF) at a 100 kHz to 300 kHz to shake up the polymeric material, gene, and medication without significant thermal effect or increasing the alternating magnetic field to 1 MHz to 10 MHz or more to induce simultaneously a thermal energy to heat up the tissue to a desired temperature as a result of the AMF frequency.
  • the thermal energy increases the surrounding tumor temperature from 37-41 degrees C or 41-50 degrees C or more which can be calculated depending on the AMF frequency and the duration of the therapy to release the gene or medication and damage the tumor
  • a focused high power ultrasound is used to heat the antibody/medication coated nanoparticles conjugated with thermosensitive polymer carrying medication, dye, monoclonal antibody or check point inhibitors, coated nanoparticles to release the medication when a temperature of 43 degrees C has been achieved and the medication is released locally without causing toxicity to the other organs of the patient and eliminating autoimmunity in the post treatment period.
  • the external thermal energy is delivered by a combination source, such as electromagnetic radiation, alternating magnetic field, radio frequency (RF), and low power ultrasound either in a compressive or pressure or high power thermal mode and the nanoparticle/tumor temperature can be imaged simultaneously with the ultrasound, MRI, CT-scan, PET-scan, photoacoustic imaging unit, etc.
  • a combination source such as electromagnetic radiation, alternating magnetic field, radio frequency (RF), and low power ultrasound either in a compressive or pressure or high power thermal mode and the nanoparticle/tumor temperature can be imaged simultaneously with the ultrasound, MRI, CT-scan, PET-scan, photoacoustic imaging unit, etc.
  • the pluralities of functionalized nanoparticles with activatable cell-penetrating peptides are conjugated with metabolically active compounds, e.g., F-18FDG used to visualize tumors and metastatic lesions using positron emission tomography-computed tomography (PET-CT) scanning in addition to the external thermal energy to visualize and damage the otherwise not sensitive tumor stem cell.
  • metabolically active compounds e.g., F-18FDG used to visualize tumors and metastatic lesions using positron emission tomography-computed tomography (PET-CT) scanning in addition to the external thermal energy to visualize and damage the otherwise not sensitive tumor stem cell.
  • PET-CT positron emission tomography-computed tomography
  • mAb coated nanoparticles administered intravenously with checkpoint inhibitors, and antineoplastic medication, taxol derivatives or Wnt inhibitors, such as WIKI4 are conjugated with thermosensitive polymers coated pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs), nanoshells or nanocages, etc.
  • neuropsychiatric syndrome associated with hyperammonemia or ARF6 inhibition associated with hyperammonemia or ARF6 inhibition.
  • nanoparticles/gemtuzumab to a site, or for systemic delivery may be combined with immunologically based B -cells or T-cells that are optionally genetically modified.
  • B- cells and T-cells may attack localized and/or unlocalized cancer cells or hematological cancers (e.g., leukemias, lymphomas, etc.).
  • one or more of these therapies are employed at lower doses when used in combination with the method of delivering antibody/medication coated nanoparticles.
  • nanoparticles of a material other than gold may be used. These include, without limitation, antibody/medication and thermosensitive coated diamond nanoparticles, platinum nanoparticles, combinations of gold, platinum, carbon, and/or diamond nanoparticles. Any of the above nanoparticles may contain at least one hydroxyl group. All such nanoparticles provide the various diagnostic and therapeutic applications as described above for gold nanoparticles. The sizes and shapes are the same as those described for gold nanoparticles. All such nanoparticles may be covalently attached to (poly)ethyleneglycol, (i.e., may be PEGylated).
  • thermotherapy to reach the temperature of 41-43 to 50 C to 56 degrees C temperature releases more cytokines from the damaged tumor cells activating the immune cell response more than if the medications were administered alone without thermotherapy, eliminating the need for CAR-T cellular therapy which requires a time consuming and expensive preparation with the chance of increased toxicity and an autoimmune response.
  • the thermal output of the thermal energy delivery system is a focused high power ultrasound which heats up the tissue located at the focal point of the focused ultrasound using an antibody/medication coated ultrasound contrast agent, such as gold nanoparticles (GNPs), gold nanorods (GNRs), gold nanoshells (GNS), graphene oxides (GOs), polypyrrole (PPy) nanocapsules, or nanoshells, nanocage magnetic, gold, silicone, carbon, mixture of magnetic Nps and PFCL, quenched fluorescein, or another dye or indicator, bubble liposome fluorescein, PFC1 nanoemulsions, etc., gold nanorods (GNRs) filled with albumen microbubbles, GNR loaded albumin microbubbles for combined ultrasound and photoacoustic or thermoacoustic imaging and thermotherapy therapy and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide
  • monoclonal antibodies, checkpoint inhibitors or antineoplastic medications, utomilumab are conjugated with antibody coated nanoparticles, nanoshells nanocage magnetic or paramagnetic, and VLP or oncolytic viruses, such as T-Vec , monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • thermosensitive polymers of the pluralities of nanoparticles with activatable cell-penetrating peptides ACPPs
  • ACPPs activatable cell-penetrating peptides
  • VLP nanoparticle conjugated medications
  • immune stimulator that are released locally at the cancer cells and have many side effects such as hyperactivation of T-cells and cytokine release causing nonspecific or specific body response (e.g., checkpoint inhibitors and vaccine therapy cause recruitment of specific T-cell to attack the tumor and potentially some specific organs that share the same checkpoint inhibitors) or cause excessive cytokine release producing Cytokine release syndrome (CRS) associated with fever, tachycardia, vascular leak, oliguria, hypotension, hyper activation of T-cells and cytokine such as those seen with checkpoint Protein Inhibitors,
  • CRS Cytokine release syndrome
  • the thermal delivery to the nanoparticle, nanoshells, or nanocages administered intravenously one can measure two distinct temperatures inside the body regardless of their location using either an electromagnetic, focused high power ultrasound, or alternating magnetic field, while releasing the medications at temperature of 41-43 degrees C, this can achieves a theranostic or diagnostic and therapeutic effect when the nanoparticles with activatable cell-penetrating peptides (ACPPs) carry a thermosensitive polymer, such as chitosan, and a dye, such as quenched fluorescein, or another dye or indicator, etc.
  • ACPPs activatable cell-penetrating peptides
  • thermotherapy with focused ultrasound or alternating magnetic field, etc. is combined with the thermal energy delivery source or combinations with low power compressive or pressure ultrasound.
  • thermotherapy with focused ultrasound or alternating magnetic field, etc. is combined with the thermosensitive polymeric compound chitosan and can carry DNA, siRNA, RNA, other genes or inhibitory genes and VLP, or oncolytic viruses, such as T-Vec , monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • oncolytic viruses such as T-Vec , monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • the thermosensitive polymeric compound is chitosan and can carry DNA, siRNA, RNA, other genes or inhibitory genes, that are released at the temperature of 41-43 degrees C along with a quenched fluorescein dye, or another dye or indicator, etc. in the circulation and can be seen in a few seconds in the circulation using a blood sample or inside a transparent butterfly tubing that fluoresces under UV radiation or other wave length indicating presence of the dye indicating that the temperature 41-43 degrees C at the site of the heated nanoparticles/tumor is achieved and the genes are released.
  • a combination of nanoparticles with activatable cell- penetrating peptides is used to have additional functionalities as well as imaging, thermoacoustic imaging for thermotherapy and imaging and tumor therapy by administering to a patient systemically or locally either antibody/medication coated copper sulfide (CuS) nanoparticles, or polymeric microbubbles of Polylactic acid or lactic acid microcapsules employing the water-in-oil-in-water double emulsion method or gold Nanorod-loaded PLA microcapsules for combined ultrasound contrast imaging and conjugated with a
  • ACPPs activatable cell- penetrating peptides
  • thermosensitive polymer such as chitosan with DNA, siRNA, RNAi, with CRISPR enzyme or with or without macrolides, such as cyclosporine A, mycophenolic acid, tactolimus or ascomycin or other drugs delivery at 41-43 degrees C temperature or monoclonal antibodies check point inhibitor, anti-neoplastic medications, or antibody coated gold nanoparticles with activatable cell-penetrating peptides (ACPPs) or antibody/medication coated gold nanoshells Perfluoro carbon liquid, or antibody coated perfluorohexane (PFH)-loaded magnetic hollow iron oxide nanoparticles (HIONs) and Fe-0 stretching vibration mode of Fe304T, or antibody coated nanoparticles, such as antibody/medication coated SPIOs or Gold Nanorod and iron oxide coating nanocapsules that absorb thermal delivery using alternating magnetic field, focused high power ultrasound and electromagnetic radiation or imaging
  • macrolides such as cyclosporine A, mycophenolic acid, tactolimus or ascomycin or other drugs delivery at
  • PET-scan simultaneously with MRI or CT contrast imaging, PET-scan, while having high X-ray absorption coefficient, to have multifunctional capabilities for cancer diagnosis and thermotherapy and thermoacoustic imaging for control of the temperature.
  • the temperature of antibody/medication coated pluralities of nanoparticles administered intravenously with activatable cell-penetrating peptides (ACPPs), nanocages, nanoshells, etc. is measured and imaged using different indicators of the temperature at the tumor site during the thermotherapy indicating temperature of either 41-43 degrees C or 44-49 degrees C or 50 degrees C or 56 degrees C using electromagnetic radiation and photoacoustic or thermoacoustic temperature imaging system or a temperature using alternating magnetic field , microwave, or radiofrequency to increase the temperature from 37-43 degrees C measured using a biocompatible dye, such as quenched fluorescein, or another dye or indicator, etc.
  • a biocompatible dye such as quenched fluorescein, or another dye or indicator, etc.
  • lipid coated nanoparticles that melt at 41-43 degrees C temperature and releases the dye in the circulation and recognized optically as in FIG. 1, or in the blood sample with UV radiation or the use of nanoshells or nanocages containing either a perfluorocarbon liquid such as perfluoro pentane or hexane that boils at the temperature of 56 degrees C, thereby creating a recordable cavitation sound recorded by a microphone of an ultrasonic receiver or, if heated with a source of energy such as alternating magnetic field (AMF) with the magnetic or paramagnetic nanoparticles with activatable cell-penetrating peptides (ACPPs) or nanoshells, etc.
  • AMF alternating magnetic field
  • ACPPs activatable cell-penetrating peptides
  • the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers or, tumor antigen, and antibody/medication coated nanoparticles and VLP and administering them with checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc., and Rock inhibitor, such as Fasudil, and its derivatives or Wnt inhibitors, such as niclosamide, to attach to the potential tumor cells while applying thermal energy to damage the recurrence of the tumor and induce reactivation of the immune response and eliminate the tumor cells and to enhance immune therapy.
  • checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitor such as Fasudil
  • Wnt inhibitors such as niclosamide
  • the administered thermosensitive polymeric/medication compound is chitosan conjugated with monoclonal antibodies (mAbs) or checkpoint inhibitors, Rock inhibitors such as Fasudil, etc., VLP, or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11 or an antineoplastic medication, daunorubicin, ara-C or cytarabine, taxol, taxane derivatives, etc.
  • mAbs monoclonal antibodies
  • checkpoint inhibitors such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11 or an antineoplastic medication, daunorubicin, ara-C or cytarabine, taxol, taxane derivatives, etc.
  • thermotherapy that are released at the temperature of 41-43 degrees C along with a quenched fluorescein dye or another quenched dye or indicator/medication, PFC nanobubbles, etc. in the circulation and can be seen in a few seconds in the circulation using a blood sample or inside a transparent butterfly tubing that fluoresces under UV radiation indicating presence of the dye indicating that the temperature 41-43 degrees C at the site of the heated nanoparticles/tumor is achieved and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP, IL-2 conjugated with antibody coated nanoparticles and administering them with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • checkpoint inhibitors such as PD-1, PD-L1, CTLA-4, Jagged 1
  • Rock inhibitors such as Fasudil or Wnt inhibitors, such as niclosamide
  • Fasudil or Wnt inhibitors such as niclosamide
  • the administered thermosensitive polymeric compound is chitosan conjugated with mAbs or checkpoint inhibitors, or antineoplastic medication, Daunorubicin, ara-C or cytarabine, taxol, taxane derivatives, etc. that are released at the temperature of 41-43 degrees C along with a quenched fluorescein dye or another quenched dye or indicator, etc.
  • thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1 , PD-L1, CTLA-4, Jagged 1 inhibitor 15D11 conjugated with antibody/medication coated nanoparticles and administering them with Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, to attach to the potential tumor cells, while applying thermal energy to damage recurrence of the tumor and
  • the administered thermosensitive polymeric compound is chitosan conjugated with mAbs or check point inhibitors, or antineoplastic medication, Artemisinins or artemisinin analogues against a variety of cancer cells, promoting apoptosis, preventing angiogenesis inhibition of Toll-like receptors, Syk, tyrosine kinase inhibitor, phospholipase Cy, PI3K Akt, MAPK, STAT- 1/3/5, NF- ⁇ , Spl, Axitinib and Nrf2/ARE signaling pathways that are released at the temperature of 41-43 degrees C along with a quenched fluorescein dye or another quenched dye or indicator, etc.
  • thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP, or oncolytic viruses such as T-Vec , monoclonal antibodies, IL-2, bee toxins or other immune stimulators conjugated with antibody coated nanoparticles and administering them with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, to enhance immune therapy etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, to attach to the potential tumor cells while applying thermal energy
  • the Wnt/ -catenin pathway can be inhibited indirectly by inhibiting GSK-3 Glycogen synthetizing Kinase, such as lithium chloride, a simple salt, or its Phosphoaminophosphonic form at an extremely low concentration of 1-3 nano-micromolar that can only be delivered using antibody coated pluralities of nanoparticles conjugated with thermosensitive polymers, such as chitosan conjugated with ACPP and quenched fluorescein, or another quenched dye or indicator/medication, etc.
  • GSK-3 Glycogen synthetizing Kinase such as lithium chloride, a simple salt, or its Phosphoaminophosphonic form
  • thermosensitive polymers such as chitosan conjugated with ACPP and quenched fluorescein, or another quenched dye or indicator/medication, etc.
  • antineoplastic medication or monoclonal antibody to release the dye and medication by thermal energy at controlled temperature of 39-41 degrees C to not only inhibit the Wnt pathway but also treat various Wnt/ -catenin signaling in a number of diseases such as inflammation, brain cancers, glioblastoma, glioma, prostate, breast, colon cancer, ovarian cancer, and Alzheimer' s Disease.
  • the Wnt/ -catenin pathway inhibitors can be used with sulfasalazine inhibitor of PIK3CA mutations, enzymes that are involved in cellular growth control signals, that is known to inhibit xCT, and can be delivered using antibody coated pluralities of nanoparticles conjugated with thermosensitive polymers, such as chitosan, conjugated with ACPP and quenched fluorescein, or another quenched dye or
  • antineoplastic medication or monoclonal antibody to release the dye and medication by thermal energy at a controlled temperature of 41-42 degrees C to not only inhibit the Wnt pathway but also treat various Wnt/ -catenin signaling in a number of diseases inhibit cCT and PIK3CA, such as in inflammation, brain cancers, glioblastoma, glioma, prostate, breast, colon cancer, and ovarian cancer.
  • the Wnt/ -catenin pathway inhibitors or Rock inhibitors can be used with sulfasalazine inhibitor of PIK3CA mutations, enzymes that are involved in cellular growth control signals, that is known to inhibit xCT and can be delivered using antibody coated pluralities of nanoparticles conjugated with thermosensitive polymers, such as chitosan, conjugated with ACPP and quenched fluorescein or another quenched dye or indicator/medication, such as inhibition of tyrosine, monoclonal antibodies to EGFR, VEGFR, PDGFR, and c-kit Cdc42: cell division control protein 42, ERK:
  • mTOR mammalian target of rapamycin
  • PI3K extracellular signal-regulated kinase
  • EGF(R) epidermal growth factor (receptor)
  • Grb2 growth factor receptor-bound protein 2
  • JNK c-Jun N-terminal kinase
  • MEK/MKK mitogen- activated protein kinase kinases
  • PDGF(R) platelet derived growth factor (receptor)
  • SOS son of sevenless
  • TAK TGF -activated kinase
  • TGF transforming growth factor
  • VEGF(R) vascular endothelial growth factor (receptor) to release the dye and medication by thermal energy at controlled temperature of 41-42 degrees C to not only inhibit the Wnt pathway but also treat various Wnt/ -catenin signaling in a number of diseases inhibit cCT and PIK3CA such as in inflammation, brain cancers, glioblastoma, glioma, glioma multiforme, prostate, breast, colon cancer, ovarian
  • the Wnt/ -catenin pathway inhibitors such as niclosamide at 500mg dose, ivermectin lgram -2 gram once a week, etc., or Rock inhibitors, such as Fasudil at 40-80 mg, etc.
  • niclosamide at 500mg dose ivermectin lgram -2 gram once a week, etc.
  • Rock inhibitors such as Fasudil at 40-80 mg, etc.
  • VEGF(R) vascular endothelial growth factor (receptor) to release the dye and medication by thermal energy at controlled temperature of 41-42
  • ERK extracellular signal-regulated kinase
  • mTOR mammalian target of rapamycin
  • PI3K phosphatidylinositol 3-kinase
  • EGF(R) epidermal growth factor (receptor)
  • Grb2 growth factor receptor-bound protein 2
  • JNK c-Jun N-terminal kinase
  • MEK/MKK mitogen-activated protein kinase kinases
  • PDGF(R) platelet derived growth factor (receptor)
  • SOS son of sevenless
  • TAK TGF -activated kinase
  • TGF transforming growth factor
  • VEGF(R) vascular endothelial growth factor (receptor) to release the dye and medication by thermal energy at controlled temperature of 41-42
  • a popping cavitation sound is produced and recorded by a microphone or an ultrasonic recorder, indicating the temperature of about 50 degrees C when the combination of sources of energy, or a combination of high power thermal energy and a low power compressive or pressure focused ultrasound is used creating a recordable cavitation sound at the temperature of about 56 degrees C and the medication is released from the nanoparticles with activatable cell- penetrating peptides (ACPPs) and the tumor cells to which the nanoparticles are attached are damaged by the thermal energy indicating sudden increase of tumor biomarkers in the circulation taken from blood samples for future antibody production in a potential repeated and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and
  • checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide
  • Fasudil or Wnt inhibitors, such as niclosamide
  • Wnt inhibitors such as niclosamide
  • antibody coated nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), and the tumor cells are recognized by the T cells, which together with killer cells phagocytose them, and enhance the immune response to the tumor and its exosomes and the circulating cells.
  • a Wnt inhibitor such as ivermectin
  • a Rock inhibitor such as Fa
  • electromagnetic energy such as laser, absorbing antibody coated nanoparticles with activatable cell-penetrating peptides (ACPPs), such iron oxide, gold, silicon carbon, magnetic, non-magnetic NPs or nanoshells, nanocages, nanotubes, nanorods, nanospheres, or any other shells produced (e.g., polylactic or polyglycolic acid, etc.) or piezoelectric nanoparticle administered systemically, topically, locally inside the tumor exposed to a thermal energy source such ultrasound or as laser light from 360-1000 nm or more or microwave or Radio frequency wave, to increase the temperature of the nanoparticles inducing a photoacoustic or thermoacoustic sound measuring the temperature of the heated tumor from 40-43- or more as needed and , the lipid component of the liposome and chitosan, melts, releasing medication and the dye at temperature of 41-43C degrees C or more to release the medication, etc.
  • ACPPs activatable cell-penetrating peptides
  • nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-Ll etc. (to disguise themselves), and the tumor cells and exosomes are recognized by the T-cells, which together with natural killer cells phagocytose them, and enhance immune response to the tumor, and its exosomes and the circulating cells.
  • the polymeric thermosensitive coating of the pluralities of nanoparticles administered with activatable cell-penetrating peptides melts and releases the medication at the melting point of the polymer, such as chitosan or liposomes containing quenched fluorescein dye or another quenched dye or indicator, medication, etc.
  • ACPPs activatable cell-penetrating peptides
  • ACPPs activatable cell-penetrating peptides
  • thermal energy i.e., confirming the presence of a tumor
  • therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and conjugated with antibody coated nanoparticle and checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, to attach to the potential tumor cells while applying thermal energy to damage recurrence of the tumor and induce reactivation of the immune response to eliminate them.
  • these biguanides administered along with aspirin can prevent cancer formation and in cancer treatment by inhibition of mTOR signaling via AMP- activated protein kinase (AMPK) -dependent and -independent path and inhibition of COX- l/COX-2 and modulation of the NFKB or STAT3 pathway and activation AMPK, and both agents may affect Notch, Wnt/ -catenin when delivered through the thermosensitive polymeric coated functionalized pluralities of nanoparticles and ACPP intravenously, orally or intracavity or intra-arterially at one nanogram to 100 nanogram concentrations interfering with the glucose metabolism of the tumor stem cells proliferation causing them to starve.
  • AMPK AMP- activated protein kinase
  • antibody/medication coated pluralities of nanoparticles administered with activatable cell-penetrating peptides (ACPPs) conjugated with Wnt inhibitor such as FH535, or rWP-2, or PNU-74654, or IWR-lendo, or rWR-exo, or demethoxycurcumin, or CCT036477, or KY02111, or ivermectin, niclosamide, metformin, or phenformin are encapsulated in nanogram concentrations within the chitosan coating of the nanoparticles, such iron oxide, gold, silicon carbon, magnetic, non-magnetic Nps or nanoshells , nanocages, nanotubes, nanorods, nanospheres, etc.
  • the lipid component of the liposome melts at temperature of 40-41 degrees C releasing the fluorescein in the circulation that can be observed by taking a blood sample or radiating the nail bed or any other part of the body with UV radiation so that now non-quenched fluorescein fluoresces as green light passing through a blue filter, indicating the temperature of 41-43 degrees C is achieved and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP, IL-2 conjugated with antibody coated nanoparticles and checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, to attach to
  • thermotherapy with focused ultrasound or alternating magnetic field, etc. is combined with antibody coated pluralities of nanoparticles are administered with activatable cell-penetrating peptides (ACPPs) conjugated with Wnt inhibitor such as FH535, or IWP-2, or PNU-74654, or IWR-lendo, or rWR-exo, or demethoxycurcumin, or
  • ACPPs activatable cell-penetrating peptides conjugated with Wnt inhibitor
  • Wnt inhibitor such as FH535, or IWP-2, or PNU-74654, or IWR-lendo, or rWR-exo, or demethoxycurcumin, or
  • CCT036477, or KY02111, or ivermectin, niclosamide, metformin, or phenformin with and without syrosingopine are encapsulated in nanogram concentrations within the chitosan coating of the nanoparticles, such as iron oxide, gold, silicon, carbon, magnetic, nonmagnetic nanoparticles or nanoshells, nanocages, nanotubes, nanorods, nanospheres, etc. inside the liposomes containing fluorescein and electromagnetic energy absorbing nanoparticles.
  • the lipid component of the liposome melts at a temperature of 40-41 degrees C, thus releasing the fluorescein in the circulation that can be observed by taking a blood sample or radiating the nail bed or any other part of the body with UV radiation so that now the non-quenched fluorescein fluoresces as green light passing through a blue filter, indicating the temperature of 41-43 degrees C is achieved and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP, IL-2 and other immune stimulators conjugated with antibody/medication coated nanoparticles and administering them with checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc
  • the nanoshells or nanocages contain one or the
  • thermoacoustic/ thermoacoustic sound wave that can be recorded by a photoacoustic/ thermoacoustic unit by which the temperature at the site of the nanoparticles are recorded depending on the amplitude of the ultrasonic wave, as an additional method to release of a dye for confirmation of the tumor 's temperature, and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers and administering them with checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc. and Rock inhibitors, such as Fasudil or Wnt inhibitors, such as niclosamide, for the future management of the patient with recurrences by making a vaccine using the biomarkers and
  • VLP/checkpoint inhibitors conjugated with antibody coated nanoparticles to attach to the potential tumor cells while applying thermal energy to damage recurrence of the tumor and induce reactivation of the immune response to eliminate them.
  • the photoacoustic or thermoacoustic unit is communicating with the thermal delivery unit via a processor to control the amount of energy released by the thermal energy delivery unit via a processor to stop or increase or decrease the temperature all below the thermal denaturation temperature of proteins which is 60 degrees C at the site of the nanoparticles as desired to prevent excessive thermal damage to the normal surrounding cells.
  • ACPPs activatable cell-penetrating peptides
  • Wnt inhibitor activatable cell-penetrating peptides
  • ACPPs activatable cell-penetrating peptides
  • Wnt inhibitor activatable cell-penetrating peptides conjugated with a Wnt inhibitor are encapsulated in the liposomes containing quenched fluorescein or liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the drug is released at the temperature or 41-43 degrees C or opens drug- permeable pores created bubble formation from the decomposition of encapsulated ammonium bicarbonate and another dye or indicator and electromagnetic energy absorbing nanoparticles, such as iron oxide, gold, silicon carbon, magnetic, non-magnetic Nps or nanoshells, nanocages nanotubes, nanorods, nanospheres, piezoelectric nanoparticles, etc.
  • nanoparticles
  • the lipid component of the liposome melts at s temperature of 40-41 degrees releasing the fluorescein in the circulation that can be observed by taking a blood sample or radiating the nail bed or any other part of the body with UV radiation so that the now non-quenched fluorescein fluoresces as green light passing through a blue wavelength absorbing filter, indicating the temperature of 41 degrees C is achieved, the fluorescent dye is recognized by an imaging unit communicating with the thermal delivery unit via a processor to control the amount of energy released by the thermal energy delivery unit via a processor to stop or increase or decrease the temperature using thermoacoustic imaging for control of the temperature, all below the thermal denaturation temperature of proteins, which is 60 degrees C, at the site of the nanoparticles as desired to prevent excessive thermal damage to the normal surrounding cells.
  • a thermal energy source such as electromagnetic radiation, microwave, or radiofrequency radiation
  • the processor of the thermal recoding unit stops the thermal energy delivery system to prevent further thermal energy delivery and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP, or oncolytic viruses such as T-Vec , monoclonal antibodies, IL-2, bee toxins or other immune stimulators conjugated with antibody coated nanoparticles and administering them with checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, to attach to the potential tumor cells while applying thermal energy to damage recurrence of the tumor and induce reactivation of the immune response to eliminate them while antibody coated nanoparticles/checkpoint inhibitors and VLP attach to the
  • the parameters of thermal heating are kept constant to prevent excess heating of the nanoparticles with activatable cell-penetrating peptides (ACPPs) / tumor is reached at 41-43 degrees C when the quenched fluorescein is used with a thermosensitive polymer or lipid or liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the drug is released at the temperature or 41-43 degrees C or opens drug-permeable pores created bubble formation from the decomposition of encapsulated ammonium bicarbonate and the fluorescent dye is recognized by an imaging unit communicating with the thermal delivery unit via a processor to control the amount of energy released by the thermal energy delivery unit via a processor to stop or increase or decrease the temperature or the temperature reaches about 56 degrees C when the perfluorohexane (PFH) or perfluoropentane incorporation in the nanocage or nanoshell evaporates, thus causing an acous
  • a Wnt inhibitor
  • a blue wavelength radiating source such as a laser or diode 28 providing a wavelength of 38o- 390 nm illuminating a finger nail bed 18 and the capillaries in the nail bed 18, which in presence of unquenched fluorescein fluoresces a green light of 410 to 420 nm passing through a blue absorbing filter 22 and reached through a fiber optic
  • a photomultiplier that converts the light to an electrical signal to a processor 26 that, in turn, is connected to the thermal delivery system indicating the temperature of 41-43 degrees C is achieved and the medication is released from the Nps
  • the fluorescent dye is recognized by imaging or thermoacoustic imaging for control of the temperature communicating with the thermal delivery unit via a processor to control the amount of energy released by the thermal energy delivery unit via a processor to stop or increase or decrease the temperature or by increasing the temperature of the nanoparticles or nanoshells, producing another popping or cavitation sound which is measured and recorded with a microphone or an ultrasonic receiver indicating
  • the increased tumor biomarker in the circulation after the thermotherapy has an important diagnostic and therapeutic value for the future vaccine production and management of tumor recurrences in the patients.
  • the microphone or the ultrasonic unit is in contact with the patient' s body surface records or images the cavitation sound of the PFCL temperature, and communicates to the energy producing unit that the desired temperature of 56 degrees C is achieved and stops further heating of the tissue to prevent expansion of the thermal energy via convection to the normal cells surrounding the tumor cells and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP, or oncolytic viruses such as T-Vec, monoclonal antibodies, IL-2, bee toxins or other immune stimulators conjugated with antibody coated nanoparticle and administering them with checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide
  • thermosensitive polymeric coating such as chitosan
  • ACPPs activatable cell-penetrating peptides
  • the fluorescence is seen in the blood sample of the patient being treated with the thermotherapy, and a processor connected to the temperature recording unit and thermal delivery unit can maintain or stop further heating of the tissue by recording and communicating the achievement of the desire temperatures via the processor's algorithm to the thermal delivery unit and the increased tumor biomarker in the circulation is obtained after the therapy has an important diagnostic and therapeutic value for the future vaccine production and management of tumor recurrences of the patient.
  • a blue wavelength radiating source such as a laser or diode 28, providing a wavelength of 380-390 nm illuminating to the finger nail 18 and the capillaries in the nail bed 18 which in presence of unquenched fluorescein fluoresces a green light of 410 to 420 nm passing through a blue absorbing filter 22 to output yellow light that is transmitted through a fiber optic 24 to a photomultiplier that converts the light to an electrical signal to a processor 26 that, in turn, is connected to the thermal delivery system indicating the temperature of 41-43 degrees C is achieved and the fluorescent dye is recognized by an imaging unit communicating with the thermal delivery unit via a processor to control the amount of energy released by the thermal energy delivery unit via a processor to stop or increase or decrease the temperature.
  • a blue wavelength radiating source such as a laser or diode 28
  • thermotherapy Further increasing the temperature by the thermal delivery unit 10 produces another a popping or cavitation sound, which is recorded with a microphone or an ultrasonic receiver 16 indicating the temperature of 56 degrees C boiling point of the PFCL has been achieved at the site of the nanoparticle/tumor cells, and the increased tumor biomarker in the circulation after the thermotherapy confirms the presence of a tumor and has an important diagnostic and therapeutic value for vaccine formation with VLP, or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators and administering them with checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide
  • antibody coated nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), and the tumor cells are recognized by the T cells, which together with killer cells phagocytose them, and enhance immune response to the tumor and its exosomes and the circulating cells.
  • thermosensitive polymers are chitosan, poly-lactic acid (PLA), poly(ethylene glycol), ethyl ether methacrylate-copoly(ethylene glycol), methyl ether methacrylate, Pluronic F127 (F127)[, poly(methylmethacrylate) (PMMA)], poly-n- isopropylacrylamide (PNIPAM), crosslinked ONIPAN hydrogels loaded with Fe304 nanoparticles, poly(ethyleneimine)-modified poly(ethylene oxide)-poly(propylene oxide)- poly(ethylene oxide) (PEO-PPO-PEO) block copolymer and nanoparticle- assembled capsules (NACs) using Poly(allylamine hydrochloride), and chitosan-poly(N-isopropylacrylamide).
  • thermosensitive polymers and methods of formulating these thermosensitive polymers with or in functionalized pluralities of nanoparticles containing chemotherapy drugs and/or a radiotracer are known in the art used with external thermal delivery, such as microwave, radio frequency (RF), or high power ultrasound, etc. to release the medication under the control of a processor at temperature of 41-43 or more degrees C as a combination of thermoradiation therapy.
  • external thermal delivery such as microwave, radio frequency (RF), or high power ultrasound, etc.
  • thermosensitive polymers with, for example, known pH sensitive polymers, magnetically controlled release agents, magnetoliposome agents, thermosensitive liposomes entrapping iron oxide nanoparticles with activatable cell- penetrating peptides (ACPPs), etc. are used, to achieve controlled drug release with external thermal delivery such as microwave, radio frequency (RF) or ultrasound, etc. under the control of a processor at temperature of 41-43 degrees C.
  • RF radio frequency
  • variable architecture thermosensitive polymers that are temperature- and pH-responsive, smart polymers, including synthetic copolymers and polymer derivatives, functionalized pluralities of nanoparticles encased in poly(N- isopropylacrylamide) (PNIPAM), poloxamers, poloxamines, polymers with thiol groups (PMA.sub.SH) are used with antineoplastic medications or monoclonal antibodies to release the medication with an external source of energy or magnetic induction under the control of a processor at temperature of 41-43 degrees C.
  • PNIPAM poly(N- isopropylacrylamide)
  • PMA.sub.SH polymers with thiol groups
  • the pluralities of functionalized pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) and temozolomide are administered intravenously, intra-arterially, locally, etc. contain at least one radioactive isotope and coated with anti-tumor antibodies, monoclonal antibody, the nanoparticles forming an antibody labeled nanoparticle-cell complex at a target site, the radioactive nanoparticle resulting in radiation therapy at a dose sufficient to damage tumor-associated vessels at the localized target site, and the DNA of the tumor stem cells while administering thermal energy simultaneously to achieve controlled drug release with external thermal delivery such as microwave, RF, or low power ultrasound etc. under the control of a processor at temperature of 41-43 degrees C in treatment of many brain tumors specially glioblastoma.
  • the pluralities of radioactive functionalized nanoparticles with activatable cell-penetrating peptides (ACPPs) coated with thermosensitive polymers and antineoplastic medication, such as taxol and or temozolomide are administered locally or systemically, intravenously, intra-arterially, inside a body cavity, and are magnetic, diamagnetic, ferromagnetic, non-ferromagnetic and/or paramagnetic nanoparticle, wherein the radioactive isotope is an alpha.-emitting isotope, a beta.-emitting isotope, or a combination thereof consisting of At.sup.211, Ac.sup.225, Bi.sup.212, Bi.sup.213,
  • an external or internal thermal delivery such as laser, microwave, RF, or low power focused ultrasound, etc.
  • ACPPs activatable cell-penetrating peptides
  • a processor under the control of a processor at a temperature of 41-43 degrees C to damage the tumor cells and their vascular supplies, the DNA of the tumor stem cells, and release the medication, such as temozolomide, in treatment of many cancers including brain tumors where thermal therapy can be repeated in the postoperative period as needed.
  • groups of non-magnetic, paramagnetic, or magnetic functionalized pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) and thermosensitive polymeric coating are produced where polymeric coating of these nanoparticles renders them less toxic to normal tissue such as Iodine.sup.131 may be crosslinked with antibody-coated nanoparticles to label the tumor or bacterial cells, both for therapy and imaging.
  • Anti-integrins or anti-VEGFs may also be incorporated or the nanoparticles may be used simultaneously for drug delivery purposes against these tumor or bacterial cells, with the drug in PDA, PGLA, dextran, dendrimers, PEG, etc.
  • the functionalized antibody/medication coated nanoparticles and thermosensitive polymeric coating may be quantum dots, magnetic and paramagnetic fullerene, encapsulate ferromagnetic nanoclusters with activatable cell-penetrating peptides (ACPPs), polymer based nanoparticles, colloidal gold nanoparticles, iron oxide nanoparticles, magnetic and paramagnetic fullerene, etc.
  • the nanoparticle size may range from 2 nm to 400 nm in one embodiment, and from 2 nm to 200 nm in another embodiment.
  • a focused ultrasound is used to heat the antibody/medication coated nanoparticle with activatable cell-penetrating peptides (ACPPs) conjugated with thermosensitive polymeric coated nanoparticles carrying a medication, dye, VLP .
  • ACPPs activatable cell-penetrating peptides
  • IL-2 monoclonal antibody or check point inhibitors alone to release the medication when a temperature of 43 degrees C has been achieved and the combination of monoclonal antibody, VLP and thermotherapy eliminate the need for the use of CAR-T cell that increases postoperative cytokine storm and autoimmune response, and the increased tumor biomarkers in the circulation after the therapy has an important diagnostic value (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for production of vaccine and administering them with checkpoint inhibitors such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • checkpoint inhibitors such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • a focused high energy ultrasound is used to heat the antibody/medication coated nanoparticle with activatable cell-penetrating peptides (ACPPs) conjugated with thermosensitive polymeric coating carrying a medication, dye, monoclonal antibody, or check point inhibitors alone to release the medication when a temperature of 43 degrees C has been achieved and the combination of monoclonal antibody and thermotherapy eliminate the need for the use of CAR-T cell that increases postoperative cytokine storm and autoimmune response, and the increased tumor biomarkers in the circulation after the therapy has an important diagnostic value (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for production of vaccine and administering them with checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc.
  • ACPPs activatable cell-penetrating peptides
  • Rock inhibitors such as Fasudil, (40-80 mg tablets) etc. or Wnt inhibitors, such as niclosamide ( ⁇ 500mg dose), ivermectin etc. 1-2 gram dose once in a week or can be repeated as needed in another month for the future management of the tumor recurrences in the patient, while the Rock inhibitor reduce TGF- ⁇ production after therapy and the subsequent scar formation.
  • a focused low energy ultrasound is used in a compressive or pressure mode which does not heat up the functionalized nanoparticles with activatable cell- penetrating peptides (ACPPs) and thermosensitive polymeric coating (e.g., the nanocages, nanoshells, etc.), but releases the coated medication, dye, monoclonal antibody, or check point inhibitors alone by shaking up the nanoparticles with activatable cell-penetrating peptides (ACPPs) to release the medication and damage the cell by the compressive action of the focused low energy ultrasound while imaging the lesion while the delivery of medication is verified by the release of the dye/medication in the circulation, as shown in FIG. 1.
  • ACPPs activatable cell- penetrating peptides
  • thermosensitive polymeric coating e.g., the nanocages, nanoshells, etc.
  • a focused low power ultrasound in a compressive or pressure mode is used alternatively with a heating high energy ultrasound which releases the coated medication, dye, monoclonal antibody or check point inhibitors alone at a lower temperature of 37-38 degrees C without damaging the tissue by shaking up the antibody/medication nanoparticles coated with cell penetrating peptides with thermosensitive polymeric coating, while keeping the temperature at ⁇ 40 degrees C that does not damage normal cells, but makes the tumor membrane permeable to medication to release the medication and damage the tumor cell by the compressive action of the compressive mode of focused low power ultrasound while imaging the lesion and delivering the medication is verified by the release of the dye/medication in the circulation as shown in FIG. 1.
  • compressive or pressure mode which does not heat up the functionalized with cell penetrating peptide and medication coated nanoparticle, but releases the coated medication, dye, monoclonal antibody or check point inhibitors by shaking up the functionalized pluralities of nanoparticles, or magnetic and paramagnetic fullerene, or encapsulate ferromagnetic nanoclusters and damages the cell membrane by the compressive action of the focused ultrasound while imaging the lesion with the compressive focused ultrasound and delivering the dye/medication is verified by the release and presence of the dye/medication in the circulation, as shown in FIG. 1.
  • a focused low power ultrasound is used in a compressive or pressure mode alternating with the thermal mode which releases the coated medication, dye, monoclonal antibody or check point inhibitors from the functionalized nanoparticles with ACPP at a lower temperature of 41 degrees C seen by release of the dye in the circulation, without damaging the normal tissue, by shaking up the pluralities of nanoparticles while keeping the temperature at about at or below 41 degrees C that does not damage normal cells, but makes the tumor membrane permeable to medication, and releases the dye/medication and damages the tumor cell by the compressive action of the focused low power ultrasound while imaging the lesion and delivering the medication is verified by the release of the dye/medication in the circulation, as shown in FIG. 1.
  • a focused low energy ultrasound in a compressive or pressure mode is alternatively used with heating focused high power ultrasound to image the lesion and release the coated medication, dye, monoclonal antibody or check point inhibitors alone at lower temperature of 41 degrees C without damaging the normal tissue by shaking up the nanoshell or nanocage containing PFCL, while keeping the temperature at about 41 degrees C that does not damage normal cells, but makes the tumor membrane permeable to medication to release the medication/dye, then increase the thermal energy or the duration of the thermal/compressive or pressure low power ultrasound causes the PFCL to reach boiling point at an earlier temperature of 47 to 50 degrees C which is lower than the boiling point of the PFCL (56 degrees C) recorded by creating a cavitation or popping sound indicating the desired temperature is achieved which is being recorded by a microphone or an ultrasonic receiver connected to the patient's body and communicates to the ultrasound delivery unit that desired temperature has been achieved, and to stop further delivery of the focused ultrasound via the processor automatically.
  • nanoparticles/tumor cell is measured by the release of dye/fluorescein form the
  • thermosensitive polymeric coating of the nanoparticle in the circulation that can be observed by UV radiation and a filter from the nail bed capillaries indicating a temperature of 41-43 degrees C at the NPs/tumor cell complex (see FIG. 1) or recording a popping sound at temperature of 56 degrees C when the nanoshell with PFCL hexane, etc. is used.
  • nanoparticles/tumor cell is measured when a focused low power ultrasound and alternating magnetic field is used to heat up the nanocage, or nanoshells to up to the boiling point of their PFCL hexane or pentane that is about 50 to 56 degrees C and image the temperature with thermoacoustic imaging system for control of the temperature.
  • nanoparticles/tumor cell is measured similarly when the combination of compressive or pressure mode and thermal focused high power ultrasound is used to heat up the nanocages or nanoshells to up to the boiling point of their PFCL hexane or pentane that is about 50 to 56 degrees C.
  • the parameters or thermal heating of the nanoparticle, nanocage or nanoshell is kept constant to heat up the antibody/medication coated pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs), the temperature reached is about 56 degrees C that is below 60 degrees C temperature of protein denaturation, the incorporation of perflourohexane PFH cause evaporation of PFH at a 56 degrees C temperature causing an acoustic sound that can be recorded with an ultrasonic receiver and the simultaneous increased tumor biomarker seen in the blood samples after the
  • thermotherapy having an important diagnostic release of the large number of tumor biomarkers and therapeutic value for making a useful vaccine and administering them with antibody coated checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc. and VLP to enhance immune therapy, and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, to stimulate a robust immune response in the future if tumor recurrences are observed in the patient while reducing TGF- ⁇ production after therapy and the subsequent scar formation.
  • antibody coated checkpoint inhibitors such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc. and VLP to enhance immune therapy
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide
  • electromagnetic radiation, microwave, radio frequency (RF) or an alternating magnetic field or focused low power ultrasound alone or with compressive or pressure mode focused ultrasound along with antibody bubble liposomes which contain air pockets or nanoemulsions of PFC carrying fluorescein, gene and/or medication coated with CPP conjugated with a thermosensitive polymer, such as chitosan, nanocages or nanoshells filled and perfluorocarbon liquid with magnetic nanoparticles, nanocages or nanoshells can be heated by different thermal energy sources to create three distinct temperatures at the tumor site, image the temperature with thermoacoustic imaging system for control of the temperature 41-43 degrees C measured by dye/medication release in the circulation or 50-56 degrees C by recorded cavitation sound when combination of compressive or pressure mode and heating of focused high energy ultrasound or alternating magnetic field are used to create a cavitation sound or the temperature of 56 degrees C if focused high power ultrasound or AMF or laser is used alone along with the PFCL Hexane nanoshells or nanocages.
  • RF radio frequency
  • either of the temperature indicators communicate via a processor that the desired temperature is achieved by a given thermal energy, to control the temperature and the duration of the desired temperature in the tissue, thereby separating the thermal drug delivery process from the cells destructive thermal delivery, as needed, via a processor that connects the temperature imaging system to the thermal delivery unit.
  • the control of the functionalized nanoparticles/tumor cell complex temperature is controlled at the desired level without overcooking the normal tissue and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value by measuring and confirming the presence of a tumor increased serum biomarkers, and dye release and therapeutic value for collecting biomarkers shortly after thermotherapy from the circulation for the future vaccine production for management of the patient with recurrences by making a vaccine using VLP , or oncolytic viruses , monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with antibody coated nanoparticles and administering them with checkpoint inhibitors, such as PD-1, PD- Ll, CTLA-4, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, to attach to the recurrent tumor cells while applying thermal energy to damage tumor recurrences and induce reactivation of the immune response to eliminate the tumor and
  • checkpoint inhibitors such as
  • the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value confirming the presence of a tumor and therapeutic value as biomarkers for the future vaccine production with management of the patient with recurrences by making a vaccine using VLP, or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators conjugated with antibody/medication coated nanoparticles and checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors or Wnt inhibitors, such as niclosamide, to attach to the tumor cells while applying thermal energy to damage recurrence of the tumor and induce reactivation of the immune response to eliminate the tumor.
  • VLP or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators conjugated with antibody/medication coated nanoparticles and checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors or Wnt inhibitors, such as niclosamide
  • the antibody/medication coated nanoparticle or nanoshell solution containing 2.67 nM gold nanorods, 2% human serum albumin, 0.04% (w/v) avidin and filling with PFC (C 3F 8) gas then modified via biotin-avidin technique to result in anti VEGF (Avastin), aflibercept to target by ultrasonography, the area of angiogenesis, such as a tumor or wet form of age related macular degeneration
  • VEGF vastin
  • aflibercept to target by ultrasonography
  • the area of angiogenesis such as a tumor or wet form of age related macular degeneration
  • the GNRs could induce
  • thermoacoustic or thermoacoustic imaging and thermal therapy under electromagnetic radiation or focused low power ultrasound showing the theranostic value of this modality on keeping the temperature at the desired temperature of 41-43 degrees C not to damage the surrounding normal tissue while treating a tumor (e.g., glioblastoma) or ARMD at low temperature and release medication and Wnt inhibitor of Rock inhibitors (Fasudil, Y-27632, small molecule inhibitor of ROCK1 and ROCK2 which act as an anti-inflammatory agent and inhibit Wnt activation, from the thermosensitive nanoparticle under the control of the temperature and the increased glioblastoma biomarkers, such as co-receptor, glypican 2 (GLP2) or cell surface GPC2 in the circulation after the therapy has an important diagnostic value (i.e., indicating the presence of a tumor) and therapeutic value as biomarkers for vaccine production and administering them with antibody coated VLP, checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor
  • the antibody/medication coated nanoparticle or nanoshell solution containing 2.67 nM gold nanorods, 2% human serum albumin, 0.04% (w/v) avidin and filling with PFC (C 3F 8) gas then modified via biotin-avidin technique to result in anti VEGF (Avastin), aflibercept or Axitinib and quenched fluorescein, bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC, gen or another dye or indicator, in a thermosensitive coating of the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) to target by ultrasonography the area of angiogenesis, such as tumor or suspicious breast cancer or ovarian cancer, while GNRs could induce photoacoustic or thermoacoustic imaging and thermal therapy under AMF or focused low power ultrasound showing the theranostic value of this modality on keeping the temperature at the desired temperature of 41-43 degrees
  • the thermal energy is applied to the body to find out if the antibody/medication coated pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) nanoshells with PFCL or nanocages, etc. are accumulated at a potential place, such as brain, breast, prostate, ovaries, etc., the nanoparticles when heated with thermal energy, such as AMF or focused low power ultrasound that can be recorded by a receiver and image the temperature, while recognizing the location of the potential tumor also in conjunction with MRI, or CT-scan or PET-scan, etc.
  • ACPPs activatable cell-penetrating peptides
  • the tumor is verified with functionalized pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) coated with multiple antibodies to biomarkers found in the serum, administered intravenously, intra-arterially, in a body cavity, subcutaneously to seek lymphatic vessels and attach to the tumor receptors located in various organs to be heated with either laser, focused high energy ultrasound or alternating magnetic field or microwave to either produce a photoacoustic or thermoacoustic sound or other sounds indicating expansion or nanoparticle or its content such as a gas, perfluorocarbon liquid expansion indicating the transition temperature from fluid to gas of 56 degrees C, which is recorded by a microphone or an ultrasound receiver and the temperature is imaged with a thermoacoustic imaging system for control of the temperature and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the patient.
  • ACPPs activatable cell-penetrating peptid
  • the recoding or temperature imaging system is connected to the thermal delivery unit via a processor to prevent increase of the temperature above predetermined level or maintaining or reducing the thermal energy or stopping it.
  • two indicators are used to indicate the temperature at the level of the tumor.
  • One indicator is quenched fluorescein, bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC, or a non-toxic dye/medication encapsulated in small functionalized liposomes that are administered simultaneously with other functionalized pluralities of nanoparticles that can act as a contrast agent and heat up when exposed to the thermal energy, where the liposome breaks up at temperature of about 42 degrees C and release its content fluorescein that can be easily recognized in the circulating blood indicating the temperature of 41 degrees C is achieved increasing the thermal energy heats up nanoparticles or nanocages to reach the transition or boiling point of perfluorocarbon liquid at 56 degrees C with a popping cavitation sound recorded by a transducer indicating the second desired temperature is achieved and via the processor to stop further heating of the tissue and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor)
  • an alternating magnetic field can heat up the antibody/medication coated nanoparticle or nanoshells in a patient having an ovarian tumor incorporated with a liquid perfluorocarbon, such as perfluoropentane, hexane, octane shells coated with Wnt inhibitor, anti-neoplastic medication and a rock inhibitor such as fasudil Rock inhibitors (HA-1077 a selective RhoA/Rho kinase (ROCK) inhibitor), Y-27632, small molecule inhibitor of ROCK1 and ROCK2 which act as an anti-inflammatory agent and inhibits Wnt activation, administered in a patient having an ovarian tumor that expands rapidly when the tissue is heated by a source of energy such as AMF or focused high power and high intensity ultrasound to reach a temperature 43 degrees C or beyond 43 degrees C to 56 degrees C which is the boiling point of these PFCLs, thereby creating a sudden gas expansion and a popping or cavitation sound that can be recorded by a source of energy such as A
  • the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides are micelles of perfluorocarbon liquids having a hydrophilic head having thermosensitive polymer such as chitosan that release the medication/dye at 41-42 degrees C when the polymer melts and an increased temperature to 56 degrees C creates a cavitation ultrasonic sound that is recorded indicating damage to the tumor cell membrane by thermal energy.
  • thermosensitive polymer of the nanoparticles or micelles carries a dye, such as fluorescein, which is released at the temperature of 41-43 degrees C in the circulation and its presence in the serum can be verified rapidly using a UV radiation indicating that the thermal energy created a temperature of 41-43 degrees C at the site of the nanoparticles.
  • a dye such as fluorescein
  • the thermosensitive polymer carries dye, medication, or a gene to be released at temperature of 42 degrees C to affect the tumor or kill the tumor cells at 56 degrees C thereby increasing the amount of the antigen released from the tumor cells which attracts more immune response to the biomarkers that are not normally available in the circulation and collecting these biomarkers can build the basis of a better cancer vaccine and administering them with antibody coated VLP or other immune stimulators and checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11, etc. to enhance immune therapy, and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide reducing TGF- ⁇ production after therapy and the subsequent scar formation and
  • the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides carry tumor monoclonal antibodies along with a Wnt inhibitor as the tumor is heated to 42 degrees C and its membrane becomes more permeable to the medications or gene released.
  • an alternating magnetic field can heat up the antibody coated nanoparticles or nanoshells in a patient having an ovarian tumor incorporated with a liquid perfluorocarbon, such as perfluoropentane, hexane, octane shells coated with Wnt inhibitor, anti-neoplastic medication and a rock inhibitor such as fasudil which acts as an antiinflammatory agent and inhibits Wnt activation, administered in a patient having an ovarian tumor that expands rapidly when the tissue is heated by a source of energy such as AMF or focused high power ultrasound to reach a temperature 43 degrees C or beyond 43 degrees C to 56 degrees C, and the temperature is measured with thermoacoustic imaging system for control of the temperature, which is the boiling point of these PFCLs, thereby creating a sudden gas expansion and a popping cavitation sound that can be recorded by a microphone indicating that the 56 degrees C temperature is achieved.
  • a liquid perfluorocarbon such as perfluoropentane, hexane,
  • the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides are micelles of perfluorocarbon liquids having a hydrophilic head with a thermosensitive polymer coating, such as chitosan, or liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the drug is released or the temperature or 41-43 C degree or opens drug-permeable pores created bubble formation from the decomposition of encapsulated ammonium bicarbonate that release the medication/dye at 41-42 degrees C when the polymer melts and an increased temperature to 56 degrees C creates a cavitation ultrasonic sound that is recorded, indicating damage to the tumor cell membrane by thermal energy.
  • a thermosensitive polymer coating such as chitosan
  • liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the
  • the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides are coated with thermosensitive polymers conjugated with medication, monoclonal antibodies and check point inhibitors, and are injected intravenously, intra-arterially, locally, systemically, subcutaneously, subconjunctival, orally, or by inhalation.
  • the nanoparticles find the tumor cells expressing the tumor surface antigen, attach to the cell membrane receptors of the tumor cells, during the thermotherapy at temperature of 41-43 degrees C when the thermosensitive polymer melts, the medication, monoclonal antibodies and checkpoint inhibitors and VLP, etc.
  • thermosensitive polymers are released precisely at the tumor site, eliminating the present practice of administrating the unbound medication, or unbound checkpoint inhibitors in the circulation, which can kill the exposed normal cells by the medication or attack the cells of the body which also express normally checkpoint inhibitors, Rock inhibitors, such as Fasudil, etc. to reduce inflammatory process and production of and TGF- ⁇ .
  • the latter induces various side effects as autoimmune response to various organs of the patient, such as the lung, digestive tract, skin, heart or kidney etc. that are difficult to manage while eliminating the tumor with a precise and localized immune response.
  • the functionalized or liposomes or nanoparticles are coated with thermosensitive polymers, conjugated with a dye and with Wnt inhibitors that reduces the post-treatment severe inflammatory response and reduces the impulse for tumor cell multiplication by inactivation of the Wnt pathway, and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the patient.
  • the pluralities of antibody/medication coated functionalized pluralities of nanoparticles or liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the drug is released or the temperature or 41-43 C degree or opens drug-permeable pores created bubble formation from the decomposition of encapsulated ammonium bicarbonate conjugated with Wnt inhibitors and rock inhibitors which act as antiinflammatory agents and inhibit Wnt activation, and thermosensitive coated polymers or a combination of small liposomes containing nanoparticles that act as thermal energy contrast agents are administered intravenously, intra-arterially, locally, etc.
  • thermotherapy to image the temperature using contrast agents, such as liposomes containing nanoshells, or nanocages containing in addition either a medication or a dye or an high power high intensity ultrasound contrast agent, such as PFCL, that creates a sound wave when heated under the control of the thermal energy delivery unit to reach the boiling point of the PFCL at temperature at 56 degrees C temperature creating a popping or cavitation sound, then recording it by a microphone indicating the 56 degrees C has been achieved and to damage to the tumor cell and do not heat up the tissue further to limit thermal damage by heat spillover to the normal cells surrounding the tumor and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the patient and enhancing the effect of immunotherapy or medication by thermotherapy and preventing production of medication resistance in these diseases.
  • contrast agents such as liposomes containing nanoshells, or nanocages containing in addition either a medication or a dye or an high power high intensity ultrasound
  • one treats excessive an immune response to the immune therapy by stopping the immune therapy shortly after it appears that the therapeutic goal has been achieved by administration of medications with antibody coated nanoparticles with activatable cell-penetrating peptides (ACPPs) conjugated with a thermosensitive polymer that releases them at the desired site at the temperature of 41-43 degrees C at lower concentrations than was previously possible with standard intravenous administration and at the site of a tumor, not the entire body, thereby preventing the tissue over-reaction to the immune therapy and enhancing the effect of immunotherapy or medication by thermotherapy and preventing production of medication resistance in these diseases.
  • ACPPs activatable cell-penetrating peptides
  • the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides are conjugated with the rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc. to dampen postoperative cytokine inflammatory response and also block the Wnt pathway inside the proliferating cancer cells or inflammatory cells seen after monoclonal antibodies or chemotherapy.
  • the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides contain medications with the rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, small molecule inhibitor of ROCK1 and ROCK2 along with slow release polymers, such as polylactic or polyglycolic acid or combinations of them carrying immunosuppressive agents to reduce the cytokine response in CAR-T cell immunotherapy.
  • the rock inhibitor Fasudil HA- 1077
  • ROCK selective RhoA/Rho kinase
  • Y-27632 small molecule inhibitor of ROCK1 and ROCK2
  • slow release polymers such as polylactic or polyglycolic acid or combinations of them carrying immunosuppressive agents to reduce the cytokine response in CAR-T cell immunotherapy.
  • tumor recurrences after initial CAR-T cells therapy in hematological malignancies such as lymphomas or leukemias or in solid tumors, such as melanoma, glioblastoma, ovarian, breast cancer, intestinal and colon cancer, prostate cancer, sarcoma, bone cancer
  • one administers intravenously, intraperitoneally, or subcutaneously, a combination of antibody coated nanoparticles conjugated with an antineoplastic medication along with Wnt inhibitors or a Rock inhibitors, ipilimumab, nivolumab, avastin, or aflibercept and applies external thermal energy to heat up the functionalized pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) to release the medications at the temperature of 41-43 degrees C or higher to inhibit Wnt activation by the metastatic cell and kill the tumor cells by increasing the nanoparticles temperature to 56 degrees C to kill the
  • ACPPs activatable cell-pen
  • antibody/medication polymeric coated nanoparticles and checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, for a lasting immune stimulation, etc., and Rock inhibitors or Wnt inhibitors, such as niclosamide, to be administered as needed, subcutaneously or systemically to attach the nanoparticles with activatable cell-penetrating peptides (ACPPs) to the potential tumor cells, applying thermal energy to release the medication from the thermosensitive polymer coated nanoparticles and damage the tumor recurrence and to reactivate the immune response for a longer time up to months and eliminate the tumor cells and its potential metastatic lesions.
  • ACPPs activatable cell-penetrating peptides
  • tumor recurrences after initial CAR-T cells therapy in hematological malignancies such as lymphomas or leukemias or in solid tumors, such as ocular or skin melanoma, glioblastoma, ovarian, breast cancer, intestinal and colon cancer, prostate cancer, sarcoma, bone cancer
  • hematological malignancies such as lymphomas or leukemias or in solid tumors, such as ocular or skin melanoma, glioblastoma, ovarian, breast cancer, intestinal and colon cancer, prostate cancer, sarcoma, bone cancer
  • Ivermectin or Niclosamide can be given orally at a low dose in conjunction with the standard immunothermotherapy procedure.
  • all medications are coated with a plurality of functionalized nanoparticles for local delivery from the thermosensitive nanoparticle to release the medication when the temperature reaches 41-43 degrees C under the control of the thermal energy delivery system processor connected to the thermal imaging recorder system and its software algorithm to control the temperature or prevent overheating the tissue.
  • Ras oncogenes and the proteins (Ras) encoded genes have had an important roles in the pathogenesis of the human cancer affecting >50% of colon and >90% of pancreatic cancers.
  • Ras proteins are membrane-bound GTP-binding proteins that serve as molecular switches in mutagenic signal transduction.
  • inhibitors such as Farnesyl or transferase, are used to block Ras activation and other relevant pathway inhibitors of PK1 and PK8 and P13K,
  • RAF/MEK/ERK, PI3 K/AKT/mTOR and RalA/B pathways that can be inhibited by administering gemcitabine, wortmannin, LY294002 and tipifarnib alone or in combination with Gemcitabine or rapamycin and AZD8055(aTORKi), everolimus, and avastin, aflibercept and quenched dye or another indicator conjugated with antibody/medication coated thermosensitive polymers and/or pluralities of nanoparticles with ACPP intravenously, intra- arterially, in the cerebrospinal fluid, locally, intraperitoneally, and release the dye and the medication by applying external, local, or internal thermal energy and damage the tumor cells that are damaged by the temperature of 41-43 degrees C to 56 degrees C, thereby enhancing the effect of immunotherapy or medication and prevent production of medication resistance in these diseases, such as pancreatic cancer, breast cancer and brain cancer, including glioblastoma, glioma, prostate cancer, ovarian cancer, and release the antigenic
  • the Wnt inhibitor Ivermectin is PEGylated with a lipid and conjugated with antibody coated nanoparticles to release the medication precisely at the site of the tumor and prevent potential toxicity of ivermectin if given in a high dose systemically that activates P2X4/P2X7 and Pannexin- 1 channels releasing ATP that enhances rapidly the toxicity of P2X7 receptor inducing sever tissue necrosis, whereas the controlled local drug delivery with thermally heating the tumor cells melts the thermosensitive polymer releasing the medication from the thermosensitive coated NPs slowly at 41-43 degrees C locally without the overreaction side effects seen after systemic, or oral ivermectin administration, thereby targeting multiple cancer cells, such as leukemia, glioblastoma, or glioma, or ovarian cancer, or breast cancer, Triple negative breast cancer cells, gastric cancer, colon cancer, or melanomas, etc. to damage the
  • Ivermectin a Wnt/ -catenin pathway inhibitor prepared as PEGylated lipid nanoemulsion conjugated with monoclonal antibody coated functionalized pluralities of nanoparticles with activatable cell -penetrating peptides (ACPPs), nanoshell or nanocage administered systemically intravenously, intra-arterially locally, intraperitoneally, or in the cerebrospinal fluid to attach to the cancer cells.
  • ACPPs activatable cell -penetrating peptides
  • the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic implication for a presence of a tumor in the body and obtaining increased tumor biomarkers from the circulation after the thermotherapy, to confirm diagnosis of the specific cancer and cold storage of the new biomarkers for future management of the patient with potential recurrences by making a vaccine by combining the biomarkers conjugated with VLP , or oncolytic viruses , monoclonal antibodies, IL-2, bee toxins or other immune stimulators and/or conjugating them with antibody/medication polymeric coated nanoparticles and checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors or Wnt inhibitors, such as niclosamide, administering them, subcutaneously or systemically, intravenously at very low dose to a patient, thereby the nanoparticles with the activatable cell-penetrating peptides (ACPPs) attach to the tumor cells, applying thermal energy to release the medication from the thermosensitive polymer coated nanoparticle
  • the pluralities of antibody/medication coated nanoparticles conjugated with either thermosensitive coated polymers or a combination of small liposomes containing functionalized pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) that act as thermal energy contrast agents are administered intravenously, intra-arterially, locally, intra-arterially, targeting multiple cancers such as leukemia, glioblastoma with cell surface GPC2 or glioma, or retinoblastoma or ovarian cancer or breast cancer, triple-negative breast cancer cells, gastric cancer colon cancer, or melanomas, acute lymphocytic leukemia (ALL), etc.
  • ALL acute lymphocytic leukemia
  • contrast agents such as a liposome containing nanoshells, or nanocages containing, in addition, either a medication Wnt inhibitor, Avastin, aflibercept, ipilimumab nivolumab, or rock inhibitor in liposomal formulations, or buformin or phenformin, gemtuzumab or a dye or an ultrasound contrast agent such as PFCL to create a popping sound when heated under the control of the thermal energy delivery unit to reach the boiling point of the PFCL at temperature of 56 degrees C temperature recording it by a microphone indicating the 56 degrees C temperature has been achieved and to damage to the tumor cell, and not heat up the tissue further to limit thermal damage by heat spillover to the normal cells surrounding the tumor, and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value by proving the presence of a tumor and/or use the biomarkers to make a vaccine for the future management of the patient recurrences.
  • contrast agents such as a liposome containing nanoshells
  • the antibody/medication and a-synuclein coated nanoparticle is conjugated with ACPP and a member of specific binding pair, a cellular receptor-agonist or antagonist pair, an enzyme-substrate pair, and/or an antibody- antigen pair, a virus, or virus like particles (VLP) or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators and Wnt inhibitor or Rock inhibitor with or without macrolides, such as cyclosporine A, mycophenolic acid, tacrolimus or ascomycin, is injected with monoclonal antibodies against the desired cancer along with a thermosensitive polymer, such as chitosan, carrying an antineoplastic medication, etc.
  • VLP virus like particles
  • monoclonal antibodies IL-2
  • bee toxins or other immune stimulators and Wnt inhibitor or Rock inhibitor with or without macrolides, such as cyclosporine A, mycophenolic acid, tacrolimus or ascomycin, is injected with monoclon
  • unquenched fluorescein or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC to be exposed to thermal energy releasing the medications at the temperature of 41-43 degrees C as evidenced by the release of the dye in the circulation to enhance the patient' s immune response to the tumor such as glioblastoma, melanoma, ocular melanoma, metastatic melanoma, breast cancer, ovarian cancer, etc. and induce an enhanced immune therapy.
  • the antibody coated nanoparticle is conjugated with ACPP and a virus, or virus like particles (VLP), or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11, and Wnt inhibitors or Rock inhibitors, and is injected with a monoclonal antibody against the desired cancer along with a thermosensitive polymer, such as chitosan, carrying an antineoplastic medication and unquenched fluorescein, or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC to be exposed to thermal energy releasing the medications, etc.
  • VLP virus, or virus like particles
  • the dye in the circulation to enhance the patient's immune response to the tumor such as glioblastoma, melanoma, ocular melanoma, metastatic melanoma, breast cancer, ovarian cancer, prostate, lung cancer, etc. along with enhanced immune therapy.
  • the antibody coated nanoparticle is conjugated with ACPP and with a member of a specific binding pair, such as a streptavidin-biotin pair, a cellular receptor-agonist or antagonist pair, an enzyme-substrate pair, and/or an antibody- antigen pair.
  • the antibody coated nanoparticle may be associated with a virus, e.g., a modified virus, Imlygic, Telomelysin, VCP inhibitor, or virus-like particles (VLP) made from modified plant viruses or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11.
  • VLP plant viral capsids or membranes lacking the genetic components of the virus and a Wnt inhibitor or Rock inhibitor is injected with monoclonal
  • thermosensitive polymer such as chitosan, carrying an antineoplastic medication and unquenched fluorescein, or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC to be exposed to thermal energy releasing the medications, etc. at the temperature of 41-43 degrees C as evidenced by the release of the dye in the circulation to enhance the patient' s immune response to the tumor, such as glioblastoma, melanoma, ocular melanoma, metastatic melanoma, breast cancer, ovarian cancer, prostate, lung cancer, etc.
  • a thermosensitive polymer such as chitosan, carrying an antineoplastic medication and unquenched fluorescein, or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC to be exposed to thermal energy releasing the medications, etc. at the temperature of 41-43 degrees C as evidenced by the release of the dye in the circulation to enhance the patient' s immune response to the tumor, such as
  • the antibody coated nanoparticle is conjugated with ACPP and with a virus, e.g., a modified virus, a tumoricidal virus, and/or an adeno-associated virus (AAV) or virus like particles (VLP) made from modified plant viruses or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11.
  • a virus e.g., a modified virus, a tumoricidal virus, and/or an adeno-associated virus (AAV) or virus like particles (VLP) made from modified plant viruses or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-L1, CTLA-4, Jagged 1 inhibitor 15D11.
  • a virus e.g., a modified virus, a tumoricidal virus, and/or an
  • VLP plant viral capsids or membranes lacking the genetic components of the virus, and thus unable to multiply in the organism
  • a Wnt inhibitor or Rock inhibitor is injected with monoclonal antibodies against the desired cancer along with a thermosensitive polymer, such as chitosan, carrying an antineoplastic medication and unquenched fluorescein or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC to be exposed to thermal energy releasing the medications etc.
  • glioblastoma such as glioblastoma, melanoma, ocular melanoma, metastatic melanoma, breast cancer, ovarian cancer, prostate, lung cancer, etc.
  • ivermectin causes an increase cell permeability makes the cell more susceptible to up take of chemotherapy, while addition of Rock inhibitors and diphenylpyrazole reduce the side effects of inflammation in the immunotherapy and thermotherapy to prevent cytokine storm.
  • ivermectin given as a non-toxic dose orally causes an increase cell permeability of the tumor makes the tumor cell more susceptible to up take of chemotherapy, while addition of Rock inhibitors, such as Fasudil, orally and
  • diphenylpyrazole reduce the side effects of inflammation in the immunotherapy and thermotherapy to prevent cytokine storm.
  • rock inhibitors Fasudil HA- 1077 a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc. in liposomal preparation are administered systemically intravenously, intra-arterially locally, intra peritoneal, or in the cerebrospinal fluid with Biologic Response Modifiers using functionalized pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) coated with rock inhibitors, Wnt inhibitors, Temozolomide, Cetuximab in thermosensitive polymers to release the medication at the desired place at a desired time or combine them with standard anti-inflammatory agents, etc., such as Steroids, Dexamethasone NASIDs, etc. and deliver pluralities of nanoparticles (i.e., biodegradable or non-biodegradable
  • ACPPs activatable cell-penetrating peptides
  • nanoparticles administered systemically intravenously, intra-arterially locally, intra peritoneal or in the cerebrospinal fluid as thermal contrast agents to prevent a cytokine storm in cancer immune therapy and remove cytokines after the therapy by electrophoresis, plasmapheresis or plasma exchange, or ARF6 inhibition, etc. to prevent the cytokine storm.
  • monoclonal antibody therapies and other biologies all are conjugated with the antibody/medication coated pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs), other biologies include soluble receptors, cytokines, and natural cytokine antagonist administered in the thermosensitive polymeric coating of the functionalized nanoparticles systemically, intravenously, locally, intra-arterially, or in a body cavity to target the tumor cells precisely to prevent the cytokine storm.
  • ACPPs activatable cell-penetrating peptides
  • humanized and chimeric antibodies are administered via thermosensitive polymer coated antibody conjugated pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs) intravenously or subcutaneously, and include TNF-alpha inhibitors, anti-B lymphocyte therapy, IL-6 inhibitors, cytotoxic T-lymphocyte- associated protein 4 (CTLA-4) fusion inhibitors, and IL-2 receptor antagonists and target lanthionine synthetase C-like 2 (LANCL2) for treating autoimmune disease administered in the thermosensitive polymeric coating of the functionalized nanoparticle systemically, intravenously, intra-arterially, locally, intraperitoneally, or in the cerebrospinal fluid, and release them at a desired place with thermotherapy to prevent a cytokine storm.
  • ACPPs activatable cell-penetrating peptides
  • rock inhibitors Fasudil HA- 1077 a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc.
  • Infliximab administered in a thermosensitive polymeric coating of the functionalized nanoparticle with activatable cell-penetrating peptides (ACPPs) systemically, intravenously, intra-arterially, locally, orally, intraperitoneally, or in the cerebrospinal fluid and release them at a desired place with thermotherapy to prevent a cytokine storm.
  • ACPPs activatable cell-penetrating peptides
  • rock inhibitors Fasudil HA- 1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc. in liposomal preparation
  • ACPPs activatable cell-penetrating peptides
  • thermosensitive nanoparticles and Adalimumab which is a humanized antibody administered subcutaneously, and release them at a desired place with thermo-immunotherapy to prevent a cytokine storm and to enhance the effect of immunotherapy or medication by thermotherapy and prevent production of medication resistance in these cancers.
  • rock inhibitors Fasudil HA- 1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor a selective RhoA/Rho kinase (ROCK) inhibitor
  • Y-27632 a small molecule inhibitor of ROCK1 and ROCK2, etc. in a functionalized liposomal preparation
  • HA- 1077 rock inhibitors Fasudil
  • ROCK selective RhoA/Rho kinase
  • Y-27632 a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • rock inhibitors Fasudil HA- 1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor a selective RhoA/Rho kinase (ROCK) inhibitor
  • Y-27632 small molecule inhibitor of ROCK1 and ROCK2 with antibody/medication coated pluralities of nanoparticles conjugated with thermosensitive nanoparticles with activatable cell-penetrating peptides (ACPPs) and Golimumab
  • ACPPs activatable cell-penetrating peptides
  • Golimumab small molecule inhibitor of ROCK1 and ROCK2 with antibody/medication coated pluralities of nanoparticles conjugated with thermosensitive nanoparticles with activatable cell-penetrating peptides
  • ACPPs activatable cell-penetrating peptides
  • Golimumab Golimumab
  • rock inhibitors Fasudil HA- 1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc.
  • a liposomal preparation containing quenched fluorescein or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC with antibody coated pluralities of nanoparticles with activatable cell-penetrating peptides (ACPPs), conjugated with thermosensitive nanoparticles and Certolizumab pegol
  • dosing depends on the condition for which it is given, although in most cases it is given as a 400-mg weekly subcutaneous injection as needed and released at a desired place with thermo- immunotherapy to prevent a cytokine storm.
  • rock inhibitors Fasudil HA-1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor or Y-27632
  • small molecule inhibitor of ROCK1 and ROCK2, etc. Avastin, aflibercept, ipilimumab, and/or nivolumab in liposomal preparation containing quenched fluorescein or another dye or indicator/medication or bubble liposomes carrying fluorescein/gene which contain air pockets or nanoemulsions of PFC
  • Avastin, aflibercept, ipilimumab, and/or nivolumab in liposomal preparation containing quenched fluorescein or another dye or indicator/medication or bubble liposomes carrying fluorescein/gene which contain air pockets or nanoemulsions of PFC are administered with antibody coated nanoparticles with activatable cell-penetrating peptides (ACPPs), conjugated with thermosensitive nanoparticles
  • rock inhibitors in liposomal preparation containing fluorescein are administered systemically, intravenously, intra-arterially, locally, intraperitoneally, or in the cerebrospinal fluid, with antibody coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), conjugated with thermosensitive nanoparticles and the IL-6 inhibitor tocilizumab and released at a desired place with thermo-immunotherapy to prevent a cytokine storm.
  • CPPs cell-penetrating peptides
  • rock inhibitors such as Botox or botulinum toxin at picogram to one nanogram concentrations is given subcutaneously, locally, or less than one picogram concentration with antibody coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), conjugated with thermosensitive nanoparticles and the IL-6 inhibitor, tocilizumab, and released at a desired place with thermo-immunotherapy to prevent a cytokine storm.
  • CPPs cell-penetrating peptides
  • rock inhibitors in a liposomal preparation containing quenched fluorescein or another indicator or bubble liposomes carrying fluorescein which contain air pockets or nanoemulsions of PFC are administered systemically, intravenously, intra- arterially, locally, intraperitoneally, or in the cerebrospinal fluid, with antibody coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), conjugated with thermosensitive nanoparticles with cell-penetrating peptides (CPPs) and the CTLA-4 fusion inhibitor abatacept, ipilimumab, nivolumab, and they are released at a desired place with thermo-immunotherapy to prevent a cytokine storm, while preventing production of medication resistance in these cancers.
  • CPPs cell-penetrating peptides
  • CPPs thermosensitive nanoparticles with cell-penetrating peptides
  • rock inhibitors Fasudil HA- 1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor a selective RhoA/Rho kinase (ROCK) inhibitor
  • Y-27632 a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • CPPs cell-penetrating peptides
  • rock inhibitors such as Botox
  • at picogram concentrations are administered with antibody coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), conjugated with thermosensitive nanoparticles and Alkylating agents and ipilimumab, nivolumab, administered systemically intravenously, intra-arterially locally, intraperitoneally, or in the cerebrospinal fluid, and they are released at a desired place with thermotherapy at the temperature of 41-43 degrees C to prevent a cytokine storm.
  • CPPs cell-penetrating peptides
  • rock inhibitors Fasudil HA- 1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor a selective RhoA/Rho kinase (ROCK) inhibitor
  • Y-27632 a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • CPPs cell-penetrating peptides
  • CPPs cell-penetrating peptides
  • CPPs thermosensitive nanoparticles with cell- penetrating peptides
  • small molecule Wnt inhibitors and a biologic response modifier or calcineurin inhibitor and paclitaxel systemically intravenously, intra-arterially locally, intraperitoneally, or in the cerebrospinal fluid, and they are released at a desired place with thermotherapy at the temperature of 41-43 degrees C to reduce inflammatory processes.
  • rock inhibitors Fasudil HA-1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc. in a liposomal preparation containing fluorescein
  • CPPs cell-penetrating peptides
  • Canakinumab is approved for use in juvenile idiopathic arthritis administered systemically, intravenously, intra-arterially, locally, intraperitoneally, or in the cerebrospinal fluid, and they are released at a desired place with thermotherapy at the temperature of 41-43 degrees C to reduce inflammatory processes while prevent production of medication resistance in these cancers.
  • rock inhibitors are administered systemically, intravenously, intra-arterially, locally, intraperitoneally, or in the cerebrospinal fluid, with antibody coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), conjugated with thermosensitive nanoparticles and Gevokizumab with or without small molecule Wnt inhibitor and they are released at a desired place with thermotherapy and a temperature of 41- 43 degrees C.
  • CPPs cell-penetrating peptides
  • Rock inhibitors are administered systemically, intravenously, intra-arterially, locally, intraperitoneally, or in the cerebrospinal fluid, with antibody coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), conjugated with thermosensitive nanoparticles and Gevokizumab with or without small molecule Wnt inhibitor, and they are released at a desired place with thermotherapy and a temperature of 41-43 degrees C or the Rock inhibitors or Wnt inhibitors are given orally at a non-toxic dose once or can be repeated monthly if needed.
  • CPPs cell-penetrating peptides
  • rock inhibitors Fasudil HA-1077
  • a selective RhoA/Rho kinase (ROCK) inhibitor a selective RhoA/Rho kinase (ROCK) inhibitor
  • Y-27632 a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • thermo-immunotherapy in a liposomal preparation containing quenched fluorescein are administered with antibody coated nanoparticles with cell-penetrating peptides (CPPs), conjugated with thermosensitive nanoparticles and Alemtuzumab, which has been shown to be beneficial for use in B-cell chronic lymphocytic leukemia, and they are released at a desired place with thermo-immunotherapy at the temperature of 41-43 degrees C to reduce inflammatory processes while preventing production of medication resistance in these cancers because the cancer cells do not survive the hyperthermia.
  • CPPs cell-penetrating peptides
  • Alemtuzumab Alemtuzumab
  • rock inhibitors are administered systemically, intravenously, intra-arterially, locally, intraperitoneally, or in the cerebrospinal fluid with antibody coated nanoparticles, conjugated with thermosensitive nanoparticles with cell-penetrating peptides (CPPs) and with Sarilumab a monoclonal antibody that targets IL-6, preventing T- and B-cell activation and differentiation and releases them at a desired place with thermotherapy at the temperature of 41-43 degrees C to reduce inflammatory processes.
  • CPPs cell-penetrating peptides
  • rock inhibitors are administered systemically intravenously, intra-arterially locally, intraperitoneally, or in the cerebrospinal fluid with
  • thermosensitive nanoparticles conjugated with thermosensitive nanoparticles with cell-penetrating peptides (CPPs) and Sirolimus and release them at a desired place with thermotherapy at the temperature of 41-43 degrees C to reduce inflammatory processes while preventing production of medication resistance in these cancers because the cancer cells do not survive the hyperthermia.
  • CPPs cell-penetrating peptides
  • rock inhibitors in liposomal preparation containing quenched fluorescein are administered systemically intravenously, intra-arterially locally,
  • thermosensitive nanoparticles with cell-penetrating peptides (CPPs) and Interferon- a2a and release them at a desired place with thermotherapy at the temperature of 41-43 degrees C while preventing production of medication resistance in these cancers because the cancer cells do not survive the hyperthermia.
  • CPPs cell-penetrating peptides
  • radiolabeled antibodies such as Ibritumomab tiuxetan
  • a thermosensitive polymer such as chitosan
  • coating antibody coated nanoparticle to deliver the medication to the tumor cell systemically intravenously, intra- arterially locally, intraperitoneally, or in the cerebrospinal fluid and release them at a desired place with thermotherapy at the temperature of 41-43 degrees C to reduce side effect of tiuxetan and Ibritumomab.
  • thermosensitive polymer of the antibody/medication coated nanoparticles with ACPP, containing a radiolabeled medication is administered systemically intravenously, intra-arterially locally, intraperitoneally, or in the cerebrospinal fluid to damage the tumor cells as a combination thermoradiotherapy and reduce their side effect while preventing production of medication resistance in these cancers because the cancer cells do not survive the hyperthermia.
  • thermosensitive polymer of the antibody/medication coated pluralities of nanoparticles such as gold nanoparticles that enhance the effect of external x- ray or particle beam radiation contains mebendazole, taxol derivatives, an anti-CCR2 antibody results in blockade of radiation-induced monocytic myeloid-derived suppressor cells infiltration that create radiation resistance, and fibrous tissue scars, in addition Rock inhibitors, such as Fasudil etc, intra-arterially locally, in the body cavity given to reduce inflammatory process and scar tissue, and quenched fluorescein or another indicator with Wnt inhibitors administered systemically intravenously, intra-arterially locally,
  • thermotherapy intraperitoneally, or in the cerebrospinal fluid released with controlled thermotherapy when administered medication to damage the tumor cells as a combination thermoradiotherapy in glioblastoma, medulloblastoma, retinoblastoma, melanoma or other brain tumors, lung cancer acting on the microtubes of the cancer cells while preventing production of medication resistance in these cancers because the cancer cells do not survive the hyperthermia, etc.
  • thermosensitive polymer of the antibody/medication coated nanoparticles contains mebendazole, taxol derivatives, and quenched fluorescein or another indicator with Rock inhibitors, checkpoint inhibitors administered systemically,
  • thermotherapy intravenously, intra-arterially locally, intraperitoneally, or in the cerebrospinal fluid released with controlled thermotherapy when administered medication to damage the tumor cells as a combination thermoradiotherapy in glioblastoma, medulloblastoma, retinoblastoma, melanoma or other brain tumors, lung cancer acting on the microtubes of the cancer cells while preventing production of medication resistance in these cancers because the cancer cells do not survive the hyperthermia, etc.
  • one or multiple agents are used to treat a number of cancers alone with a monoclonal antibody conjugated with thermosensitive antibody coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), nanoshell or nanocage treatment with temperature sensitive polymers conjugated with the nanoparticles having ipilimumab, nivolumab, and/or temozolomide combined a small molecule Wnt inhibitor, ivermectin, or niclosamide along with paclitaxel, or mebendazole, VLP or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators along with checkpoint inhibitors, such as PD-1, PD-11, CTLA-4, Jagged 1 inhibitor 15D11 administered intravenously or intra-arterially by injection, systemically intravenously, locally,
  • CPPs cell-penetrating peptides
  • temozolomide combined a small molecule Wnt inhibitor, ivermect
  • thermotherapy with either alternating magnetic field or focused low power ultrasound, a laser and simultaneous release of a dye such as quenched fluorescein or indocyanine green from the nanoparticle 's thermosensitive polymer or magnetic nanoshells containing fluorescein and PFH under observation with MRI or focused ultrasonic units, a CT-scan, or PET-scan, and release the medication at a desired place with thermotherapy at the temperature of 41-43 degrees C or higher up to 60 or more degrees C as recorded by a microphone or an ultrasonic receiver, and imaged with a thermoacoustic imaging system for control of the temperature and to induce an enhanced immunotherapy.
  • a dye such as quenched fluorescein or indocyanine green
  • the focused low power ultrasound is combined with the alternating magnetic field to simultaneously heat and image the tissue while heating and killing the tumor at a lower time duration
  • the pluralities of nanoparticles with cell- penetrating peptides (CPPs) are coated with thermosensitive polymers for drug release at temp, of 41-43 degrees C, imaged with a thermoacoustic imaging system for control of the temperature and the popping sound of the cavitation sound is heard at the temperature of 56 degrees C killing the tumor cells releasing their antigenic content to stimulate immune response to remove all the tumor cells, thereby eliminating the need for cellular immune therapy, and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the patient.
  • CPPs cell- penetrating peptides
  • the focused ultrasound can be directed by a computer controlled head to the exact location and borders of the tumor mass, which may be marked by focused high power ultrasound to heat up and create bubbles after heating the nanoshell or nanocage filled with PFCL hexane to its transition temperature of 56 degrees C that can be recorded by an additional ultrasound receiver or a microphone as a cavitation sound creating a precise ablation of a tumor under ultrasonic observation and drug Wnt inhibitors and a Rho inhibitor or mebendazole, diphenylpyrazole, release to kill the tumor while dampening the post-therapy inflammation, thus enhancing the person's immune response cells eliminating the need for cellular immune therapy, and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the patient having
  • glioblastoma medulloblastoma, ovarian tumor, or breast cancer, retinoblastoma, glioma, etc.
  • a known Wnt inhibitor such as Ivermectin or mebendazole, conjugated with ipilimumab, nivolumab, and thermosensitive polymer antibody coated nanoparticles, nanocages, nanoshells having the temperature markers fluorescein and PFCL intravenously, intra-arterially locally, or in the cerebrospinal fluid systemically intravenously, locally, intraperitoneally to attach to the tumor receptors and heated with either an alternating magnetic field, or ultrasound or microwave to heat up the tumors and release the medication under control of the temperature or 41-43 degrees C, then 56 degrees C, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy releasing more biomarkers from the tumor to enhance immune response cells by releasing increased tumor biomarkers in the circulation and eliminating the need for cellular immune therapy, and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e.,
  • a known Wnt inhibitor such as Ivermectin and taxol or other medication conjugated with thermosensitive polymer antibody/ medication coated nanoparticles, nanocages, nanoshells having the temperature markers quenched fluorescein or another indicator and PFCL intravenously, intra-arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, or focused low power ultrasound or microwave to heat up the tumors and release the medication under control of the temperature and a processor connected to thermal energy delivery system and thermal imaging unit to release the medication and kill the tumor simultaneously at a defined thermal energy temperature, and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the patient and production of vaccine for the patient's tumor and administering it with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fa
  • a small molecule Wnt inhibitor and taxane are conjugated with a thermosensitive polymer having ipilimumab and/or nivolumab, antibody coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein and PFCL intravenously, intra- arterially locally, in the bladder, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, low power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future of the patient and production of vaccine for the patient's tumor and administering it with checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wn
  • a known Wnt inhibitor Ivermectin PEGylated lipid nanoemulsion conjugated along with the Rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, a small molecule inhibitor of ROCK1 and ROCK2, etc. and decitabine with thermosensitive polymer antibody coated nanoparticles, nanocages, or nanoshells having the temperature markers fluorescein and PFCL
  • thermoacoustic imaging system for control of the temperature to reduce the production of inflammatory cytokines by macrophages and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the patient and production of vaccine for the patient's tumor and administering it with VLP and checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide to enhance immunotherapy, while Rock inhibitors and Wnt inhibtors reduce TGF- ⁇ production after therapy and the subsequent scar inflammation and formation.
  • VLP and checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide to enhance immunotherapy
  • Rock inhibitors and Wnt inhibtors reduce TGF- ⁇ production after therapy and the subsequent scar inflammation
  • Ivermectin the Wnt/ -catenin pathway prepared as PEGylated lipid nanoemulsion conjugated with a monoclonal antibody and ipilimumab or nivolumab and coated pluralities of nanoparticles with cell-penetrating peptides (CPPs), nanoshells or nanocages administered systemically intravenously, intra-arterially, locally, or in the cerebrospinal fluid and other body cavities or tissues through lymphatic vessels to reach the lymph nodes to attach to the tumor receptors and heated with either an alternating magnetic field, low power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature, imaged with a thermoacoustic imaging system for control of the temperature, a processor connected to the thermal delivery unit to release the medication at about 41-43 degrees C when the lipid melts targeting multiple cancers, such as leukemia, glioblastoma, glioma, or ovarian cancer or breast
  • CPPs cell-pen
  • thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the tumor recurrences in the patient and production of vaccine for the patient' s tumor and administering it with antibody coated VLP, checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, etc to enhance immune therapy while reducing inflammation.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil
  • Wnt inhibitors such as niclosamide
  • a known Wnt inhibitor and Wnt-TCF pathway Ivermectin the Wnt/ -catenin pathway prepared as PEGylated lipid nanoemulsion conjugated and a rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc. are conjugated with a thermosensitive polymer antibody coated nanoparticles, nanocages, or nanoshells having the temperature markers fluorescein and PFCL intravenously, intra-arterially, locally, or is the cerebrospinal fluid, etc.
  • the oral dose for ivermectin is about 40-150 microgram/kg, once in 7 days and the systemic dose is (10-100 ng/ml) in 1-5 (ml) in physiologic solution, or more doses as needed, and the oral dose of Fasudil is 40 mg-80 mg and the systemic dose is 50 nanograms to a few micrograms.
  • a small molecule Wnt inhibitor PKF118-3 lOthe Wnt/ -catenin pathway inhibitor and fasudil a rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, small molecule inhibitor of ROCK1 and ROCK2, etc.
  • thermosensitive polymer antibody coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein or another indicator and PFCL administered intravenously, intra-arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, focused ultrasound, or microwaves to heat up the tumors and release the medication and/or gemtuzumab under control of the temperature, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the leukemic cells, inducing leukemic cell death at a low micro molar concentration of Ivermectin or other hematological malignancies simultaneously by thermal energy and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers vaccine for the future management of the tumor recurr
  • a small molecule Wnt inhibitor PKF118-3 lOthe Wnt/ -catenin pathway inhibitor and fasudil a rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • thermosensitive polymer antibody coated pluralities of nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein or another indicator and PFCL administered intravenously, intra-arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, focused ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the tumor cells and starve those that might escape treatment preventing their important glucose metabolism.
  • a small molecule Wnt inhibitor PKF118-310 the Wnt/ ⁇ - catenin pathway inhibitor and fasudil, a rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • thermosensitive polymer antibody/medication coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein or another indicator and PFCL administered intravenously, intra- arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, low power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the tumor cells and starve those that might escape treatment preventing their important glucose metabolism.
  • a known Wnt inhibitor Ivermectin and doxorubicin Ivermectin and doxorubicin
  • thermosensitive polymer antibody coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein or another indicator and PFCL conjugated with Niclosamide, Rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, a small molecule inhibitor of ROCK1 and ROCK2 and femformin, etc.
  • thermoacoustic imaging system for control of the temperature initially and a processor to control the temperature, imaged at 41-43 degrees C with the release of quenched fluorescein to release the medication and kill the tumor simultaneously by thermal energy subsequently at 50-56 degrees C with the low power focused ultrasound receiver showing the ultrasound cavitation at a boiling point of PFCL in triple negative breast cancer or in ovarian cancer, glioblastoma, melanoma, retinoblastoma, gastric cancer, etc.
  • the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP or oncolytic viruses, monoclonal antibodies, IL-2, bee toxins or other immune stimulators conjugated with antibody coated nanoparticles to attach to the potential tumor cells and administering it with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, while applying thermal energy to damage recurrence of the tumor and induce reactivation of the immune response to eliminate them and prevent the tumor's important glucose metabolism with metformin, buformin, while antibody coated
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as nic
  • nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 ( to disguise themselves), and the tumor cells are recognized by the T cells, which together with killer cells phagocytose them, and enhance the immune response to the tumor and its exosomes and the circulating cells.
  • a checkpoint protein such as PD-L1 ( to disguise themselves)
  • T cells which together with killer cells phagocytose them, and enhance the immune response to the tumor and its exosomes and the circulating cells.
  • a known Wnt inhibitor, Ivermectin and doxorubicin is a known Wnt inhibitor, Ivermectin and doxorubicin
  • thermosensitive polymer antibody coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein or another dye or indicator and PFCL conjugated with Niclosamide, Rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, a small molecule inhibitor of ROCK1 and ROCK2 and femformin, etc.
  • thermotherapy has an important diagnostic value (i.e., confirming the presence of a tumor) and therapeutic value as biomarkers for the future management of the patient with recurrences by making a vaccine using the biomarkers and VLP or oncolytic viruses conjugated with antibody coated nanoparticles and administering it with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such Fasudil, or Wnt inhibitors, such as niclosamide, to attach to the potential tumor cells while applying thermal energy to damage recurrence of the tumor and induce reactivation of the immune response to eliminate them and prevent the tumor's important glucose metabolism with metformin, buformin and syrosingopine or cinnamaldehyde.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such Fasudil, or Wnt inhibitors, such as niclosamide
  • a known Wnt inhibitor, Ivermectin, Niclosamide, metformin, or buformin are conjugated with thermosensitive polymer antibody/medication coated nanoparticles, nanocages, nanoshells having the temperature markers quenched fluorescein or another indicator and PFCL intravenously, intra-arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, high power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature of 41-43 degrees C or 50-56 degrees C, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e. indicating presence of a tumor) and therapeutic value as biomarkers for the future management of the
  • a known Wnt inhibitor, niclosamide and taxol or
  • daunorubicin, ara-C or cytarabine or metformin, buformin or phenformin with or without syrosingopine are conjugated with a thermosensitive polymer antibody coated nanoparticles and ACPP, nanocages or nanoshells having the temperature markers quenched fluorescein and PFCL intravenously, intra-arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer, gastric cancer, colon cancer, or melanomas, Mantle cell lymphoma preferentially killing the cancer stem cells, etc.
  • thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP attached to antibody coated nanoparticles and administering it with checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, with cell-penetrating peptides (CPPs) for the management of the future recurrences in the patient and eliminating the need for CAR-T administration to the patient, and preventing their important glucose metabolism with metformin, buformin or phenformin while antibody coated nanoparticles/checkpoint inhibitors and VLP attach to the localized or circulating tumor cells and their exosomes, which are carrying a checkpoint protein, such as PD-L1 (to disguise themselves), and the tumor cells are recognized by the T cells, which together with killer cells phagocytose them, and enhance the immune response to
  • checkpoint inhibitors such as PD-1
  • a known Wnt inhibitor, niclosamide and taxol or
  • daunorubicin, ara-C or cytarabine or metformin, buformin or phenformin with or without syrosingopine are conjugated with a thermosensitive polymer antibody/medication coated nanoparticles and ACPP, nanocages or nanoshells having the temperature markers quenched fluorescein and PFCL intravenously, intra-arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, low power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy, imaged with a thermoacoustic imaging system for control of the temperature in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer, gastric cancer, colon cancer, or melanomas, Mantle cell lymphoma preferentially killing the cancer stem cells, etc.
  • thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP or oncolytic viruses attached to antibody coated nanoparticles and administering them with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil or Wnt inhibitors, such as niclosamide, with cell-penetrating peptides (CPPs) for the management of the future recurrences in the patient and eliminating the need for CAR-T administration to the patient, and preventing their important glucose metabolism with metformin, buformin or phenformin and syrosingopine.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil or Wnt inhibitors, such as niclosamide
  • CPPs cell-penetrating peptides
  • a known Wnt inhibitor, Niclosamide and VLP, and checkpoint inhibitors and metformin, or buformin are conjugated with a thermosensitive polymer antibody coated nanoparticles with ACPP, nanocages, or nanoshells having the temperature markers quenched fluorescein or another indicator and PFCL intravenously, intra- arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, high power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature of 42 to 43 degrees C and, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer and the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e.
  • biomarkers for vaccine production with or without VLP or oncolytic viruses attached to antibody coated nanoparticles, and administering them with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, with cell- penetrating peptides (CPPs) for the management of the future recurrences in the patient and eliminating the need for CAR-T administration to the patient and preventing their important glucose metabolism at the tumor site and enhance immunotherapy.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil
  • Wnt inhibitors such as niclosamide
  • the Wnt inhibitor ivermectin
  • the Wnt inhibitor may be given separately orally or systemically at low concentration of 1-90 nanograms/ml with thermotherapy of the glioblastoma, breast, ovarian, prostate cancer, lymphoma, leukemia along with thermotherapy using antibody/medication coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein and PFCL intravenously, intra- arterially locally, intra- arterially locally, in the body cavity such as bladder , mouth, nasal cavity, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, low power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature, imaged with a thermoacoustic imaging system for control of the temperature, and a processor to release the medication and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer and the increased tumor bio
  • the treatment can be repeated in weekly or monthly intervals until the cancer disappears.
  • a known Wnt inhibitor, niclosamide, and rituximab are conjugated with thermosensitive polymer antibody/medication coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein and PFCL intravenously, and checkpoint inhibitors, Rock inhibitors, such as Fasudil, etc., intra-arterially locally, in the body cavity, such as in the bladder, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, focused ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer, gastric cancer colon cancer, or melanomas, bladder or prostate cancer, etc.
  • a known Wnt inhibitor Niclosamide and rituximab are conjugated with thermosensitive polymer antibody / medication coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein and PFCL intravenously, and checkpoint inhibitors, but Rock inhibitors, such as Fasudil, etc., given orally or locally, intra-arterially locally, in the body cavity, such as in the bladder, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, high power focused ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer, gastric cancer colon cancer, or melanomas, bladder or prostate cancer, etc.
  • a known Wnt inhibitor is conjugated with rock inhibitor Fasudil (HA- 1077), a selective RhoA/Rho kinase (ROCK) inhibitor, or Y-27632, a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • rock inhibitor Fasudil HA- 1077
  • ROCK selective RhoA/Rho kinase
  • Y-27632 a small molecule inhibitor of ROCK1 and ROCK2, etc.
  • thermosensitive polymer antibody coated nanoparticles, nanocages, or nanoshells having the temperature markers quenched fluorescein or other dyes or indicators and PFCL intravenously, intra-arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, low power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication targeting multiple signaling pathways (NF- ⁇ , Wnt/ -catenin, Notch, ROS, mTORCl, and Stat3 and kill the tumors simultaneously by thermal energy in the treatment of glioblastoma or glioma, or ovarian cancer or breast cancer, gastric cancer, colon cancer, or melanomas etc.
  • NF- ⁇ , Wnt/ -catenin, Notch, ROS, mTORCl, and Stat3 to kill the tumors simultaneously by thermal energy in the
  • thermotherapy has an important diagnostic (i.e. indicating presence of a tumor) and therapeutic value as biomarkers in vaccine production along with antibody coated nanoparticles conjugated with VLP, and administering them with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc. for enhanced localized immunotherapy and Rock inhibitors, such as Fasudil or Wnt inhibitors, such as niclosamide, for the future management of the recurrences in patients and reduce the cytokine storm.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil or Wnt inhibitors, such as niclosamide
  • ivermectin or a small molecule Wnt inhibitor PKF118-310 or Curcumin, and doxycycline derivatives, or vitamin D are conjugated with thermosensitive polymer antibody/medication coated nanoparticles with CPP, nanocages, or nanoshells having the temperature markers quenched fluorescein or other dyes or indicators and PFCL intravenously, intra-arterially, locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, high power ultrasound, or microwaves to heat up the tumors and release the medication under control of the temperature 43 degrees C to 50-56 degrees C temperature, imaged with a thermoacoustic imaging system for control of the temperature and a processor to release the medication and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer, colon cancer, prostate cancer, or breast cancer, mammary stem cells cancers and the increased tumor biomarkers in the circulation after
  • TNF tumor necrosis factor
  • interleukin-6 granulocyte colony-stimulating factor
  • interleukin- ⁇ interleukin-17
  • IVIG Intravenous immune globulin
  • niclosamide, a salicylamide targets the Wnt/p-catenin pathway prepared as a PEGylated lipid nanoemulsion conjugated with carboplatin, monoclonal antibody, metformin, buformin or phenformin coated pluralities of nanoparticles with cell- penetrating peptides (CPPs), nanoshells, or nanocages administered systemically
  • cancers such as leukemia, glioblastoma or glioma, or ovarian cancer, drug- resistant ovarian cancer, ovarian tumor-initiating cells (OTIC) or breast cancer, gastric cancer
  • thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP attached to antibody coated nanoparticles and administering them with, VLP, checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc. and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, with cell-penetrating peptides (CPPs) for the management the future recurrences in the patient and eliminating the need for CAR-T administration to the patient.
  • VLP checkpoint inhibitors
  • PD-1 checkpoint inhibitors
  • Jagged 1 inhibitor 15D11 Jagged 1 inhibitor 15D11
  • Rock inhibitors such as Fasudil
  • Wnt inhibitors such as niclosamide
  • Niclosamide targets the Wnt/p-catenin pathway prepared as PEGylated lipid nanoemulsion conjugated with carboplatin, or other
  • antineoplastic medications or temozolomide, monoclonal antibody coated pluralities of nanoparticles with cell -penetrating peptides (CPPs), nanoshells, or nanocages administered systemically intravenously, intra-arterially locally, in the body cavity, such as in the bladder, mouth, nasal cavity, intraperitoneally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, focused ultrasound, or microwaves to heat up the tumors and release the medication under control of the
  • thermoacoustic imaging system for control of the temperature with a processor connected to the thermal delivery and thermal imaging unit to release the medication at about 41-43 degrees C when the lipid melts targeting multiple cancers such as leukemia, glioblastoma or glioma, or ovarian cancer, drug-resistant ovarian cancer, ovarian tumor-initiating cells (OTIC) or breast cancer, gastric cancer, colon cancer, or melanomas, etc.
  • OTIC ovarian tumor-initiating cells
  • thermotherapy has an diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP attached to antibody coated nanoparticles, and administering them with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc., and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, with cell- penetrating peptides (CPPs) for the management the future recurrences in the patient and eliminating the need for CAR-T administration to the patient.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil
  • Wnt inhibitors such as niclosamide
  • niclosamide is given orally instead of intravenously at the dose of 1-2 grams or less for one day and repeated monthly as needed simultaneously with thermoimmunotherapy to achieve the killing of the tumors such as glioblastoma, ovarian cancer, breast cancer, or medulloblastoma, and the therapy is repeated as needed.
  • antibody coated nanoparticles conjugated with niclosamide is given intraperitoneally along with the thermoimmunotherapy as a cyclodextrin conjugate at
  • thermoimmunotherapy to achieve killing of the tumors, such as glioblastoma, ovarian cancer, breast cancer, medulloblastoma, and the therapy is repeated as needed.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil
  • Wnt inhibitors such as niclosamide
  • the vaccination may be repeated in weekly or monthly intervals, as needed, until no sign of the tumor metastasis is found demonstrated by disappearance of the blood biomarkers.
  • a known enzyme that dissolves the tumor cells' membrane and mitochondrial cell membrane is conjugated with a thermosensitive polymer antibody/medication coated nanoparticles and ACPP, gold, silica, magnetic, paramagnetic, and nonmagnetic nanoparticles, nanocages or nanoshells, with or without macrolides, such as cyclosporine A, mycophenolic acid, tacrolimus or ascomycin, having the temperature markers quenched fluorescein or other indicators and PFCL, bubble liposomes containing air pockets or nanoemulsions of PFC carrying fluorescein gene intravenously, intra- arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, focused ultrasound, or microwaves, radiofrequency to heat up the nanoparticle/tumors complex and release the medication under control of the temperature, and imaged with a thermoacoustic imaging system for control of the temperature and a processor
  • thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP attached to antibody/medication coated nanoparticles and administering them with checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc. and Rock inhibitor, Fasudil or Wnt inhibitors, such as niclosamide, with cell-penetrating peptides (CPPs) for the management of the future recurrences in the patient and eliminating the need for CAR-T administration to the patient.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc. and Rock inhibitor, Fasudil or Wnt inhibitors, such as niclosamide
  • CPPs cell-penetrating peptides
  • a known enzyme that dissolves the tumor cells' membrane and mitochondrial cell membrane e.g., granzyme, metalloproteinase (MMP), chymotrypsin, as pepsin, alpha chymotrypsin, or trypsin
  • macrolides such as cyclosporine A, mycophenolic acid, tacrolimus or ascomycin
  • a thermosensitive polymer antibody/medication coated nanoparticles and ACPP gold, silica, magnetic, paramagnetic, and nonmagnetic nanoparticles, nanocages or nanoshells having the temperature markers quenched fluorescein or other indicators and PFCL, or bubble liposomes containing air pockets or nanoemulsions of PFC carrying fluorescein, gene and intravenously, intra- arterially locally, or in the cerebrospinal fluid to attach to the tumor receptors and heated with either an alternating magnetic field, ultrasound, microwaves, or radiofrequency to heat up
  • MMP metalloproteinase
  • thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP attached to antibody/medication coated nanoparticles and administering them with checkpoint inhibitors, such as PD-1, Jagged 1 inhibitor 15D11, etc. and Rock inhibitors, such as Fasudil, or Wnt inhibitors, such as niclosamide, with cell-penetrating peptides (CPPs) for the management of the future recurrences in the patient and eliminating the need for CAR-T administration to the patient.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil, or Wnt inhibitors, such as niclosamide
  • CPPs cell-penetrating peptides
  • a known enzyme that dissolves the tumor cells' membrane and mitochondrial cell membrane e.g., granzyme, metalloproteinase (MMP), chymotrypsin, as pepsin, alpha chymotrypsin, or trypsin
  • MMP metalloproteinase
  • chymotrypsin as pepsin, alpha chymotrypsin, or trypsin
  • Rock inhibitors such as Fasudil, etc.
  • thermoacoustic imaging system for control of the temperature and a processor to release the medication dye at 41-43 degrees C and kill the tumor simultaneously by thermal energy in treatment of glioblastoma or glioma, or ovarian cancer or breast cancer, gastric cancer, colon cancer, or
  • thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP attached to antibody coated nanoparticles and administering them with checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc. and Rock inhibitors, such as Fasudil or Wnt inhibitors, such as niclosamide, with cell-penetrating peptides (CPPs) for the management of the future recurrences in the patient and eliminating the need for CAR-T administration to the patient.
  • checkpoint inhibitors such as PD-1, Jagged 1 inhibitor 15D11, etc.
  • Rock inhibitors such as Fasudil or Wnt inhibitors, such as niclosamide
  • CPPs cell-penetrating peptides
  • the pluralities of antibody/medication coated nanoparticles including piezoelectric nanoparticles carry with their thermosensitive polymeric coating one or multiple antineoplastic medications, checkpoint inhibitors, Perforin, cytolysins, complement component 9(C9) granzyme, VLP etc.
  • thermoacoustic imaging system for control of the temperature, along with low-intensity (1-3 V/cm), intermediate frequency (100-300 kHz), electric fields achieving thermotherapy, electroacoustic imaging and dielectric effect on the cellular components of the tumors, such as brain tumors, lung cancer, ovarian cancer, breast cancer, prostate cancer, and other cancers, which can be repeated numerous times post therapy to eliminate the cancer with simultaneous aspiration of a lysed tumor mass through a 20-30 gauge needle to reduce the toxic burden on the body while the increased tumor biomarkers in the circulation after the thermotherapy has an important diagnostic (i.e., indicating presence of a tumor) and therapeutic value as biomarkers for vaccine production with or without VLP attached to antibody coated nanoparticles, and administering them with checkpoint inhibitors such as PD-1, Ja
  • CPPs cell-penetrating peptides
  • tocilizumab a monoclonal antibody that targets interleukin-6 and ornithine phenylacetate an ammonia scavenger, or ARF6 inhibition for treatment of hepatic encephalopathy, a neuropsychiatric syndrome associated with hyperammonemia.
  • a known enzyme that dissolves the wall of the bacteria, viruses, fungi, parasites such as MRSA, granzyme, metalloproteinase (MMP), chymotrypsin, as pepsin, alpha chymotrypsin, trypsin conjugated with a thermosensitive polymer antibody/medication coated nanoparticles of iron, iron oxide, gold, silica, magnetic, paramagnetic, and nonmagnetic nanoparticles, nanocages or nanoshells having the temperature markers quenched fluorescein or other indicators or with Liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying a liposome opens a channel through which the drug is released at the temperature or 41-43 degrees C or opens drug-permeable pores created bubble formation from the decomposition of encapsulated ammonium bicarbonate injected intravenously, intra- arterially, locally, or in the cerebrospinal fluid
  • tocilizumab a monoclonal antibody that targets interleukin-6 and ornithine phenylacetate an ammonia scavenger, for treatment of hepatic encephalopathy, a neuropsychiatric syndrome associated with hyperammonemia or ARF6 inhibition.
  • thermal and electrical energy is used to influence the permeability of the tumor cell membrane so as to utilize the effect or thermotherapy with pluralities of nanoparticles or liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the drug is released at the temperature or 41-43 degrees C or opens drug-permeable pores created bubble formation from the decomposition of encapsulated ammonium bicarbonate injected in the body to enhance the effect of chemotherapy or immune therapy, gene therapy, gene modification using antibody coated nanoparticles conjugated with CRISPR-cas9 in malignant cells, such as brain and spinal cord tumors, breast cancer, lung cancer, prostate and ovarian cancer and melanoma, glioblastoma,
  • retinoblastoma retinoblastoma, meduloblastoma, gastrointestinal and genitourinary tumor of sarcoma, or benign expanding tumor, such as memningioma, uterus fibroma, etc.
  • benign expanding tumor such as memningioma, uterus fibroma, etc.
  • a DNA-cutting Cas9 enzyme targets the checkpoint inhibitor regions removing checkpoint inhibitors, but Rock inhibitors, such as Fasudil, etc.
  • HS-TK thymidine kinase
  • cytosine deaminase etc.
  • pro-apoptatic genes etc.
  • antibody coated nanoparticles conjugated with CRISPR cas9 mediated Homology - Independent Targeted Integration (HITI) or Homology Directed Repair (HDR) further enhancing an immune response which is not being blocked by the checkpoint inhibitors of the tumor cells.
  • Antibody coated nanoparticles are used having pyroelectric or piezoelectric characteristics, and the antibody coated nanoparticle tube complex is imaged with an electroacoustic computed tomography imaging system.
  • the antibody/medication coated pluralities of gold, piezoelectric, iron oxide, nanocage, nanotube, nanoshell, magnetic, paramagnetic, pyroelectric and/or piezoelectric nanoparticles are coated with thermosensitive polymers, or with liposomes having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the drug is released at the temperature or 41-43 degrees C or opens drug-permeable pores created bubble formation from the decomposition of encapsulated ammonium bicarbonate or polymers, such as chitosan, quenched with fluorescein or other indicators conjugated with antineoplastic medication, gene, CRISPR-cas9 conjugated with cell penetrating agents or activatable cell penetrating agents injected in the patient's circulation, lymphatic vessels, inside a body cavity, cerebrospinal fluid, etc.
  • thermosensitive polymers or with liposomes having temperature triggered opening of a leucine zipper peptid
  • an acoustic response is produced by electrical stimulation of piezoelectric nanoparticle inside the body that is called an electroacoustic sound or signal which can be recorded with a transducer, or microphone, converted to an electrical signal and forwarded to a processor to be converted to a ID, 2D
  • HITI Homology -Independent Targeted Integration
  • HDR Homology Directed Repair
  • thermoacoustic or photoacoustic imaging system for control of the temperature.
  • thermal and electrical energy is used to influence the permeability of the tumor cell membrane so as to utilize the effect or thermotherapy to enhance the effect of chemotherapy or immune therapy, gene therapy, gene modification using CRISPR-cas9 in malignant cells, such as brain and spinal cord tumors, breast cancer, lung cancer, prostate and ovarian cancer and melanoma, glioblastoma, retinoblastoma, meduloblastoma, gastrointestinal and genitourinary tumor of sarcoma, or benign expanding tumors, such as meningioma, uterus fibroma, etc., based on the antibody coated nanoparticles conjugated with CRISPR/Cas9 system, in which a DNA-cutting Cas9 enzyme or combination with antibody/medication coated nanoparticles or with liposomes injected in the body having temperature triggered opening of a leucine zipper peptide inserted in the membrane of a dye and medication carrying liposome opens a channel through which the drug
  • thermal and electrical energy is used to influence the permeability of the tumor cell membrane so as to utilize the effect or thermotherapy to enhance the effect of chemotherapy or immune therapy, gene therapy, gene modification using antibody coated nanoparticles having pyroelectric or piezoelectric characteristics, and the nanoparticle tube complex is imaged with an electroacoustic computed tomography imaging system and conjugated with CRISPR-cas9 mediated Homology -Independent Targeted Integration (HITI) or Homology Directed Repair (HDR) further enhancing an immune response which is not being blocked by the checkpoint inhibitors in malignant cells, such as brain tumors and spinal cord tumors, breast cancer, lung cancer, prostate and ovarian cancer and melanoma, glioblastoma, retinoblastoma, meduloblastoma, gastrointestinal and genitourinary tumor of sarcoma, or benign expanding tumors, such as meningioma, uterus fibroma, etc., based on
  • the systemic administration of antibody coated nanoparticles conjugated with CRISPR Cas9 cells eliminating the body's checkpoint inhibitor of the immune responsive cells, using antibody/medication coated nanoparticles or liposomes in treatment of severe autoimmune diseases involving the digestive tract, central nervous system, such as multiple sclerosis and Gillian-barre syndrome or respiratory diseases, such as asthma and arthritis, giant cell arthritis and arteriosclerosis, etc.
  • Rock inhibitors such as Fasudil or other small molecule Rho kinase inhibitors or GSK 269962 inhibitor
  • Wnt inhibitors such as ivermectin, niclosamide, apicularen and bafilomycin
  • antibody coated nanoparticles are conjugated with non-toxic dose of Rock inhibitors, Wnt inhibitors, combined with macrolide immunosuppressants such as cyclosporine, mycophenolic acid, tacrolimus, ascomycin etc. to suppress an excessive immune response and inflammation in the normal tissue by blocking the IL-17 from the memory Thl7 cells with the existing severe immune response in the skin, gut, joint, etc.
  • Rock inhibitors Wnt inhibitors
  • macrolide immunosuppressants such as cyclosporine, mycophenolic acid, tacrolimus, ascomycin etc.
  • the use of antibody coated pluralities of nanoparticles can be conjugated with Rock inhibitors, Wnt inhibitors and GSK 269962 inhibitors along with antibody coated nanoparticles with cyclosporine A, or mycophenolic acid or tacrolimus etc. to significantly damage the tumor cells and their metastatic lesion with or without thermotherapy.
  • the treatment of a tumor with a metastatic lesion is done with simultaneous administration of pluralities of the antibody coated nanoparticles conjugated with various medications such as Wnt inhibitors, GSK inhibitors, or Rock inhibitors with macrolide antibodies such as cyclosporine A, Ascomycin, mycophenolic acid or tacrolimus, anti VEGFs such as Avastin, etc. and simultaneous administration of checkpoint inhibitors, such as PD-1, PD-Ll, CTLA-4, Jagged 1 inhibitor 15D11, etc. and immune stimulators, such as VLP , IL-2, IL-4 TLR7 etc.
  • various medications such as Wnt inhibitors, GSK inhibitors, or Rock inhibitors with macrolide antibodies such as cyclosporine A, Ascomycin, mycophenolic acid or tacrolimus, anti VEGFs such as Avastin, etc. and simultaneous administration of checkpoint inhibitors, such as PD-1, PD-Ll, CTLA-4, Jagged 1 inhibitor 15D11, etc. and immune stimul
  • kidney dialysis is done to remove the dead normal cells and hybrid cells and exosomes, and toxins, etc. from the blood that can still cleaned and repurposed to the patient.
  • the antibody obtained from the patient's body or in tissue culture from the hybrid cells are used to coat the nanoparticles conjugated with CRSPR cas9 and with cytotoxic siRNA to damage the hybrid tumor cell.
  • the nanoparticles carry both the tumor antibody and the hybrid cell antibody to deliver cytotoxic siRNA to both kinds of the cells and protect the normal cells.
  • this procedure is repeated with simultaneous thermotherapy with an external source of energy to damage these cells, and the treatment is repeated weekly or monthly along with either Rock inhibitors, Wnt inhibitors, or GSK 269962 inhibitor lithium along with antibody coated nanoparticles to inhibit GSK and repeated until the hybrid cells or tumor cells are not found from the patient's blood samples.
  • the combination therapy for the late metastatic disease may produce an autoimmune response.
  • the gene therapy is combined with Rock inhibitors, etc. and macrolides to reduce post-treatment immune and inflammatory responses.
  • the pluralities of antibody coated nanoparticles are conjugated with Rock inhibitors, Wnt inhibitors, GSK inhibitors, anti-VEGFs, such as Avastin, NSAIDS such as Diclofenac, acetyls alicylic acid, aspirin, Lovastatins to block phosphoinositol activation as a results of hypoxia inducible factor- 1 which induces VEGF production.
  • HIF1A induces Myelodysplastic Syndromes which can be treated with antibody coated nanoparticles conjugated with Wnt inhibitors, GSK269962, inhibitor and anti-VEGFs.
  • CDK12 cyclin-dependent Kinase 12
  • ADP]-ribose poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitors or platinum chemotherapies such as cisplatin, carboplatin.
  • PARP poly(adenosine diphosphate [ADP]-ribose) polymerase
  • pluralities of antibody coated nanoparticles are conjugated with a polymer carrying Rock or Wnt of GSK inhibitors and PARP and or platinum chemotherapy and VLP or oncolytic viruses, anti-VEGFs and checkpoint inhibitors injected locally or systemically, intravenously or preferably intra-arterially, into the feeding artery, applying and exposing the nanoparticles to the tumor cells prior to their systemic uptake by the Reticuloendothelial cells (REC) applying optionally external thermotherapy, laser, LIFU, HIFU, alternating magnetic field in non-thermal or thermal frequencies while controlling the temperature with a thermal imaging unit and a software from 37 degrees C to 41 degrees C or more as needed, releasing the medication to kill damaged tumor cell and their exosomes or CTC or their exosomes.
  • REC Reticuloendothelial cells
  • cytotoxic T lymphocytes become exhausted and lose their normally present anti-tumor action and, in contrast, produce on their surface membrane inhibitory makers and angiogenetic factors, and cannot produce other anti-tumor chemokines, such as IFN- ⁇ or granzyme ⁇ thus, encouraging tumor resistance and development of metastatic disease or chemotherapy resistant tumor cells, release of IL- 1 activates NF- ⁇ encouraging metastatic disease and limits the use of immunotherapy with T Cell Receptor (TCR) or chimeric antigen receptor (CAR) in these patients.
  • TCR T Cell Receptor
  • CAR chimeric antigen receptor
  • the patient is treated with antibody coated nanoparticles conjugated with Rock inhibitors, such as Fasudil, etc. or Wnt inhibitors or GSK269962 inhibitors with or without macrolides and subsequent plasmapheresis or dielectrophoresis to remove toxins and dead cells from the patient' s body, and the clean blood can be re-infused to the patient.
  • the treatment is systemic, and in another embodiment, the treatment is done locally, or orally combined with anti-VEGF coated nanoparticles.
  • the pluralities of antibody coated nanoparticles are conjugated with Wnt inhibitors, Rock inhibitors or GSK inhibitors in combination with a MAP/ERK inhibitor such as sorafenib an RAF kinase inhibitor or Raf inhibitor sB 590885, PLX4720, RAF 265, XL281, ecorafenib, darafenib,vemurafenib or MEK inhibitors such as cobimetinib, Bimimetinib, selumetinib or Trametinib or GSK 112020212 along with an anti- VEGF, such as Avastin, and checkpoint inhibitors, and IL-2 ,VLP, or oncolytic viruses such as Reovirus, etc. in a thermosensitive polymeric coating to release the medication after thermotherapy at temperatures of 39 to 43 degrees C or more to damage the tumor cells but control the inflammatory process and prevent a cytokine storm formation or autoimmune response.
  • a MAP/ERK inhibitor such
  • the antibodies found on the regular tumor cell can be grown on the cancer stem cells (CSC), it may not be easy to target these cells.
  • the antibody coated nanoparticles are conjugated with a slow release polymer, such as polylactic or glycolic acid or porous silicon that can stay around the tumor cells or CSC and their vasculature to damage the genetic components of the cancer stem cells and their vascular supply, while carrying VLP or oncolytic viruses, checkpoint inhibitors and Rock inhibitors, Rho-kinases GSK 269962, potent and selective Rock inhibitor GSK 429286, Selective Rho- kinase (ROCK) inhibitor HI 152 dihydrochloride, Selective Rho-kinase (ROCK) inhibitor Glycyl H 1152 dihydrochloride, Selective Rho-kinase (ROCK) inhibitor; more selective analogue of HI 152, Cell-permeable, selective Rho-kinase inhibitor OXA 06 dihydrochloride, potent ROCK inhibitor
  • pluralities of radioactive functionalized nanoparticles are used to treat hybrid tumor cells in conjunction with an immunotherapy treatment method that administers nanoparticles containing a radioactive material to the body of a cancer patient.
  • the nanoparticles containing the radioactive material are conjugated with cell penetrating peptides (CPPs) or activatable cell penetrating peptides (ACPPs) for penetration and uptake into the hybrid tumor cells.
  • CPPs cell penetrating peptides
  • ACPPs activatable cell penetrating peptides
  • the radioactive material is disposed inside the hybrid tumor cells. Once inside the hybrid tumor cells, the radioactive material of the nanoparticles damages the genetic component of the hybrid tumor cells, thereby preventing the proliferation of the hybrid tumor cells and enabling the hybrid tumor cells to be destroyed.
  • viruses are causes of the development of the cancer, such as Merkel-cell carcinoma, papilloma virus, Herpes virus, creutzfeldt- Jakob disease, Epstein- Barr virus, that can produce both skin cancer or brain cancer, or may lead to
  • the prions can cause inflammation in the brain, the inflammatory process caused by the viruses, or prions activates p38 MAPK synaptotoxic signaling pathway and causes neuronal damage
  • Rock inhibitors control the inflammatory process and prevent cell death particularly contributing to neuronal regeneration and branching of the axons, similarly GSK-3 beta inhibition by Lithium causes spreading the cerebellar neurons and has implication in treatment of Alzheimer's disease, or Parkinson disease.
  • the Rock inhibitor or GSK inhibitors conjugated with polymeric nanoparticle such as polylactic or glycolic acid can be administered intranasally to travel to the brain via the olfactory nerve.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Rehabilitation Therapy (AREA)
  • Dispersion Chemistry (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Dermatology (AREA)
  • Medical Informatics (AREA)
  • Anesthesiology (AREA)
  • Pain & Pain Management (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Radiology & Medical Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des méthodes de traitement du cancer faisant appel à une thermothérapie et/ou à une immunothérapie améliorée. Dans un mode de réalisation, la méthode comprend les étapes consistant à administrer une pluralité de nanoparticules pour cibler une tumeur chez un patient, les nanoparticules étant revêtues d'un anticorps anti-tumoral, des peptides de pénétration cellulaire (CPP), et un polymère, et les nanoparticules contenant un médicament et/ou un gène, et un colorant ou un indicateur dans le revêtement polymère, au moins certaines des nanoparticules se liant à des antigènes de surface de cellules tumorales de sorte à former un complexe cellule tumorale/nanoparticule ; à exciter les nanoparticules à l'aide d'une source ultrasonore générant une onde ultrasonore de sorte à décoller le revêtement polymère des nanoparticules, libérant ainsi le colorant ou l'indicateur dans la circulation du patient et le médicament et/ou le gène au niveau du site tumoral ; et à imager une région corporelle du patient de sorte à détecter le colorant ou l'indicateur libéré dans la circulation du patient.
PCT/US2018/054880 2015-12-21 2018-10-08 Méthodes de traitement du cancer faisant appel à une thermothérapie et/ou à une immunothérapie améliorée WO2019071261A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US16/200,195 US11433260B2 (en) 2015-12-21 2018-11-26 Cancer treatment methods using thermotherapy and/or enhanced immunotherapy
US16/843,831 US11660229B2 (en) 2015-12-21 2020-04-08 Cancer treatment methods using thermotherapy and/or enhanced immunotherapy
US16/988,697 US20210002338A1 (en) 2015-12-21 2020-08-09 Cancer Treatment Methods Using Thermotherapy And/Or Enhanced Immunotherapy
US17/220,878 US20210220480A1 (en) 2015-12-21 2021-04-01 Cancer Treatment And Imaging Methods Using Thermotherapy And Drug Delivery
US17/902,814 US20230000981A1 (en) 2015-12-21 2022-09-03 Cancer Treatment Methods Using Thermotherapy And/Or Enhanced Immunotherapy

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201762569592P 2017-10-08 2017-10-08
US62/569,592 2017-10-08
US201762577485P 2017-10-26 2017-10-26
US62/577,485 2017-10-26
US201862614456P 2018-01-07 2018-01-07
US62/614,456 2018-01-07
US201862720258P 2018-08-21 2018-08-21
US62/720,258 2018-08-21

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/976,321 Continuation-In-Part US10136820B2 (en) 2015-12-21 2015-12-21 Method to visualize very early stage neoplasm or other lesions

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US14/976,321 Continuation-In-Part US10136820B2 (en) 2015-12-21 2015-12-21 Method to visualize very early stage neoplasm or other lesions
US16/200,195 Continuation-In-Part US11433260B2 (en) 2015-12-21 2018-11-26 Cancer treatment methods using thermotherapy and/or enhanced immunotherapy
US16/843,831 Continuation-In-Part US11660229B2 (en) 2015-12-21 2020-04-08 Cancer treatment methods using thermotherapy and/or enhanced immunotherapy

Publications (1)

Publication Number Publication Date
WO2019071261A1 true WO2019071261A1 (fr) 2019-04-11

Family

ID=65994363

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/054880 WO2019071261A1 (fr) 2015-12-21 2018-10-08 Méthodes de traitement du cancer faisant appel à une thermothérapie et/ou à une immunothérapie améliorée

Country Status (1)

Country Link
WO (1) WO2019071261A1 (fr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111632266A (zh) * 2020-06-08 2020-09-08 洛阳康贝生物工程有限公司 一种脑功能调控治疗***
CN112076319A (zh) * 2020-10-16 2020-12-15 福州大学 青蒿素及其衍生物在制备热动力治疗敏化剂中的应用
CN113164749A (zh) * 2019-11-15 2021-07-23 科利耳有限公司 全身性地循环的治疗物质的局部释放
WO2021224687A1 (fr) * 2020-05-08 2021-11-11 Novocure Gmbh Compositions et procédés d'application de champs électriques alternatifs à des cellules souches pluripotentes
CN113967256A (zh) * 2021-10-26 2022-01-25 山东大学 一种具有光热-化疗-免疫功能的纳米粒及其制备方法和应用
WO2022263984A1 (fr) * 2021-06-14 2022-12-22 Novocure Gmbh Méthodes de traitement et de prévention du cancer au moyen de champs électriques alternatifs, de particules radioactives et d'un traitement systémique
CN116524999A (zh) * 2023-07-03 2023-08-01 迈杰转化医学研究(苏州)有限公司 一种用于检测结直肠癌微小残留病灶的靶标集合的筛选方法及其检测***

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110287035A1 (en) * 2002-02-14 2011-11-24 Peyman Gholam A Method and composition for hyperthermally treating cells
US20150202466A1 (en) * 2009-10-12 2015-07-23 Kona Medical, Inc. Energetic modulation of nerves
US20160022976A1 (en) * 2002-02-14 2016-01-28 Gholam A. Peyman Method and composition for hyperthermally treating cells

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110287035A1 (en) * 2002-02-14 2011-11-24 Peyman Gholam A Method and composition for hyperthermally treating cells
US20160022976A1 (en) * 2002-02-14 2016-01-28 Gholam A. Peyman Method and composition for hyperthermally treating cells
US20150202466A1 (en) * 2009-10-12 2015-07-23 Kona Medical, Inc. Energetic modulation of nerves

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HUSSEINI ET AL.: "Ultrasonic-Activated Micellar Drug Delivery for Cancer Treatment", J PHARM SCI, vol. 98, no. 3, 27 May 2008 (2008-05-27), pages 795 - 811 *
KONG ET AL.: "Efficacy of Liposomes and Hyperthermia in a Human Tumor Xenograft Model: Importance of Triggered Drug Release", CANCER RESEARCH, vol. 60, 15 December 2000 (2000-12-15), pages 6950 - 6957, XP002760235 *
PHENIX ET AL.: "High Intensity Focused Ultrasound Technology, Its Scope and Applications in Therapy and Drug Delivery", JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES, vol. 17, no. 1, 31 March 2014 (2014-03-31), pages 136 - 153 *
TAYLOR ET AL.: "Glycogen Synthase Kinase 3 Inactivation Drives T-bet-Mediated Downregulation of Co-receptor PD-1 to Enhance CD 8+ Cytolytic T Cell Responses", IMMUNITY, vol. 44, no. 2, 16 February 2016 (2016-02-16), pages 274 - 286, XP029428481, DOI: doi:10.1016/j.immuni.2016.01.018 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113164749A (zh) * 2019-11-15 2021-07-23 科利耳有限公司 全身性地循环的治疗物质的局部释放
EP4057990A4 (fr) * 2019-11-15 2024-01-03 Cochlear Limited Libération localisée de substances thérapeutiques à circulation systémique
WO2021224687A1 (fr) * 2020-05-08 2021-11-11 Novocure Gmbh Compositions et procédés d'application de champs électriques alternatifs à des cellules souches pluripotentes
US11850421B2 (en) 2020-05-08 2023-12-26 Novocure Gmbh Compositions and methods of applying alternating electric fields to pluripotent stem cells
CN111632266A (zh) * 2020-06-08 2020-09-08 洛阳康贝生物工程有限公司 一种脑功能调控治疗***
CN111632266B (zh) * 2020-06-08 2024-03-01 洛阳康贝生物工程有限公司 一种脑功能调控治疗***
CN112076319A (zh) * 2020-10-16 2020-12-15 福州大学 青蒿素及其衍生物在制备热动力治疗敏化剂中的应用
WO2022263984A1 (fr) * 2021-06-14 2022-12-22 Novocure Gmbh Méthodes de traitement et de prévention du cancer au moyen de champs électriques alternatifs, de particules radioactives et d'un traitement systémique
CN113967256A (zh) * 2021-10-26 2022-01-25 山东大学 一种具有光热-化疗-免疫功能的纳米粒及其制备方法和应用
CN116524999A (zh) * 2023-07-03 2023-08-01 迈杰转化医学研究(苏州)有限公司 一种用于检测结直肠癌微小残留病灶的靶标集合的筛选方法及其检测***
CN116524999B (zh) * 2023-07-03 2023-09-19 迈杰转化医学研究(苏州)有限公司 一种用于检测结直肠癌微小残留病灶的靶标集合的筛选方法及其检测***

Similar Documents

Publication Publication Date Title
US11660229B2 (en) Cancer treatment methods using thermotherapy and/or enhanced immunotherapy
WO2019071261A1 (fr) Méthodes de traitement du cancer faisant appel à une thermothérapie et/ou à une immunothérapie améliorée
US20210220480A1 (en) Cancer Treatment And Imaging Methods Using Thermotherapy And Drug Delivery
US20210002338A1 (en) Cancer Treatment Methods Using Thermotherapy And/Or Enhanced Immunotherapy
Lan et al. Microneedles loaded with anti-PD-1–cisplatin nanoparticles for synergistic cancer immuno-chemotherapy
Yang et al. Thermal immuno-nanomedicine in cancer
US9393396B2 (en) Method and composition for hyperthermally treating cells
US20200390889A1 (en) Remotely triggered therapy
US9302087B2 (en) Method and composition for hyperthermally treating cells
US9320813B2 (en) Method and composition for hyperthermally treating cells
Day et al. Magnetic systems for cancer immunotherapy
US11260128B2 (en) Synergistic nanotherapy systems and methods of use thereof
US9289491B2 (en) Method and composition for hyperthermally treating cells
US8932636B2 (en) Method and composition for hyperthermally treating cells
US8668935B2 (en) Method and composition for hyperthermally treating cells
Sun et al. Ultrasound microbubbles mediated sonosensitizer and antibody co-delivery for highly efficient synergistic therapy on HER2-positive gastric cancer
US8709488B2 (en) Method and composition for hyperthermally treating cells
US9017729B2 (en) Method and composition for hyperthermally treating cells
Escoffre et al. Bubble-assisted ultrasound: application in immunotherapy and vaccination
US8795251B2 (en) Method and composition for hyperthermally treating cells
Sawant et al. Cancer research and therapy: Where are we today
CN103893761B (zh) 一种超声辅助基于光热剂的肿瘤细胞杀伤方法、***和应用
US9233157B2 (en) Method and composition for hyperthermally treating cells
Grumezescu Biomedical applications of nanoparticles
Carney et al. Harnessing nanomedicine for enhanced immunotherapy for breast cancer brain metastases

Legal Events

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

Ref document number: 18864099

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18864099

Country of ref document: EP

Kind code of ref document: A1