<<

US 20170172932A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2017/0172932 A1 Peyman (43) Pub. Date: Jun. 22, 2017

(54) EARLY DETECTION AND A 6LX 39/395 (2006.01) ENHANCED A61R 4I/00 (2006.01) (52) U.S. Cl. (71) Applicant: Gholam A. Peyman, Sun City, AZ CPC ...... A61K 9/50 (2013.01); A61K 39/39558 (US) (2013.01); A61K 4I/0052 (2013.01); A61 K 48/00 (2013.01); A61K 35/17 (2013.01); A61 K (72) Inventor: sham A. Peyman, Sun City, AZ 35/15 (2013.01); A61K 2035/124 (2013.01)

(21) Appl. No.: 15/143,981 (57) ABSTRACT (22) Filed: May 2, 2016 A method of therapy for a tumor or other pathology by administering a combination of thermotherapy and immu Related U.S. Application Data notherapy optionally combined with gene delivery. The combination therapy beneficially treats the tumor and pre (63) Continuation-in-part of application No. 14/976,321, vents tumor recurrence, either locally or at a different site, by filed on Dec. 21, 2015. boosting the patient’s immune response both at the time or original therapy and/or for later therapy. With respect to Publication Classification gene delivery, the inventive method may be used in cancer (51) Int. Cl. therapy, but is not limited to such use; it will be appreciated A 6LX 9/50 (2006.01) that the inventive method may be used for gene delivery in A6 IK 35/5 (2006.01) general. The controlled and precise application of thermal A6 IK 4.8/00 (2006.01) energy enhances gene transfer to any cell, whether the cell A 6LX 35/7 (2006.01) is a neoplastic cell, a pre-neoplastic cell, or a normal cell. US 2017/0172932 A1 Jun. 22, 2017

EARLY CANCER DETECTION AND 0007. One embodiment is a method for evaluating treat ENHANCED IMMUNOTHERAPY ment outcome in a patient having a genetic predisposition for a malignant neoplasm before clinical manifestation of 0001. This application is a continuation-in part of co the neoplasm can be seen radiographically. The method pending application Ser. No. 14/976,321 filed Dec. 21, 2015 permits visualization of any tumor, whether located exter which is expressly incorporated by reference herein in its nally on a patient’s body or located internally in the body, entirety. and as Small as 2 mm in diameter, producing a biomarker, 0002 Various factors may lead one to suspect the pres either a biomarker specific for the tumor or a general ence of a small cancerous or neoplastic tumor in a patient. biomarker. Such factors include the patient's genetic history, environ 0008. In general, a biomarker indicates a disease process. mental conditions to which the patient is or has been As Subsequently described, a biomarker can be a , exposed, the presence of biomarkers in the patient’s blood, , enzyme, hormone, carbohydrate, toxin, DNA, an or the presence of a lesion on a patient's skin or mucosal organism such as , tumor cell, exosome, or indirectly Surface. A small neoplasm, however, is often not recognized an , present in a liquid biopsy specimen. It can be unless and until it produces some clinical symptom. produced by the plasma cells, against a tumor antigen, etc. 0003. In a patient having a genetic mutation indicating a 0009. The method uses conjugated with nano predisposition to cancer, prophylactic Surgical intervention, particles which include but are not limited to quantum dots, Such as a bilateral mastectomy performed in a patient having with the conjugated form collectively termed functionalized a genetic mutation indicating a predisposition to breast nanoparticles, that are heated under specified conditions to cancer, is seldom performed. Additionally, a genetic predis produce a photoacoustic signal that is then visualized to position to one type of cancer may not lead to that type of locate the tumor to which the nanoparticles are attached. cancer, e.g. , but it may lead to another unsus Nanoparticles may be used for qualitative and quantitative pected type of cancer, e.g. malignant . Even if the assessment of an analyte in the blood or other tissue using other type of cancer is suspected, because of the finding of photoacoustic technology, U.S. Pat. No. 8,554.296. As pre biomarkers in the blood, a small internal lesion may not be viously stated, as used herein, unless specifically stated seen on radiography, or may not be accessible by Surgery, or otherwise, nanoparticles include but are not limited to quan the collateral complications may not be acceptable. It may tum dots. not suffice to just know the biomarker for a tumor, because 00.10 Early stage small neoplastic cells produce bio this information may not indicate whether the tumor is a markers that are either specific to the tumor cells or that primary site or a metastatic site, the tissue of its origin, represent the body's response to the tumor as an antibody. and/or its location. It is appreciated that some treatment The biomarkers can be proteomic, genetic, epigenetic or techniques such as Surgery or radiation may be useful, but glycomic biomolecules. These biomolecules can be recog only if the tumor is tissue specific. Radiation and chemo nized in the patient’s tissue samples or in the blood. Their therapy also have their own side effects, and may not destroy existence can be demonstrated thus far chemically using, the tumor completely. Larger tumors present a much com e.g., immunoassay or PCR methods. Quantitation of these plex problem, e.g., mutations in one area of the tumor are biomarkers is also important to determine disease progres usually different from mutations in another area of the same sion and prognosis of the disease. tumor. 0011 Biomarkers for many diseases are found in the 0004. It is clearly preferable, then, to manage small early blood. As Subsequently disclosed, biomarkers detected in a neoplasms that have not progressed to a larger tumor to liquid biopsy sample are used to generate antibodies against provide the patient an improved clinical prognosis. them using known methods in the art. The anti-tumor 0005. The invention includes a method of therapy for a antibodies are used to coat nanoparticles in the inventive non-Surgically accessible tumor by administering a combi method, where a lesion can be imaged regardless of the nation of thermotherapy and immunotherapy combined with lesion size or location in the body. The method is not limited gene delivery. The combination therapy beneficially treats to tumor detection and/or therapy. As only one example, the tumor and prevents tumor recurrence, either locally or at detecting an antibody against anti-beta-amyloid protein a different site, by boosting the patient’s immune response present in Alzheimer's disease in a liquid biopsy specimen, both at the time or original therapy and/or for later therapy the method renders the plaque visible with the nanoparticles as a “booster to the original therapy. With respect to gene and accessible to the inventive treatment. As another delivery, the inventive method may be used in cancer example, the method can also be used to detect and/or treat therapy, but is not limited to such use; it will be appreciated inflammatory processes, etc. that the inventive method may be used for gene delivery in 0012. The inventive method is applicable to any pro general. For example, the inventive method facilitates cel cesses or diseases that produce a biomarker detectable in a lular gene uptake by current methods that lack a thermal liquid biopsy specimen. It is applicable to a lesion including energy component, Such as electroporation, quantum dot an abscess, an ulcer, a tumor either benign or malignant, an delivery, etc. The controlled and precise application of ischemic area of a stroke and/or an area of the brain affected thermal energy enhances gene transfer to any cell, whether by a stroke whether visible or microscopically. the cell is a neoplastic cell, a pre-neoplastic cell, or a normal 0013 Well over a thousand are differentially cell. expressed in and thus may serve as biomark 0006. The inventive method provides in vitro and in vivo ers. Such proteins play a role in cancer-related processes precision immunotherapy to decrease or eradicate a malig Such as angiogenesis, , cell differentiation, cell nant neoplasm at an early stage of the disease. This method signaling, hematopoiesis, hormonal control, immune reac provides a vaccination effect to prevent at least the same tions, etc. Exemplary biomarkers include, but are not limited kind of cancer or recurrences. to, CEA for both malignant pleural effusion and peritoneal US 2017/0172932 A1 Jun. 22, 2017

cancer dissemination; HER-2/neu for stage IV breast cancer; endoscope and a cutting probe, has a fast post-operative bladder tumor antigen for urothelial cell carcinoma; thyro recovery. Such a method is in contrast to completely remov globulin for thyroid cancer metastasis; a-fetoprotein for ing the uterus with the tumor intact out of caution that the hepatocellular carcinoma; PSA for ; CA 125 tumor may harbor neoplastic cells, but using a large incision for non-small cell lung cancer; CA 19.9 for pancreatic with significantly higher operative risks and post-operative cancer; CA 15.3 for breast cancer; the combination of leptin, complication probabilities. Another, more problematic prolactin, , and IGF-II for , the example, is the decision for a woman having genetic dis combination of CD98, fascin, spIgR, and 14-3-3 eta for lung position to breast cancer without any physical or radiologi cancer; troponin I for myocardial infarction, and B-type cal manifestation. The woman must endure the stress and natriuretic peptide for congestive . While the fear not knowing if or when she may develop breast cancer, previous nine proteins are the only approved markers for and must consider prophylactic removal of both breasts. As cancer testing to date, they are but a small fraction of the another example, a personal decision whether or not to total number of available biomarkers, and their sensitivity undergo radiation therapy when a nevus is discovered under and specific vary. the retina, and biopsy results that often do not provide 0014. Other common biomarkers include the estrogen definitive information because of the diversity of the cells in receptor/progesterone receptor (ER/PR), HER-2/neu, and the entire area of the tumor. ESFR for breast cancer, and TIMP-1-associated with serum 0019. When the tumor site is unknown, locating a bio HER2-positive breast cancer; KRAS and UGT1A1 for col marker in the circulation may be akin to finding a needle in orectal cancer; HER-2/neu for gastric cancer, c-KIT, CD20 a hay stack. For any particular tumor or cancer, not all antigen, CD30, and FIP1L1-PDGRF alpha, and PDGFR for biomarkers are even known. Similarly, finding a micro DNA GIST: Philadelphia Chromosome (BCR/ABL)/PML/RAR in the circulation may not provide an answer when the tumor alpha and TPMT/UGT1A1/ALK EGFR for leukemia/lym is either invisible or has already metastasized. An example phoma; KRAS/EGFR for lung cancer, and BRAF and S100 of this occurs in patients with uveal , having a for melanoma. mortality rate of about 50%, even if the tumors undergoes 0015. Other examples of biomarkers include tumor sup radiation, at the time the ophthalmologist discovers the pressors that are lost in cancers, such as BRCA1, BRCA2; tumor. This points to the fact that a malignant tumor can RNA such as mRNA, microRNA; proteins found in body metastasize very early, at times even when the size of the fluids or tissue such as prostate specific antigen and CA-125; tumor is about 2 mm in diameter which is equal to about one gene and protein based biomarkers; and nonspecific bio million cells. In general, these lesions do not have any markers such as glycosaminoglycans in body fluids; alkaline symptoms. phosphatase and urinary hydroxyproline in skeletal involve 0020. The inventive method makes it possible to evaluate ment; hyaluronic acid excretion and urinary hydroxyproline a patient with genetic predisposition of a malignant neo in disease, and combinations thereof. plasm before its clinical manifestation can be seen radio 0016. In malignancies, the biomarkers may be released graphically. into the circulation either prior to or after the tumor has 0021. In one embodiment, the presence of one or more grown Sufficiently to become metastatic. Small tumors (less biomarkers is evaluated in any body fluid or organ. Exem than about 2 mm) seldom have any clinical manifestations, plary bodily fluids include, but are not limited to, urine, however even Such small tumors can release chemical blood, cerebrospinal fluid (CSF), eye cavity fluid, tear film, and/or biomarkers into the circulation. sputum, fluid obtained from the trachea, bronchi, abdominal 0017. The existence of biomarkers in the circulation has cavity, vagina, uterus etc. The biomarkers are analyzed in been known, but has not met the threshold for locating tumor vitro by methods known in the art, e.g., immunoassays cells that could not be imaged radiographically or by ultra including enzyme-linked immunoassay (ELISA), Western Sound as long as the tumors were asymptomatic. Available blots, fluorescence in situ hybridization (FISH), polymerase imaging methods such as X-ray, magnetic resonance imaging chain reaction (PCR), etc. The biomarkers are then conju (MRI), functional MRI, computed tomography (CT) scans, gated with functionalized antibody coated nanoparticles CT ultrasound, etc. may not permit visualization of lesions and/or quantum dots, as known in the art. smaller than about 3 mm in diameter. This has been the case 0022. In one embodiment one obtains a liquid biopsy for most malignant tumors, or when a malignant tumor is sample. Such a sample may be obtained from, e.g., blood, created from a benign precursor lesion Such as nevus, breast urine, cerebrospinal fluid (CFS), aqueous or vitreous or unspecific cyst or unspecific scar, prostate tumors along with abdominal cavity fluid, lymph node fluid, bladder fluid, milk benign prostate hypertrophy, or uterus cancer inside the duct fluid, sputum, gastric fluid, bile duct fluid, sinus fluid, uterus fibroma, melanoma inside a skin nevus or in a etc. The patient may or may not have any clinical symptom. seborrheic keratosis, etc. Moreover, it is often difficult to The patient may or may not have history of a family follow a cancerous tumor which has been irradiated but may disposition for tumors in and/or cancer of the breast, brain, still harbor malignant cells, and that can start growing with lung, prostate, ovary, pancreas, etc., or a genetic abnormality time and metastasize before it shows a local growth that is leading to progression in diseases such as, e.g., Alzheimer’s, detected by conventional imaging or other methods. Parkinson's, post traumatic brain syndrome, brain tumor, 0018. The diagnosis of a malignant tumor may be other neurological disease, age related macular degenera extremely difficult, even when a tumor is visible clinically or tion, an infectious disease, an immune response, etc. The radiologically, e.g. a uterus fibroma that may have some method evaluates the components of the sample for cell free malignant transformation. Moreover, a diagnosis also affects nucleic acid-based biomarkers including but not limited to the decision whether or not and also how to remove the micoRNA and microDNA; protein-based biomarkers, extra tumor. As one example, accessing the uterus through a small cellular vesicle (EV)-based biomarkers that are contained incision, and removing the tumor piece by piece using an within exosomes, extracellular vesicles, or microvesicles, US 2017/0172932 A1 Jun. 22, 2017

and circulating tumor cell (CTC)-based biomarkers. The 0028. In another embodiment, self-assembling bio/nano method uses methodologies Such as next generation hybrid material consisting of two constituents at the nano sequencing (NGS) or recombinant affinity reagents fabri meter or molecular level composed of inorganic and organic cated into nanostructures such as carbon nanotubes, compounds, having amphiphilic characteristics, i.e., hydro nanowires, quantum dots, or gold nanoshells, to enhance philic and lipophilic components or micelles, which may be their detection with the use of, e.g., Surface-enhanced radioactive (e.g., Cu) or radioactive (e.g., tin) are prepared Raman scattering (SERS), as known in the art. with biocompatible coatings and administered in the body 0023 For example, if a known tumor exists and there is for both therapy and imaging. a known biomarker for the tumor, one may have or prepare 0029. In one embodiment, the functionalized nanopar an antibody against the tumor to be used in both imaging and ticles travel in the body and attach to receptors of desired therapy. Large tumors with symptoms can be imaged, but cells, e.g., tumors, Alzheimer's plaque, drusen of the retina, before the inventive method, there was a problem when a etc. These nanoparticles are imaged by applying external biomarker was present in a liquid biopsy specimen but the thermal energy and/or by applying a reversible or alternating tumor was invisible, e.g., an early stage of a tumor, and there magnetic field. The thermal energy causes the nanoparticles was no symptomatic or radiographic evidence of the tumor. to expand, producing an ultrasound wave in the tissue. The 0024 Detecting a tumor biomarker, typically a protein or ultrasound wave can be detected by an ultrasonic receiver a glycoprotein, in a liquid biopsy specimen is facilitated by which is imaged in two to three dimensional formats as a the inventive method. Once detected, an antibody against tomogram. In another embodiment the plaques in Alzheim that tumor biomarker can be prepared. The antitumor bio er's disease, and the drusen in age related macular degen marker antibody is used to located the tumor. Antibody eration, are rendered visible using silica coated nanopar production is a well-known method in the art, and it will be ticles <2 nm in diameter administered with turmeric, appreciated that the antibody against either or both of the glycosaminoglycan, amyloid antibody, or percolan, etc. and tumor biomarker and the tumor cell may be recombinant, are quantified. In another embodiment, the nanoparticles are monoclonal, polyclonal, or an aptamer. The prepared anti conjugated with antibodies, , sterols, antibiotics, tumor cell antibodies are conjugated with nanoparticles and antifungals, antibacterials, antiproliferative agents, etc. that administered to a patient, where they target the tumor cells can be released from silica coated gold nanoparticles when and can be detected and/or treated. Detection is by photoa coated with thermosensitive polymers, e.g., chitosan coated coustic imaging technology. Treatment is at least by one of nanoparticles heated to 40° C.-42 C., to treat various thermal energy. The photoacoustic detection and thermal diseases including bacteria, fungi, parasites, plaque, drusen, treatment is described herein. etc. In another embodiment, the plaques and drusen can be 0025. In one embodiment, any specific tumor related quantified by imaging using , MRI, photoacoustic tech biomarker may be used. One example uses or nology imaging, etc. herceptin, a recombinant , against the 0030. In another embodiment, the functionalized anti oncogene HER-2, previously mentioned, which is a member biomarker-conjugated nanoparticle, ranges in size from 1 of the human epidermal receptor (HER/ nm to 900 nm. In another embodiment, the functionalized EGFR/ERBB) family. Other examples of known monoclo biomarker ranges in size from 1 nm to 8 nm, chosen to nal antibodies or biologics include, but are not limited to, enhance their elimination through the kidney for facilitated , , racotunomab, obinotuZumab, pertu clearance. Zumab, belaniatumomab, , . ofatu 0031. In one embodiment, the nanoparticles are rendered mumab, botezomib, , ipilumumab, pembroli magnetic by coating with a thin film of iron oxide prior to Zumab, and daratumumab. their conjugation with biomarkers antibodies. 0026. In one embodiment, in the absence of a specific 0032. In one embodiment, the nanoparticles are rendered biomarker, antibodies against biomarkers that are shared by more biocompatible by coating with a compound, including a number of the tumors may be used. Such biomarkers but not limited to the following: (poly)ethylene glycol, cell include glycosaminoglycan, which is specific for a group of penetrating peptide (CPP), activating CPP (ACPP), biotin, cancers such as bladder, gastrointestinal, , etc. streptavidin, etc., as known in the art, prior to their injection Antibodies against Such biomarkers are then conjugated in the body. with nanoparticles, termed functionalized nanoparticles. The 0033. Thermal energy in the form of electromagnetic term “functionalized indicates nanoparticles that have been radiation, ultrasound, or an alternating magnetic field is coated to render them soluble, biocompatible, and/or tar applied, under the control of a photoacoustic imaging sys geted by conjugating them with a biomolecule Such as an tem, to the organ Suspected of potentially harboring an as yet antibody. invisible neoplasm. The thermal energy applied increases 0027. In one embodiment the nanoparticle may be one or the temperature of the exposed nanoparticle, and creates a more of the following compounds or contain one or more of photoacoustic image or tomogram of the accumulated the following components: quantum dots, nanowires, nano heated nanoparticles. This image or tomogram represents a tubes, nanoshells, nanocages, perovskites, nanoparticles that Suspected neoplasm in that organ, and is compared to an are magnetic Such as iron or iron oxide, paramagnetic, or image taken without the thermal application radiographi nanoparticles that are non-magnetic Such as gold, gold cally. silica, gold-iron, silica coated gold nanospheres and nan 0034. In one embodiment, one administers functionalized orods, ferritic, quartz, graphene, carbon, Zinc oxide, piezo antibody-coated nanoparticles that, once attached to tumor electric, etc. Any of these nanoparticles, alone or in cells, become visible with a photoacoustic imaging unit that combination, may be conjugated or otherwise associated corroborates with an image obtained or not seen with other with the biomarkers antibodies, using methods known in technology such as ultrasound, MRI, PET, CT scan, etc. In the art. one embodiment, the images obtained with other instru US 2017/0172932 A1 Jun. 22, 2017 ments are either overlapped using a processor or are taken creating a photoacoustic response for imaging in the body by simultaneously during photacoustic imaging. In one light stimulation in the eye for simultaneous imaging and embodiment, after administration of the antibody-coated therapy. nanoparticle, an MRI image is overlapped with the photoa 0040. In one embodiment, using antibody coated quan coustic image and compared by a processor to verify the tum dots and light of a specific wavelength that is absorbed changes in the imaged area. by the quantum dot and emits light of a different wavelength, 0035. In one embodiment, the nanoparticles are incorpo one can render the moving tumor cells and extracellular rated in liposomes. In this embodiment, they may contain vesicle visible attached to the quantum dots in the retinal or medications that, upon attainment of a specific tumor tem choroidal vessels, or vessels and tumors of the skin, or perature, are released. The type of is not limited, tumors located beneath the skin and their feeding vessels, by and can include anti-bacterial, anti-viral, anti-fungal, anti light absorbed by the quantum dots circulating in the vessels, neoplastic, anti-inflammatory Such as acetyl cycline, anti as is done in fluorescence angiography with appropriate beta-amyloid protein, other antibodies, non-steroidal antiin filters and camera. flammatory drugs, immune stimulating agents, anti-VEGF 0041. In another embodiment, a gold quantum dot in a agents, anti-aggregation agents such as sterols, etc. mesoporous silica shell or cage is coated with an antibody or 0036. In another embodiment, antibody-coated nanopar a biomarker to any cell, e.g., neuronal or tumor cells, retinal ticles conjugated with thermosensitive polymers such as drusen, Alzheimer plaques, etc. for delivering medication or chitosan, carrying any medication including but not limited gene to an organ, e.g., retina or brain. to sterol, squalamine, lanosterol, is administered to a patient 0042. In another embodiment, the extent of plaque or having a neurologic pathology Such as Alzheimer's disease, drusen, as an indicator of disease progression in the brain or Parkinson's disease, or age related retinal drusen, etc. In this eye, respectively, can be evaluated by conjugating nanopar embodiment, administration is either intravenous or local in ticles with antibodies to glycosaminoglycan, heparin Sulfate, the cerebrospinal fluid or vitreous cavity, respectively, or at glycosaminoglycan, and/or heparin Sulfate proteoglycan, another local site. After controllably increasing the tempera and injecting the composition into the body or locally to ture of the functionalized nanoparticle to between 40° adhere to plaques or drusen for diagnosis, quantitation, C.-43° C. by increased energy delivery through a delivery and/or therapy with antibodies and medication. Source, under the control of the photoacoustic imaging 0043. In another embodiment the nanoparticles are used system and a processor, the temperature-sensitive coating for simultaneous imaging and thermotherapy of very Small polymers such as chitosan melts and release medications tumors. The nanoparticles are heated to a temperature rang specific to the pathology. For example, a medication to ing from 41° C.-43° C. releasing anti-cancer medication, dissolve would be administered to a patient along with inhibitory siRNA, or modify a gene using the with Alzheimer's disease; a medication to remove retinal CRISPR/cas9 system or another CRISPR system, addition drusen would be administered to a patient with age related ally releasing checkpoint inhibitors such as CTLA-4 or PD-1 retinal disease, etc. along with tumoricidal vectors, etc. 0037. In one embodiment, the functionalized nanopar 0044. In one embodiment, the nanoparticles are rendered ticle, e.g., a nanoshell, nanocage, etc., is combined with radioactive by coating with alpha or beta radiators that are biodendrimers that are conjugated with biomarkers and antibody specific or nonspecific biomarkers of the tumor. monoclonal antibodies and/or genes, e.g., siRNA, mRNA, The nanoparticles can also be coated with heat sensitive etc., for simultaneous visualization and therapy. polymers, including but not limited to chitosan, PEG, poly 0038. In another embodiment, after thermal imaging one amino esters, etc. increases the temperature of the functionalized nanopar 0045. In one embodiment, checkpoint inhibitors defined ticles. This is achieved by increased energy delivered by a as components that act as co-stimulatory or thermal delivery source under the control of the photoacous co-inhibitory molecules, poisons such as bee or Snake tic imaging system connected to a processor. The energy Venom, or other toxic agents that damage tumor cell mem delivery unit increases the temperature of the functionalized branes, or genes that inhibit tumor growth, siRNA, siLDNA, nanoparticles to 42° C.-43° C. to melt the temperature miRNA, mDNA along with the CRISPR/cas 9 complex or sensitive coating polymers such as chitosan and release variations of these may be used. anticancer medications, or inhibitory genes, siRNA, 0046. In one embodiment, the nanoparticles are coated miRNA, or checkpoint inhibitors, or introduce missing with a specific or a nonspecific biomarker Such as gly genes, or add any other genes for gene editing from the cosaminoglycan and injected into the circulation, into a thermosensitive coating of the nanoparticles along with a body fluid such as the lymphatic system or cerebrospinal CRISPR complex to modify the genetic composition of the fluid (CSF), or inside a body cavity. Examples of injection tumor cells, etc. In another embodiment, the temperature of sites include, but are not limited to, eye, sinuses, abdominal the functionalized nanoparticles is increased, by the thermal cavity, bladder, uterus, etc. The nanoparticles may also be delivery unit via a processor under the control of the injected into the breast ducts, e.g., through the nipple, inside photoacoustic imaging unit to image the temperature and the brain, into the prostate or other organ, or may even be control it to 45° C.-47 C., to 47°C., or to 50° C. to kill the applied topically. The injected nanoparticles circulate and Suspected tumor to which the antibody-coated nanoparticles seek cells bearing a receptor to their antibody, or perhaps are attached. cells with specific receptors or biomolecules, and readily 0039. In one embodiment, one synthetizes hybrid, very attach within minutes or hours. Small (1 nim-8 nm) gold silica nanoparticles that have a dual 0047. In one embodiment, specific or non-specific bio function, the nanoparticles antibody coated for imaging, and markers' antibodies are conjugated with nanoparticles and having photovoltatic and magnetic properties, to release one injected either into circulation or locally into a body cavity. or more gene(s) or medication(s) at certain temperatures, The nanoparticles travel and seek cells bearing specific US 2017/0172932 A1 Jun. 22, 2017

receptors or biomolecules, and attach within a few hours. less than 2 mm in diameter, which are radiographically The patient’s body or organ is then scanned, with the thermal invisible without knowing their exact location in the body. energy producing radiation or an alternating or reversible 0053. In one embodiment using photoacoustic technol magnetic field to heat the nanoparticles. Using photoacous ogy and a specific or non-specific tumor biomarker, a very tic technology, the Sound wave generated by the thermal Small lesion (<2 mm in diameter) is imaged in the body expansion of the nanoparticle induced by absorption of the when the tumor has not caused any clinical symptom. The thermal energy is recorded. The Sound wave signals may inventive method thus is used to differentiate a malignant originate from any part of the body, or from a specific organ. lesion from a benign lesion, even if the cancerous lesion is inside a begin lesion. It is noteworthy that biopsy of these 0048. In one embodiment, an alternating magnetic field very Small tumors, even when the lesion is visible, e.g., on produces heat in magnetic nanoparticles as a result of rapid skin or under the retina, may not yield malignant cells if the circular or semicircular motion of the nanoparticles. The biopsy is performed on a part of the lesion that contains patient’s body is scanned within the reversible magnetic benign cells. With tumors in the brain, it is most often the field, and the photoacoustic sound is recorded as a tempera case that the tumors will not be noted absent a neurological ture profile of the site of the nanoparticle/cell membrane symptom. imaged and location of the lesion is verified. 0054. In one embodiment, the inventive method is used 0049. In another embodiment, other source of thermal with specific biomarkers of a tumor such as breast cancer, energy are used. Such sources include, but are not limited to, prostate cancer, glioma, pancreatic malignancies, along with electromagnetic radiation, visible light, invisible light, infra nonspecific biomarkers. The location and size of a malignant red radiation, microwaves, or radiofrequency waves, etc. tumor in any organ is imaged in a patient with a genetic The nanoparticles are heated from body temperature of 37 propensity to develop a tumor. The thermal energy may also C. to 40° C. or 43°C., or if needed to 45° C. At the desired be applied, if desired, to treat the lesion simultaneously with temperature, e.g., 41° C.-43°C., the heat sensitive coating of providing the photoacoustic effect. Subsequent evaluation of the nanoparticle melts, releasing its cargo of e.g., medica the level of these biomarkers in the blood indicate if the tion, gene, etc., thus facilitating or enhancing passage of lesion was damaged or eliminated by the method, including these compounds through the membrane of the neoplastic use of medicaments and/or other treatment agents delivered cells. by the method as cargo in the nanoparticles. 0055. In one embodiment, a combination of biomarkers 0050. In another embodiment, use of a photoacoustic can be used in an early stage. For example, specific or technology unit controls the thermal delivery unit and the nonspecific bio-markers such as glycosaminoglycans can be thermal energy delivered to the nanoparticles to maintain or used in imaging a malignant lesion using antibody-coated reach a predetermined temperature for a desired time. nanoparticles to photoacoustically image the presence of a 0051. In one embodiment, the temperatures rise of the very Small early stage tumor anywhere in the body. nanoparticles expands them, producing a photoacoustic 0056. In another embodiment, the inventive method is Sound wave. This photoacoustic Sound wave is recorded by employed to determine residual tumor cells that may have one or multiple ultrasonic receivers located on the patients left at the site of a tumor resection or elsewhere in the body, skin. The signal can be obtained from any part of the body, and to treat or eliminate the residual tumor cells. or from a specific organ, since the signal travels through the 0057. In another embodiment, the functionalized nano body as a wave. The signal or Sound pulse is converted to an particles are conjugated with one of the recombinant, mono electric pulse in the receiver, then is amplified and imaged clonal, or or aptamers known in the art on a monitor. A processor produces a two- or three-dimen and administered along with either one or more toxin(s) or sion image of the lesion, localizing the location of the Sound antibodies, along with a medication that is provided at a and indicating the size of a lesion and its temperature by the much lower dose systemically to kill the already compro amplitude of the Sound pulse, mised tumor cells. Monoclonal antibodies that may be used 0.052 In one embodiment, photoacoustic imaging is used include, but are not limited to, those shown in Table 1, e.g., for a very early stage diagnosis of cancerous lesion that are rituximab, obinuZumab, oftumumab, etc. TABLE 1. Name Trade name Type Source Target Use

3F8 mab OSC GD2 8H9 mab OSC B7-E3 neuroblastoma, sarcoma, metastatic brain cancers mab OSC CA-125 (imitation) ovarian cancer ReoPro Fab chimeric CD41 ( platelet aggregation alpha-IIb) inhibitor mab humanized CD51 C8Ce mab human integrin C437 inflammatory bowel disease, , Crohn's disease mab human Clostridium difficile Clostridium difficile Humira mab human TNF-C. , Crohn's Disease, Plaque , , , US 2017/0172932 A1 Jun. 22, 2017

TABLE 1-continued

Name Trade name Type Source Target Use Juvenile Idiopathic Arthritis, Hemolytic disease of the newborn 8) human EpCAM prostate and breast C8CC 8) human beta-amyloid Alzheimer's disease F(ab') mouse TNF-C. Afutuzumab 8) humanized CD2O F(ab')2 humanized VEGFR2 C8CC ALD518 humanized IL-6 rheumatoid arthritis Campath, 8) humanized CD52 MabCampath 8) human NARP-1 hypercholesterolemia Altumomab pentetate Hybri-ceaker 8) OSC CEA (diagnosis) 8) chimeric mesothelin C8CC Anatumomab Fab OSC TAG-72 non-Small cell lung mafenatox carcinoma Anetumab ravtansine 8) 8 MSLN C8CC 8) 8 C.B systemic receptor erythematosus 8) humanized L-13 (=IMA-638 8) humanized HLA-DR2 hematological cancers Arcitumomab CEA-Scan Fab OSC CEA gastrointestinal cancers (diagnosis) AScrinvacumab 8) 8 -like C8CC kinase 1 8) humanized L-selectin (CD62L) severely injured patients 8) humanized CD274 Atinumab 8) 8 RTN4 Atlizumab Actemra, 8) humanized L-6 receptor rheumatoid arthritis (=) RoActemra 8) 8 Rhesus factor hemolytic disease of the newborn citation needed 8) humanized beta amyloid Alzheimer's disease Simulect 8) chimeric CD25 (C. chain of IL prevention of organ 2 receptor) transplant rejections 8) chimeric phosphatidylserine cancer, viral LymphoScan Fab OSC CD22 non-Hodgkin's lymphoma (detection) Begelomab 8) OSC DPP4 Benlysta, 8) human BAFF non-Hodgkin LymphoStat-B lymphoma etc. 8) humanized CD125 8) human CCL11 (eotaxin-1) severe allergic disorders Scintinun 8) OSC CEA-related antigen inflammatory lesions and metastases (detection) Bevacizumab Avastin 8) humanized VEGF-A metastatic cancer, retinopathy of prematurity 8) 8 Clostridium difficile Clostridium difficile infection Biciiromab FibriScint Fab OSC fibrin II, beta chain thromboembolism (diagnosis) Bimagirumab 8) 8 ACVR2B inhibitor 8) humanized IL17A and IL17F Bivatuzumab 8) humanized CD44 w8 squamous cell mertansine carcinoma BTE OSC CD19 C8CC 8) humanized SOST osteoporosis 8) humanized neural apoptosis dyslipidemia regulated proteinase 1 8) chimeric CD30 (TNFRSF8) hematologic cancers Briakimumab 8) 8 IL-12, IL-23 psoriasis, rheumatoid arthritis, inflammatory bowel diseases, multiple Sclerosis 8) 8 IL-17 inflammatory diseases 8) humanized VEGFA 8) Notch 1 C8CC US 2017/0172932 A1 Jun. 22, 2017

TABLE 1-continued

Name Trade name Source Target Use

Canakinumab Ilaris mab human IL-12 rheumatoid arthritis Cantuzumab mab humanized mucin CanAg colorectal cancer etc. mertansine Cantuzumab mab humanized MUC1 ravtansine mab humanized thrombotic thrombocytopenic purpura, thrombosis Capromab pendetide Prostascint mab OSC prostatic carcinoma prostate cancer cells (detection) mab human MCP-1 oncology immune indications Removab 3funct rat?mouse hybrid EpCAM, CD3 ovarian cancer, malignant ascites, gastric cancer cBR96-doxorubicin mab humanized Lewis-Y antigen C8CC immunoconjugate mab humanized CD4 prevention of organ transplant rejections, treatment of autoimmune diseases Cimzia humanized Crohn's disease Cetuximab Erbitux chimeric metastatic colorectal cancer and head and neck cancer Ch.14.18 chimeric 999 neuroblastoma humanize EpCAM ovarian cancer and other solid tumors 8 IGF-1 receptor solid tumors humanize Oryctolagus rheumatoid arthritis clinicatius chimeric CD4 rheumatoid arthritis Clivatuzumab hPAM4-Cide humanize MUC1 tetraxetan Codrituzumab humanize glypican 3 Coltuximab ravitansine chimeric CD19 8 TRAIL-R2 Concizumab humanize TFPI bleeding humanize 1-40-f-amyloid Alzheimer's disease CR6261 8 Influenza. A infectious hemagglutinin diseasefinfluenza. A humanize CD40 hematologic cancers Zenapax humanize CD25 (C. chain of IL prevention of organ 2 receptor) transplant rejections humanize -like growth cancer etc. actor I receptor Dapirolizumab pegol humanize CD40 ligand Daratumumab 8 CD38 (cyclic ADP ribose hydrolase) Dectrekumab 8 L-13 humanize DLL4 Denintuzumab humanize CD19 mafodotin Prolia 8 RANKL osteoporosis, bone metastases etc. Derlotuximab biotin chimeric histone complex recurrent glioblastoma multiforme OSC B-lymphoma cell lymphoma chimeric ganglioside GD2 neuroblastoma human hemagglutinin influenza. A Dorlimomab aritox OSC human DRS cancer etc. Duligotumab human HER3 human atopic diseases human CD274 C8CC human LGF2 C8CC chimeric GD3 ganglioside malignant melanoma Ecullizumab Soliris humanized C5 paroxysmall nocturnal hemoglobinuria OSC endotoxin sepsis caused by Gram-negative bacteria Panorex OSC EpCAM colorectal carcinoma Raptiva humanized LFA-1 (CD11a) psoriasis (blocks T-cell migration) US 2017/0172932 A1 Jun. 22, 2017

TABLE 1-continued

Name Trade name Type Source Target Use

Efungumab Mycograb ScFw 8 Hsp90 invasive Candida infection 8 Crohn's disease, induced protein ulcerative colitis Elgemtumab 8 ERBB3 C8CC humanize SLAMF7 OSC humanize humanize HHGFR C8CC humanize TWEAK receptor cancer etc. 8 AGS-22M6 cancer expressing Nectin-4 Enlimomab pegol OSC CAM-1 (CD54) humanize B7-EH3 Enokizumab humanize asthma 8 DLL4 chimeric SAC Epitumomab cituxetan OSC episialin humanize CD22 cancer, SLE humanize TGB2 (CD18) heart attack, stroke, traumatic shock Rexomun 3funct rat?mouse hybrid HER2/neu, CD3 breast cancer etc. Etairacizumab Abegrin 8) humanize integrin CVB3 melanoma, prostate cancer, ovarian cancer etc. humanize integrin C7 B7 inflammatory bowel disease 8 3 dyslipidemia 8 PCSK9 hypercholesterolemia 8 surface hepatitis B antigen Fanolesomab NeutroSpec OSC CD15 appendicitis (diagnosis) 8) OSC interferon receptor 8) humanized folate receptor 1 ovarian cancer 8) HNGF acute sciatic pain FBTAOS Lymphomun 3funct hybrid CD2O chronic lymphocytic leukaemia humanized respiratory syncytial respiratory syncytial virus infection Fezakimumab 8 IL-22 rheumatoid arthritis, psoriasis humanized HGF cancer etc. 8 IGF-1 receptor adrenocortical carcinoma, non-Small cell lung carcinoma etc. Firivumab 8 influenza A virus hemagglutinin 8 TYRP1 (glycoprotein melanoma 75) Fletikumab 8 IL. 20 rheumatoid arthritis HuzAF humanized Crohn's disease etc. 8 CD3 epsilon 8 rabies virus rabies (prophylaxis) glycoprotein 8 idiopathic pulmonary fibrosis, focal segmental glomerulosclerosis, C8CC mab human NGF pain mab chimeric EGFR mab chimeric B-cell lymphoma mab human IGF-I C8CC mab human beta amyloid Alzheimer's disease mab OSC CD147 () graft versus host disease Gemtuzumab Mylotarg mab humanized CD33 acute myelogenous OZogamicin leukemia mab humanized IL-1B etc. Rencarex mab chimeric carbonic anhydrase clear cell renal cell 9 (CA-1X) carcinoma81 Glembatumumab mab human GPNMB melanoma, breast vedotin C8CC US 2017/0172932 A1 Jun. 22, 2017

TABLE 1-continued

Name Trade name Source Target Use Simponi human rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis Gomiliximab chimeric CD23 (IgE receptor) allergic asthma human IL23 psoriasis balizumab humanized CD4 HIV infection britumomab tiuxetan Zevalin OSC non-Hodgkin's lymphoma crucumab human VEGFR-1 cancer etc. darucizumab humanized dabigatran reversal of effects of dabigatran govomab Indimacis-125 OSC CA-125 ovarian cancer (diagnosis) MAB362 human CLDN18.2 gastrointestinal adenocarcinomas and pancreatic tumor malumab human MIF C8CC mciromab Myoscint OSC cardiac myosin cardiac imaging mgatuzumab humanized EGFR C8CC human selectin P ndatuximab ravtansine chimeric SDC1 indusatumab vedotin human GUCY2C C8CC nfliximab Remicade chimeric TNF-C. rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis, Crohn's disease, ulcerative colitis 8 CDS1 solid tumors (prostate cancer, melanoma) nolimomab OSC CD25 (C. chain of IL graft versus host 2 receptor) disease notuzumab humanized CD22 OZogamicin pilimumab Yervoy 8 CD152 melanoma ratumumab 8 CD30 (TNFRSF8) Hodgkin's lymphoma Satuximab chimeric CD38 C8CC tolizumab humanized CD6 Xekizumab humanized IL-17A autoimmune diseases chimeric CD4 chronic asthma CEA-Cide humanized CEA colorectal cancer Lambrolizumab humanized PDCD1 antineoplastic agent humanized CFD humanized IL-13 asthma OSC NCA-90 diagnostic agent (granulocyte antigen) 8 CSF2 8 TGF beta 2 reduction of scarring after glaucoma Surgery 8 TRAIL-R2 C8CC 8 hepatitis B surface hepatitis B antigen humanized phosphate-sodium co-transporter humanized IGHE severe asthma and chronic spontaneous urticarial satetraxetan OSC humanized 8 Lodelcizumab humanized hypercholesterolemia veterinary Canis lupus familiaris IL31 Lorvotuzumab humanized CDS6 mertansine 8 CD40 multiple myeloma, non Hodgkin's lymphoma, Hodgkin's lymphoma Lulizumab pegol humanized CD28 autoimmune diseases chimeric CD23 (IgE receptor) chronic lymphocytic leukemia humanized ERBB3 8 TRAIL-R1 US 2017/0172932 A1 Jun. 22, 2017 10

TABLE 1-continued

Name Trade name Source Target Use humanized ch4D5 OSC T-cell receptor human GMCSF receptor C rheumatoid arthritis chain humanized EGFR colorectal, lung and stomach cancer Bosatria humanized IL-5 asthma and white blood cell diseases human TGF beta 1 systemic humanized CD74 multiple myeloma and other hematological malignancies OSC TAG-72 tumor detection (and therapy?) Mirvetuximab chimeric folate receptor alpha Soravitansine OSC GD3 ganglioside Small cell lung carcinoma humanized CCR4 human Rhesus factor Numax humanized respiratory syncytial respiratory syncytial virus virus (prevention) Moxetumomab OSC CD22 C8CC pasudotox Muromonab-CD3 Orthoclone OSC CD3 prevention of organ OKT3 transplant rejections OSC C242 antigen colorectal cancer human CSF2 Naptumomab OSC ST4 non-Small cell lung estafenatox carcinoma, human RON C8CC Tysabri humanized integrin C4 multiple Sclerosis, Crohn's disease human endotoxin sepsis human EGFR non-Small cell lung carcinoma Nemolizumab humanized IL31RA OSC TNF-C. human angiopoietin 2 C8CC Theracim, : humanized EGFR squamous cell Theraloc carcinoma, head and neck cancer, nasopharyngeal cancer, glioma Nivolumab human PD-1 C8CC Nofetumomab Verluma OSC cancer (diagnosis) merpentan Obilitoxaximab chimeric Bacilius anthracis Bacilius anthracis anthrax spores Gazyva humanized CD2O Chronic lymphatic leukemia humanized C8CC humanized rheumatoid arthritis, etc. OSC LFA-1 (CD11a) prevention of organ transplant rejections, immunological diseases Arzerra human chronic lymphocytic leukemia etc. human PDGF-RC C8CC humanized IL6 Xolair humanized IgE Fc region allergic asthma : humanized human scatter factor C8CC receptor kinase chimeric/humanized TEM1 C8CC human LINGO-1 multiple Sclerosis humanized EpCAM C8CC OwaRex OSC CA-125 ovarian cancer human oxLDL chimeric/humanized CD3 diabetes mellitus type 1 humanized CD37 C8CC human OX-40 asthma US 2017/0172932 A1 Jun. 22, 2017 11

TABLE 1-continued

Name Trade name Source Target Use humanized NOGO-A ALS and multiple sclerosis humanized TNF-C. chimeric lipoteichoic acid sepsis (Staphylococcus) PaliwiZumab Synagis, humanized F protein of respiratory syncytial Abbosynagis respiratory syncytial virus (prevention) virus Wectibix 8 EGFR colorectal cancer Pankomab humanized tumor specific ovarian cancer glycosylation of MUC1 8 Pseudomonas Pseudomonas aeruginosa aeruginosa infection 8 EGFL7 C8CC humanize IL-4 asthma Pasotuxizumab chimeric/humanized folate hydrolase humanize LTA 8 HER3 Pembroizumab humanize PDCD1 cancer etc. Theragyn OSC MUC1 Perakizumab humanize IL17A arthritis Omnitarg humanize HER2/neu. C8CC humanize reduction of side effects of cardiac Surgery humanize PD-1 cancer and infectious diseases humanize CD22 C8CC Pintumomab OSC adenocarcinoma adenocarcinoma antigen (imaging) Placulumab 8 human TNF humanize CD79B C8CC humanize human beta-amyloid Alzheimer's disease chimeric CD4 Crohn's disease, multiple Sclerosis Pritoxaximab chimeric E. coli Shiga toxin type-1 8 vimentin brain cancer PRO 140 humanized CCR5 HIV infection humanized IGHE asthma OSC N-glycolylneuraminic C8CC acid 8 fibronectin extra C8CC domain-B 8 rabies virus rabies (prophylaxis) glycoprotein Ralpancizumab humanized neural apoptosis dyslipidemia regulated proteinase 1 Cyramza 8 VEGFR2 solid tumors Lucentis humanized VEGF-A (wet form) 8 anthrax toxin, anthrax (prophylaxis protective antigen and treatment) Refanezumab humanized -associated recovery of motor glycoprotein function after stroke 8 cytomegalovirus cytomegalovirus glycoprotein B infection humanized IL-5 of the airways, skin and gastrointestinal tract 8 HGF solid tumors 8 platelet-derived neovascular age growth factor related macular receptor beta degeneration Rituximab MabThera, 8 chimeric CD2O , Rituxan leukemias, some autoimmune disorders human IGF-1 receptor C8CC Roledumab human RHD humanized sclerostin osteoporosis : humanized IFN-O. systemic lupus erythematosus LeukArrest 8 humanized CD11, CD18 haemorrhagic shock etc. US 2017/0172932 A1 Jun. 22, 2017 12

TABLE 1-continued

Name Trade name Source Target Use Antova humanized CD154 (CD4OL) rheumatic diseases humanized tumor-associated C8CC calcium signal transducer 2 humanized CD2OO C8CC 8 IL6 rheumatoid arthritis, ankylosing spondylitis Satumomab pendetide OSC TAG-72 cancer (diagnosis) 8 IL-17A , rheumatoid arthritis psoriasis 8 ERBB3 C8CC Setoxaximab chimeric E. coli Shiga toxin type-2 8 cytomegalovirus cytomegalovirus infection humanize FAP C8CC SGN-CD19A humanize CD19 acute lymphoblastic leukemia and B-cell non-Hodgkin lymphoma SGN-CD33A humanize CD33 Acute myeloid leukemia Sifallimumab humanize IFN-O. SLE, dermatomyositis, polymyositis Situximab chimeric IL-6 humanize LOXL2 fibrosis humanize CD2 psoriasis, graft-versus host disease (prevention) 8 IL-6 rheumatoid arthritis Softuzumab vedotin humanize CA 12S ovarian cancer humanize beta amyloid Alzheimer's disease : OSC EpCAM Sonepcizumab humanize sphingosine-1- choroidal and retinal phosphate neovascularization Somtuzumab humanize episialin 8 myostatin muscular dystrophy Sulesomab LeukoScan OSC NCA-90 osteomyelitis (imaging) (granulocyte antigen) humanized HIV-1 viral infections 8 BAFF B-cell cancers Tacatuzumab AFP-Cide humanized alpha-fetoprotein C8CC etraxetan adocizumab humanized integrin C.IIbf83 percutaneous coronary intervention humanized IgE allergic reaction humanized NGF pain OSC CD19 cancercitation needed 8 Notch receptor C8CC Aurexis humanized clumping factor A Staphylococcusatiretts infection Telimomab aritox OSC OSC tenascin C chimeric CD40 humanized CD3 diabetes mellitus type 1 8 CD221 hematologic tumors Tesidolumab 8 TGN1412 humanized CD28 chronic lymphocytic leukemia, rheumatoid arthritis Ticilimumab 8 CTLA-4 (=) humanized IL23 immunologically mediated inflammatory disorders humanized TRAIL-R2 C8CC TNX-6SO humanized IL-13 Hodgkin's lymphoma Tocilizumab6 (= Actemra, humanized IL-6 receptor rheumatoid arthritis atlizumab) RoActemra humanized CD154 (CD4OL) rheumatoid arthritis, etc. Tosatoxumab 8 Staphylococcus (iiietS US 2017/0172932 A1 Jun. 22, 2017 13

TABLE 1-continued Name Trade name Type Source Target Use

Tositumomab Bexxar OSC CD2O 8) 8 8) 8 IL-13 asthma etc. Trastuzumab Herceptin 8) humanized HER2/neu. breast cancer TRBSO7 Ektomab 3funct 2 GD2 melanoma 8) humanized CD4 Tremelimumab 8) 8 8) 8 growth muscle atrophy due to differentiation factor 8 orthopedic disuse and sarcopenia Tucotuzumab 8) humanized EpCAM C8CC celmoleukin 8 hepatitis B virus chronic hepatitis B 8) chimeric 8) 8 C—X—C hematologic receptor type 4 malignancies 8) 8 4-1BB cancer etc. 8) humanize Escherichia coi diarrhoea caused by E. coli Stelara 8) 8 IL-12, IL-23 multiple Sclerosis, psoriasis, psoriatic arthritis Vandortuzumab 8) humanize STEAP1 C8CC vedotin 8) 8 Frizzled receptor C8CC 8) humanize angiopoietin 2 C8CC 8) chimeric AOC3 (VAP-1) 8) 8 CD27 8) humanize ITGA2 8) humanize integrin C437 Crohn's disease, ulcerative colitis 8) humanize CD2O non-Hodgkin's lymphoma 8) OSC AOC3 (VAP-1) inflammation Vesencumab 8) 8 NRP1 Nuvion 8) humanize CD3 Crohn's disease, ulcerative colitis 8) chimeric integrin C.531 solid tumors Vorsetuzumab 8) humanize mafodotin Votumumab HumaSPECT mab 8 tumor antigen colorectal tumors CTA A16.88 HuMax-EGFr mab 8 EGFR squamous cell carcinoma of the head and neck HuMax-CD4 8) 8 CD4 rheumatoid arthritis, psoriasis, T-cell lymphoma Zatuximab 8) chimeric HER1 C8CC Ziralimumab 8) 8 CD147 (basigin) Zolimomab aritox 8) OSC CDS systemic lupus erythematosus, graft versus-host disease

0058. In another embodiment using photoacoustic tech ticles expand and create a photoacoustic response that is nology, the circulating tumor, exosomes, or extracellular recorded by the photoacoustic receiver. vesicles in the blood are quantified non-invasively by having 0060. The probe can be moved in any direction, e.g., up a thermal energy source Such as laser microwave, RF, or and down, side to side, etc., over the skin while recording the other unit mounted on the patient’s wrist, neck, etc. and a Sound waves from the nanoparticles. Using a processor in receiver to count and record the Sound wave generated by the photoacoustic unit, one uses the photoacoustic response circulating cells to which the antibody-coated nanoparticles data to construct a two- or three-dimensional image of the are attached. tumor. The hand held probe permits scanning any bodily 0059 In another embodiment, the ultrasonic receiver of surface, including but not limited to breast, eye, CNS, spinal the photacoustic unit is an array of ultrasonic receivers cord, extremities, internal organs, eye, nose, chest, trachea, mounted on a handheld probe. The hand held probe contacts throat, abdomen, and urogenital organs. The data from the the patient’s skin via a gel placed over the area suspected to ultrasonic array probe of the photoacoustic unit is stored in contain a tumor or lesion. It simultaneously records multiple a computer during the probe's motion, permitting video photoacoustic signals from the lesion during thermal energy construction showing tumor shape, structure, location, etc. application. Thermal energy applied pulses can range from for video presentation, evaluation, and archiving. one per second to a million times or more per second. Each 0061. In one embodiment, the unit is capable of storing time a thermal pulse reaches the nanoparticles, the nanopar vast quantities of data from photoacoustic signals. The unit US 2017/0172932 A1 Jun. 22, 2017

is also capable of storing vast quantities of data from to the Smaller exosome size. All exosomes or other types of non-stationary tissues, e.g., circulating tumor cells and exo extracellular vesicles are less than one micron; in contrast, Somes in blood vessels, that have accumulated antibody tumor cells are five to twenty times larger than exosomes. coated nanoparticles on their cell membranes. The targeted The inventive system for the first time permits in vivo cells can also be any normal or abnormal circulating cell in observation and separation of exosomes from tumor cells, the blood or lymphatic system. The photoacoustic unit and separation of circulating tumor cells from a tumor mass. reproduces signals from these mobile cells and/or exosomes The separated cells or cell structures can be observed, as photoacoustic cinematography/angiography or video. counted, and quantified to assess the therapeutic effect of a 0062. In one embodiment, the cinematography or video procedure on tumor cells. recording is done by the photoacoustic unit recording at least 0066. In another embodiment, after imaging and therapy, 30 frames/second of photoacoustic signals, and converting the biomarkers are collected from liquid biopsies and com them into an image of a moving object. A cinematography pared with those obtained prior to therapy in different or video is performed by obtaining at least 30 frames of post-operative periods to confirm the therapeutic effect of photos of a moving object per second. In photoacoustic the procedure and prognosticate the condition. Videography or photoacoustic angiography, 30 or more 0067. In another embodiment, the antibody coated nano frames of pulse signals from the heated nanoparticles per particles are conjugated and administered with checkpoint second are needed to reproduce or convert the still images to inhibitors along with known immune therapy agents and a moving object, e.g., blood flow, etc. by the unit. Use of vaccines to facilitate circulating killer cells attack and Such a system is known: Peyman et al. Ophthalmic Surg removal of tumor cells. Laser Imaging 43 (2012) 143-51 doi: 10.3928.15428877 0068. In another embodiment, polymeric nanoparticles or 20120105-01 showing, however, lower resolution because polysaccharide or synthetic polymers conjugated with bio no nanoparticles or photoacoustic imaging system was markers are administered to enhance a vaccination effect and employed, and expressly incorporated by reference herein in are taken up by antigen presenting cells. its entirety. 0069. In one embodiment, genetic analysis of the patient 0063. In one embodiment the photoacoustic processor is performed to determine a sequence of the gene that is converts the microscopic still images to a video or photoa mutated. A sample of the patient’s blood is analyzed for any coustic angiography; since the only moving parts in the of the following indicia of the presence or a neoplasm or a vessels that are targeted with antibody coated nanoparticles predisposition to a neoplasm: Specific tumor biomarker(s), are the circulating tumor cells or exosomes, extracellular non-specific tumor biomarker(s), extravascular vesicles, cir vesicles or bubbles covered with antibody coated nanopar culating tumor cells, tumor microRNA, micro DNA, or any ticles that are heated by a pulse of thermal energy produces other tumor indicator. RNA sequencing reflects the dynamic an internal ultrasonic pulse signal recorded by the photoa of gene expression for detection of RNA fragments, coustic receiver. A moving image of the cells and exosomes including mRNA, noncoding RNA, chimeric RNA, patho can be created by the unit whether the cells are on the tumor gen RNA, extracellular RNA, etc. interior or exterior. 0070. Examples of biomarkers have been previously dis 0.064 Nanoparticle assisted photoacoustic video-angiog closed. Other biomarkers include DNA hypermethylation, raphy or nanoparticle assisted photoacoustic cinematogra the presence of ZNF154 in colon, lung, breast, stomach, and phy is novel and inventive. All “photoacoustic' terminology endometrial tumor, and the stem cell marker NANOG, a has previously been used for describing tissue heating or the mitochondrial oxidative phosphorylation/fatty acid oxida difference in the temperature between two tissues, vessels tion molecule in highly malignant tumor-initiating stem-like vs. skin, and has been done with light alone, not in combi cells (TICs) that reprograms mitochondrial metabolism. nation with nanoparticles. In one embodiment, the method is 0071 Use of results from a patient’s genetic analysis performed for therapy by providing the patient with at least advantageously permits selection of a therapeutic agent, one antibody-coated functionalized nanoparticle having a along with antibody-coated nanoparticles conjugated with detectable property, with the antibody targeting the func thermosensitive polymers and thermotherapy, to provide the tionalized nanoparticle to a specific patient site, then heating greatest efficacy against cancers that are Smaller than 4 mm the nanoparticles to generate a photoacoustic signal, i.e., in diameter. In general. Such cancers have not grown to a thermal therapy, and imaging to visualize any localized size whereby they show genetic differentiation of the cancer nanoparticle at the site. The ultrasonic receiver of the cells. Treatment of these Small cancer cells can thus include photoacoustic unit is an array of ultrasonic receivers treatment of the (s). In one embodiment, mounted on a hand held probe simultaneously recording nanoparticles activated by electromagnetic radiation, either multiple photoacoustic signals from the lesion during ther in vitro or in Vivo, enhance both gene transfer and cell mal energy application which in one embodiment is pulsat proliferation of any desired cell, including stem cells ing. The array of ultrasonic receivers of the photoacoustic 0072. In one embodiment, the patient’s blood is pro unit mounted on a hand held probe simultaneously records cessed to isolate the patients own natural killer (NK) cells, multiple photoacoustic signals from the lesion or vessels i.e., a type of lymphocyte that is part of the patients innate during thermal energy application, reproducing motion of immune system, and dendritic cells, i.e., immune cells that moving nanoparticles and/or cells as a nanoparticle assisted process antigen material and present it on the cell Surface to photoacoustic video-angiography or nanoparticle assisted T cells of the immune system). NK cells and dendritic cells photoacoustic cinematography. are isolated from a patient’s blood using commercially 0065. In another embodiment, software associated with available kits known in the art, e.g., EasySepTM and Roset the photoacoustic unit can enhance either or both the pho teSepTM STEMCELL Technologies Inc., Tukwila Wash.; toacoustic signals and resulting images. Enhancement may NK Cell Isolation Kit, Meltinyi Biotech, Bergisch Gladbach facilitate differentiating exosomes from circulating cells due Germany. The natural killer cells/dendritic cells are rendered US 2017/0172932 A1 Jun. 22, 2017

sensitized to the tumor in vitro. Sensitization is accom anti-tumor antibody coated nanoparticle. This beneficially plished by co-culturing the patient’s natural killer cells increases the hyperthermal damage to cancer and other and/or dendritic cells with IL-2 and the antibody-coated undesirable cells due to toxin release from the nanoparticles nanoparticles containing the optional penetration-enhancing coating of thermosensitive polymer at 40° C.-43° C. agents and/or thermosensitive polymers as previously 0078. In one embodiment, genes are provided that have a described. The patient’s sensitized natural killer cells/den stimulatory action in response to light or ultrasound. An dritic cells are then administered to the patient at intervals to example of Such a gene is the opsin gene and members of the provide a booster immune therapy, much as a vaccine opsin family. In this embodiment, Such genes are provided booster injection does. IL-2 is a protein produced by the T to regulate cell membrane polarization and depolarization. cells. When conjugated with the thermosensitive antibody Such genes can thus controllably create an action potential coated nanoparticle, upon controllable temperature release, in the membrane of an excitable cell. Such as a retinal cell, IL-2 is systemically available to enhance a T-cell response in or a non-excitable cell such as a tumor cell. Controllable the patient by cell sensitization and proliferation. regulation may drive a permanent depolarization state to 0073. Thermal damage to the tumor cell membrane as a render the cells accessible to a desired medicament for cell part of nanoparticle assisted thermotherapy releases destruction. that, in vivo, activate and stimulate a dendritic cell immu 0079. In one embodiment, combinations of genes can be nogenic response. The activated dendritic cells induce a used for controllable regulation. As an example, genes signal that additionally activates -driven tumor cell responding to light to produce action potential, combined damage or killing. with genes that can modifying a defective gene(s) in the cells 0.074. In one embodiment, the medium used to culture of an organ, e.g., eye, brain, lung, spinal cord, peripheral NK/dendritic cells contains viral like particles (VLP). The nerve, lung, digestive tract, can be used in combination to NK/dendritic cells pick up the VLP and enhance sensitiza facilitate regulation of actions including Swallowing, breath tion against the tumor. If tumor cell biopsy specimens are ing, gland secretion, etc., to restore the normal function of available, NK cells/dendritic cells are cultured from these the organ. As another example, genes responding to light to biopsy specimens which additionally contain tumor lysate, produce an action potential, combined with inhibitory genes killed circulating tumor (CT) cells, and their extracellular such as siRNA, RNAi, microRNA, can be used to inhibit vesicles (ECV). In one embodiment, nanoparticles with tumor function by simultaneous depolarization of the tumor thermosensitive polymers and conjugated with tumor anti cells. These genes can additionally be combined with che body and VLP are administered to the patient intravenously, motherapeutic agent to work synergistically and damage the as the first step of tumor vaccination and therapy. The tumor cells. nanoparticles become attached to the tumor cells within a 0080 Systemic administration of tumor antibody coated few minutes after administration. nanoparticles, coated with thermosensitive polymers and a 0075. In one embodiment, the tumor biomarkers from a cell penetration facilitating agent, targets the nanoparticles patient’s blood are identified, and anti-tumor antibodies are toward the tumor cell membrane. External energy is applied prepared, using conventional antibody techniques known in by a thermal delivery device that uses energy (electromag the art. The antibodies may be monoclonal, polyclonal, netic radiation, microwave radiation, radiofrequency waves, humanized, etc.; tumor antibodies also includes aptamers an alternating magnet, focused ultrasound, etc.) to increase (oligonucleotide or peptides that bind to a specific target). the temperature of the nanoparticle. The heated nanoparticle The antibodies/aptamers are then coated on diagnostic or absorbs more energy than the tissue Surrounding the nano therapeutic nanoparticles or quantum dots, which are then particle. The temperature increase causes the nanoparticles systemically administered to the patient. In vivo, the tumor to expand. Expansion of the nanoparticles creates a photoa antibody-coated nanoparticles seeks the tumor cells via the coustic, thermoacoustic, or ultrasound wave, whose Sound specificity of the anti-tumor antibody component. In one wave amplitude correlates with the amount of the tempera embodiment, adding a cell penetration enhancing agent to ture increase, i.e., the degree of the temperature rise. the polymer or other coating facilitates penetration of the 0081. In one embodiment, the ultrasound wave is tumor-antibody-coated nanoparticles into a tumor cell. Cell recorded by a transducer and is transmitted to a unit to image penetration enhancing agents render the nanoparticle com the nanoparticle increase in temperature as one-, two- or plex more biocompatible, and have been previously dis three-dimensional images. This unit is connected to the closed; they include cell penetrating peptide (CPP), thermal delivery device via a computer to maintain the activated CPP (APCC), (poly)ethylene glycol (PEG), biotin amount of thermal energy needed for the time required to streptavidin, etc. heat the nanoparticles to the desired temperature and for the 0076. In one embodiment, as previously disclosed, the desired time period and thus release medicament(s), gene(s), tumor-antibody-coated nanoparticles are also coated with a VLP, etc. These agents may also be against microorganisms, thermosensitive polymer that dissolves at a particular tem e.g., bacterial, viral, fungal, or parasitic agents, that have perature, e.g., a polymer Such as chitosan that dissolves at a developed resistance to the therapeutic agents. For example, temperature of 40°C.-43°C., and/or an arginine rich poly heated bacteria become more permeable to diffusion of mer, etc. appropriate medication; in contrast, non-heated bacteria 0077. This coating, in addition to its thermosensitive remain resistant. properties, and include one or more medicaments, genes, I0082 In one embodiment, nanoparticles coated with the etc. thus providing additional therapy to the patient upon desired antibody (e.g., anti-tumor antibody, anti-bacterial administration and thermotherapy. In one embodiment, add protein antibody, etc.) are administered to the patient to ing a phospholipase, anti-phospholipid antibody, toxin assure that the antibody-nanoparticle complex is in contact (Snake, Scorpion, bee venom, etc. to the polymer or other with the appropriate cells or tissues. It will be appreciated coating enhances the damage to the cell membrane from an that the appropriate cells or tissues may include both circu US 2017/0172932 A1 Jun. 22, 2017

lating cells (e.g., ECV, endosomes, leukemic cells, etc.) and heart attack, intestinal bleeding, kidney disease, dis non-circulating cells (e.g., Solid tumor). ease, etc. As another example, release of phospholipase 0083. In one embodiment, a small hand held photoacous enzymes can create a hole in the membrane of tumors or tic unit with a small thermal delivery unit e.g. laser, micro other cells to provide or facilitate entry of a medicament(s) wave, or radiofrequency unit is placed externally over a and/or gene(s) entry into a cell. Subcutaneously located vessel to deliver a pulse of energy I0088. In the inventive precision nanoparticle assisted and to heat the nanoparticles attached to the circulating thermotherapy imaging (NATTI), the temperature of the tumor cells and create a photoacoustic Sound as they heat up. tumor cell to which the nanoparticle is conjugated is con This records the Sound wave each time a tumor cell passes trollably precisely increased. The temperature increases (a) by the external hand held unit, adjusts the temperature from releases a medicament(s) and/or gene(s) from a thermosen 37°C.-43°C., thus assessing and quantifying non-invasively sitive coating on the nanoparticle, and (b) enhances pen the circulating tumor cells using the hand held thermal etration of the medicament(s) and/or gene(s) through the imaging device. open pores of the tumor cell membrane. NATTI technology 0084. In one embodiment, photoacoustic technology includes a computer-controlled thermal energy delivery unit imaging is controlled to a low temperature of 37°C.-43°C., to ensure attainment of a desired increased temperature of thus assessing and non-invasively measuring circulating the tumor for achieving the therapeutic goal. Controlling cells using a hand held thermal imaging device. Imaging thermal energy delivery to achieve a temperature from 38° may be used in combination with any standard method, C. to 42° C. for drug delivery or more in the tumor including but not limited to radiography, computed tomog nanoparticle complex to a tumor, or to another tissue raphy (CT), magnetic resonance imaging (MRI), ultrasound, affected by a disease as directed by antibody binding to a positron and other molecular imaging devices. corresponding antigen. It will be appreciated that the 0085. In one embodiment, nanoparticles are conjugated increased temperature may be maintained at the controlled with VLP derived from plant . In this embodiment, desired level for any desired time interval, e.g., up to 1, 2, the VLP are used for cancer therapy by carrying sRNA, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 minutes, or even RNAi, etc. The host of these viruses are plants e.g. tobacco longer, depending upon the need. mosaic virus (TMV), bean yellow dwarf virus (BeYDV), I0089. Typically, normal healthy cell membranes are com etc., which cannot infect the patient. Thermal application of prised of the phospholipids phosphatidylserine (PS) and the antibody coated nanoparticles provides the control over phosphatidylethanolamine (PE), which are located within when and where these particles are released to provide the inner membrane and oriented toward the cell interior. maximum benefit in immunotherapy. The VLP are generally However, for cancer cells, the orientations of PS and PE are immunogenic and do not require adjuncts to induce an flipped, so each oriented toward the cell exterior. The immune response. These modified viruses are devoid of melting point of PS and PE is about 65° C.-70° C. degree. genetic components and cannot replicate in the body. How When the nanoparticles are heated to this temperature, the ever, if a specific gene of a specific protein e.g. an antibody, exposed PS and PE lipids of the cell membrane melt, and is conjugated with them and injected in the plant, the create a dehiscence in the cell membrane through which modified viruses produce large amounts of the antibody or chemotherapeutic agents can freely flow into the tumor cell, protein in the plant, which can Subsequently be extracted and killing the tumor cell. used in human infective or non-infective diseases or to 0090. For a medicament that is a chemotherapeutic agent, produce a vaccine to treat e.g. Alzheimer's disease etc. Once local release permits a higher concentration of a chemo the antibody is produced, it can be used in diagnosing or therapeutic agent to be contained in the polymer coating, guiding treatyment to the affected area in combination with i.e., a Supratherapeutic concentration, because it is confined nanoparticles and drug delivery. to a localized site and would not result in Systemic toxicity, I0086. With respect to a gene(s) present in the polymer yet still would achieve a higher therapeutic level at the tumor coating, e.g., an inhibitory gene Such as siRNA, siLNA, site. In one embodiment, a known existing antitumor che RNAi, or an appropriate checkpoint inhibitor, may be used. motherapeutic agent is administered at a concentration that Checkpoint inhibitors enhance cellular immune responses to exceeds that of a concentration that would be administered tumor specific proteins in the cancer cells, as previously under typical therapy, yet that does not result in patient disclosed. In one embodiment, a checkpoint inhibitor Such toxicity. Similarly, one can use a toxic medicament(s) to as nivolumab is combined with nanoparticle-assisted tar locally perturb a cellular metabolic pathway or a specific cell geted immunotherapy as an adaptive T-cell transfer mecha cycle, e.g., local tumor cell perturbation. Such agents are nism. These, along with the a CRISPR/cas9 or CRISPR generally not administered intravenously or orally because interference (CRISPRi) complex, may perturb or modify the of their serious or fatal systemic toxicity and side effects. tumor genes. The concentration of the chemotherapeutic medicament(s) 0087. With respect to a medicament(s) present in the delivered by the inventive method is, in general, high polymer coating, the medicament(s) would be released locally, but less the /1000 of the concentration that would be locally. For a medicament that is a biologic, local release required if the medicament(s) were to be effective if deliv permits agents to be concentrated at the desired site without ered systemically. resulting in Systemic toxicity the medicament may otherwise 0091 Use of precision nanoparticle assisted thermo cause. As one example, anti VEGF agents, TNF inhibitors, therapy and imaging (NATTI) may be used to fine-tune the antineoplastic medications such as taxol, , approach to perturb Survival mechanisms of tumors or other antiinflammatory agents, steroids, antibiotics antiviral agent, pathological cells. It will be thus appreciated that the inven etc. can localize at a tumor or other site at significantly tive method may be used as a rapid en mass treatment of a higher concentrations to stop tumor neovascular growth, cancer or an organ. For example, it may be used as a without causing the known systemic complications such as preliminary treatment in advance of other therapies that in US 2017/0172932 A1 Jun. 22, 2017

general have severe debilitating systemic complications cellular immune response due to the additional tumor anti Such as immune Suppression, etc. and which may take longer gens present, and drawing the patient's immune cells, den to obtain approval for their administration. Thus, nanopar dritic cell, T-lymphocytes, B-lymphocytes, , ticle assisted thermotherapy and drug delivery NATTI etc., to the tumor location. This mechanism also advanta avoids the complication of damaging the geously provides immune memory functioning as an internal patient’s immune system as a result of one or multiple vaccination method. Specifically, local release of antigens chemotherapeutic agents used in these late metastatic cancer from damaged tumor cells enhances a patient’s immune patients. response to a large amount of other tumor cell associated 0092. The nanoparticle assisted thermoacoustic imaging antigens, creating a form of in Vivo Vaccination. Such technology, along with thermal energy, drives the medica vaccination can be provided as needed, e.g., annually, at ment(s), gene(s), VLP into the , with simultane specific intervals, upon specific events, etc. to prime the ous generation of an immune response to the cancer cell and patient’s immune cells against any future tumor cells, and inhibition of cancer cell proliferation by including siRNA, protects against reappearance of any tumors with similar siLDNA, etc., along with simultaneously enhancing gene antigenic components. For example, vaccination may be therapy by conjugating a CRISPR/cas9 or CRISPRi com administered annually orbiannually or between annual and plex to the nanoparticle to correct or inhibit a genetic biannual administration indefinitely, unless new biomarkers component of a tumor. Once inside a cell, e.g., a tumor cell are discovered in the patient, necessitating additional and/or or the cell of an organ, the gene(s), along with the CRISPR/ earlier therapy. cas9 complex or CRISPRi, enter the or mito 0096. In addition, the inflammatory process created as a chondria and precisely modify the gene pool of those cells. result of the immune therapy and cellular response increases The RNA-guided bacterial endonuclease Cas9 is the effector the temperature of the tissue involved, which is also protein of the type IICRISPR/Cas9 system that detects and recorded using photoacoustic technology imaging to image subsequently generates a double-strand break (DSB) in the tumor location and its potential metastatic lesions any target DNA. This may treat a disease caused by a gene where in the body. deficiency, or add a new useful gene(s), or remove and 0097. This embodiment results in precise, local, inter possible replace a gene, in the cell nucleus or the mitochon nally-induced immunotherapy and simultaneous vaccina dria. tion. The antigen, e.g. VLP is delivered intravenously with 0093. The gene(s) and/or medicaments(s) may be deliv thermosensitive polymers conjugated with antitumor anti ered to a specific site, but not released in the circulation from body coated nanoparticles. The VLP are released from these the nanoparticles until reaching the required elevated tem nanoparticles only when the temperature of the nanoparticle perature and after attaching to the tumor or other desired is increased, and the nanoparticles are localized only at the cells or desired location. It will be appreciated that the tumor site, due to the specificity of the anti-tumor antibody inventive method can be used in therapy for non-neoplastic with which the nanoparticles are conjugated. diseases. As one example, the amyloid plaques present in 0098. As previously disclosed, various nanoparticle Alzheimer's disease may be used to produce anti-amyloid types, compositions, configurations, etc. are possible, plaque antibodies and treated by the inventive method. As including the following non-limiting examples: organic, another example, bacteria in patients with severe sepsis synthetic, metallic, non-metallic, magnetic, non-magnetic, refractory to standard antibiotic therapy, e.g., patients with paramagnetic, etc., configurations such as a nanosphere, methicillin-resistant Staphylococcus aureus (MRSA) etc., nanotube, nanoshell, nanocage, nanocarbon, etc., including may be used to produce anti-bacterial antibodies and treated quantum dots, dendrimers, liposomes, piezoelectric nano by the inventive method. In this example, the method may particles, etc. be combined with extracorporeal treatment of blood, using 0099. In one embodiment, piezoelectric nanoparticles are a thermal energy delivery unit to provide electromagnetic stimulated by an ultrasonic unit, providing a therapeutic radiation, radiofrequency waves, microwaves, focused ultra effect by inducing an electric current in cells. Depending Sound, an alternating magnetic field, etc., under the control upon the frequency, this exposure can kill cells on one hand, of the described NATTI unit, to controllably achieve a or it can enhance growth of specific cells on the other hand. temperature of 42° C.-45° C. to kill the bacteria prior to Application of thermal energy at a frequency in the range of cooling the blood to the normal 37°C. prior to reinfusion to 1 HZ-20 HZ promotes cell growth. Application of thermal the patient. energy at a frequency greater than about 60 Hz, damages 0094. In addition, increasing the temperature of the nano cells. Cell death is desirable for treating pathologies such as particles incrementally from 37°C.-43°C. allows precision cancer. However, cell proliferation is desirable to facilitating nanoparticle assisted thermotherapy and imaging (NATTI) tissue regeneration. For example, in this embodiment, a to release a gene(s) or medicament(s) from the nanopar patient with a stroke, or a myocardial infarction, or a spinal ticles. It will be appreciated that the method beneficially cord injury, may be treated to regenerate brain, heart, nerve permits imaging a tumor or other desired cells, such as tissue respectively. In this embodiment, the antibody used is Alzheimer's plaques, that are present in a small lesion targeted to the damaged cells, i.e., neurons, cardiac cells, otherwise invisible by conventional imaging methods such etc., and treatment is with a pulsed frequency of 1 HZ-20 Hz as radiography. or more is provided for 1 min-10 min. It will be appreciated 0095. The immune response is generated by two different that this embodiment permits stem cells to be controllably mechanisms. One mechanism is by releasing the VLP, which either stimulated or inhibited. then stimulate a cellular immune response at the site of the 0100. In one embodiment, the nanoparticle stimulates tumor. The other mechanism is by the thermotherapy-dam proliferation of in vitro cultured cells when the nanoparticle aged tumor cells releasing their antigenic material in and is exposed to and absorbs light pulses of low frequency, i.e., beyond the Surrounding tissues, creating a more active frequencies in the range of 1 HZ-20 Hz. Conversely, in one US 2017/0172932 A1 Jun. 22, 2017

embodiment, the nanoparticle inhibits cell proliferation be thought of as cellular debris or detritus, to prevent an when the nanoparticle is exposed to and absorbs light pulses immunogenic storm in the body. of very high frequency, i.e., frequencies in the range of >30 0106. In one embodiment, the patient receiving the HZ-100 Hz). Thus, selecting the frequency of the thermo inventive therapy undergoes plasmapheresis to remove, e.g., therapy, and thus the frequency to which the tumor antibody Such , enzymes, dead cells, etc. from the circula coated nanoparticles are exposed, adds to the mechanisms of tion. Plasmaphoresis is a known method to remove compo therapy the patient receives if the light pulses are at low nents from . Because the patient’s plasma is frequencies, i.e., no higher than about 20 HZ. treated extracorporeally, then reinfused, in contrast to rein 0101. In one embodiment, after sensitization of the fusing only cellular components of the patient’s blood, immune cells with the tumor antigen, functionalized quan plasmaphoresis also beneficially detoxifies the patients tum dots with antibody coated against cell membrane of plasma without compromising blood Volume and with mini immune cells is added so that the cell membranes of the mal or no fluid loss. This technique avoids the serious immune cells carry a marker that can be made visible with complications and side effects of simply returning the cel specific wavelength of light extracorporally. lular components of the blood to the patient. Additionally, all 0102. In one embodiment, antibody coated nanoparticles precautions are observed to avoid hypotension and loss of are conjugated with thermosensitive polymers containing calcium ions in the process of citrate anticoagulation that VLP/medication/genes, and intravenously administered to a this procedure requires. The patient can be treated initially patient. The VLP are released from the thermosensitive with presently available such as heparin, nanoparticles by thermal application at temperatures of 41 coumadin, etc., which can be immediately neutralized post C.-43° C. The increase in temperature is achieved using, procedure. Neutralization uses standard techniques known e.g., activation by light, electromagnetic radiation, micro in the art, such as calcium, etc. Hemofiltration treatment is wave radiation, radiofrequency waves, focused ultrasound, performed with activated carbon, treatment on non-ionic or alternating magnetic field to preferentially heat the nano exchange resins, etc. for removing free toxin and also toxin particles because of their high Surface to Volume ratio, and bound with plasma proteins, etc. as in renal dialysis meth because the selected molecular composition of the nanopar ods. The process may be instituted or repeated as needed, ticles preferentially absorbs more thermal energy than the e.g., if the tumor reappears. surrounding normal cells. The tumor cells to which the 0107. In one embodiment, to prevent a severe autoim nanoparticles are attached are also heated. mune response after tumor immunotherapy, one uses extra 0103. In one embodiment, the increase in the temperature corporeal plasmapheresis. A strong pulse of light energy is of the nanoparticles results in their thermal expansion. applied to a tube containing blood cells to achieve a tem Thermal expansion of the nanoparticles produces an ultra perature up to 60° C. to kill immune cells containing Sonic wave that passes through the body, is captured by a quantum dots. The blood is then passed through a dielec receiver, the ultrasonic pulse is converted and amplified by trophoresis system to characterize and remove dead or live an ultrasonic, photoacoustic, or thermoacoustic unit, imaged T-cells, sensitized killer cells, and tumor cells, prior to as a thermoacoustic signal or as nanoparticle assisted ther re-infusing the same blood or performing a blood transfu moacoustic signal, and converted by a computer to images, sion in the patient while simultaneously administering in one-, two-, or three-dimensions, of the temperature and immunosuppressive agents, including a biologic, to reduce the lesion. the severe autoimmune response often seen after tumor 0104. In one embodiment, the photoacoustic or nanopar immunotherapy. ticle assisted thermoacoustic unit controls the thermal 0108. The size of the nanoparticle may vary, and may energy delivery unit via a processor to maintain the tem vary depending on the site of therapy and imaging as well as perature of the nanoparticles at a predetermined temperature other factors. In one embodiment, the nanoparticle size as a closed circuit once the nanoparticles have attached to ranges from 1 nm to 999 nm or more. In one embodiment, the tumor cells. An increase in the temperature to which the the nanoparticle size ranges from 1 nm to 20 nm, which is nanoparticles are exposed, i.e., at the nanoparticle level. ideal for use in the eye and central to permit from 37° C. to 41° C.-43° C. melts the thermosensitive the nanoparticle access to the intercellular space, and also polymers coating the nanoparticles, releasing under control ideal for renal clearance without generating systemic side the conjugated VLP. medication/gene which are attached to effects. Nanoparticles having a size less than 10 nm in the thermosensitive antibody coated nanoparticles locally at diameter, and not bound to a tumor, i.e., nanoparticles that the desired site. This method is particularly effective in small are free in the circulation, undergo rapid renal elimination tumors, i.e., tumors less than 4 mm in diameter, because the from the body within a few hours of administration. Only tumor stem cells are still present at the original tumor site nanoparticles attached to the tumor cells remain in the body. and can be simultaneously killed and eliminated before This results in a novel form of simultaneous local thermo metastasis has occurred. therapy and vaccination. 0105. In one embodiment, a plurality of the antibody 0109 The localized thermotherapy component of the coated nanoparticles are injected into a patient’s circulation method damages the tumor cells, thus disseminating tumor with the cultured and tumor-sensitized NK cells/dendritic cell-associated antigens into the circulation, generating a cells to target the tumor. It will be appreciated that such cellular response to the various tumor biomarkers that were thermal damage to tumor cells, and a NK cellular response, originally present. This dual thermotherapy and cellular generates and releases relatively large quantities of lytic response augments the effect of both immunotherapy and enzymes and other cellular contents. In the case of Smaller thermotherapy. The inventive method augments immuno tumors, the released Substances are of Smaller quantities, but therapy methods that relied on T-cells that had been sensi for larger tumors it become necessary to remove these tized to just a few tumor markers, or that relied only on Substances released into a patient’s circulation, which may checkpoint inhibitors to prevent the tumor cells’ sequestra US 2017/0172932 A1 Jun. 22, 2017

tion from T-lymphocytes. Previous methods of tumor vac 0117 The patient’s blood is assessed for new biomarkers cination used intradermal or Subcutaneous antigen adminis from the dead cells. tration, with the antigen taken up by antigen presenting cells, 0118. The cultured T-cells and dendritic cells are har e.g., dendritic cells, to generate specific killer cells only at a vested, along with the nanoparticle-coated antigen plus VLP location remote from the specific tumor site. The inventive or RNA or DNA phages. These are stored under appropriate method augments previous thermotherapy methods by com conditions, and reinjected into the patient with low dose bining immunotherapy to act synergistically with thermo coated nanoparticles or systemic medicaments to be admin therapy, providing additional therapeutic mechanisms to istered as needed, e.g., semi-annually, annually, biannually, attack the tumor. etc. with repetition as needed. This is followed up with 0110. In one embodiment, cultured killer cells sensitized administering free circulating DNA and exosomes of circu to a tumor are administered simultaneously with the anti lating cells. tumor antibody coated nanoparticle-conjugated VLP to attack the tumor cells and remove the dead tumor cells. For EXAMPLE 2 example, an intradermally administered antitumor antibody 0119) A checkpoint inhibitor is added to a thermosensi coated nanoparticle with VLP can be administered in sub tive polymer coating a nanoparticle for controlled release of sequent rounds of therapy during a postoperative period to the checkpoint inhibitor using the inventive NATTI to treat induce an immune response as needed. This embodiment a patient with breast, colorectal, pancreatic, and/or ovarian decreases the likelihood of or prevents potential recurrences cancer. A checkpoint inhibitor Such as nivolumab is com of the tumor. bined with nanoparticle assisted targeted immunotherapy for 0111. In one embodiment for use in larger tumors of a adaptive T-cell transfer to overcome the limitations of stan sufficient size for biopsy, an antibody directed to the tumor dard immunotherapy. lysate (TL) is used as a Source of tumor-associated antigens (TAAS), and is conjugated with the nanoparticles for gen EXAMPLE 3 erating therapeutic anti-tumor immune responses. One can I0120 Nanoparticles are conjugated with a chimeric generate in vivo immunity against multiple TAA simultane receptor, a CD19 protein that is found only on B cells, along ously from the killed or damaged tumor cells during the with the T-cells cultured in vitro that expresses a chimeric thermotherapy. This embodiment broadens the repertoire of antigen receptor (chimeric antigen receptor T (CAR TAA-specific T-cell clones available for activation to a given T)-cells) to target abnormal B cells seen in leukemia. The tumor. reappearance of new biomarkers as neoantigens in these 0112. In one embodiment, after an initial thermotherapy patients can be also treated in the postoperative period using procedure, a blood sample is obtained from the patient. This the inventive method. blood sample contains released tumor antigens that are I0121 Plasmaphoresis is simultaneously performed or recoverable prior to treatment by the inventive method using performed after treatment. various immunoassays or methods of searching for biomark 0.122 This example treats acute and chronic hematologic ers. The tumor antigens are then used to generate, in vitro, malignancies such as acute lymphoblastic leukemia, non additional T-cells that are sensitized to many TAA for future Hodgkin lymphoma, chronic lymphocytic leukemia, chronic use in, along with VLP for vaccination of the same patient. myelogenous leukemia, etc. 0113. In one embodiment, immunostimulatory oligo nucleotide-loaded cationic graphene oxide, carbon nano EXAMPLE 4 tube, gold/iron, iron/zinc oxide, or cadmium Sulfate nano I0123 Nanoparticles are conjugated with all-trans retinoic particles are combined with photothermally enhanced acid (ATRA) and arsenic trioxide to target leukaemia cells in immunogenicity to achieve combined thermo-immune acute promyelocytic leukemia and used in the inventive therapy. In one embodiment, RNA oligonucleotides/gra method. The all-trans retinoic acid is released at the site of phene or graphene oxide, or long double stranded RNA/ the tumor without exposing the entire body to the toxic graphene oxide induces a controlled immunostimulation in medication, simultaneously, plasmophoresis is performed to combination with oncogene silencing RNAi. clear all toxin released in the blood, along with leukemic 0114 Nanoparticles, dendrimers, carbon nanotubes, cells. It is appreciated that other blood cell cancers are lipid-based carriers, micelles, gold nanoshells/nanocages, removed in the same session. PLGA, chitosan, PEI cationic lipid, and cationic polymers are useful gene therapy, gene delivery, and immunotherapy. EXAMPLE 5 These have the advantages of being easily prepared, biode 0.124. In a patient with a hematologic malignancy that is gradable, non-immunogenic, non-toxic, and water soluble. resistant to chemotherapeutic agents or immune therapy, NATTI is performed with gene delivery, along with chemo EXAMPLE 1. therapeutic agents, to target all immune cells initially with 0115 T cells and dendritic cells are obtained from a out Subjecting the patient to systemic heavy chemotherapy, patient’s blood, and grown in culture along with a tumor or followed by transplantation, without exposing other antigen, plus nanoparticles coated with thermosensi the entire body to systemic chemotherapy. tive polymers conjugated with antigen and VLP using cul ture methods known in the art. EXAMPLE 6 0116. The nanoparticle complex is injecting them along 0.125 Nanoparticles are conjugated with RNA that con with checkpoint inhibitors and IL-2. The inventive method tains an aptamer, ribosomes, and siRNA in a thermosensitive is applied, killing tumor cells, and increasing the response of polymer and administered to using NATTI to target specific T-cells and dendritic cell. tumor cells. US 2017/0172932 A1 Jun. 22, 2017 20

EXAMPLE 7 thermotherapy comprises systemically administering a tumor-antibody-coated nanoparticle, optionally also 0126 The microenvironment of the cancer cell is modi containing an agent that facilitates cell penetration and fied by delivering medicaments that block the uptake of optionally coated with a thermosensitive polymer; and exosomal signals and prevent the uptake of ECV. Such immunotherapy comprises systemically administering the medications include choloroquine, heparin, cytochalasin D, patient’s natural killer (NK) cells/dendritic cells pre and ethyl-isopropyl amiloride. These medications are sensitized in vitro to the tumor. approved for patient use. The medicaments are provided 2. The method of claim 1 where immunotherapy is using NATTI in conjunction with chemotherapeutic agents. administered at intervals to the patient after the initial therapy acting as a booster to the original immunotherapy EXAMPLE 8 and reduce or prevent tumor recurrences at a same or 0127. The inventive method provides nanoparticle different site. assisted localized immunothermotherapy and thermotherapy 3. The method of claim 1 where immunotherapy further for delivery of customized vaccines to target core mutations comprises administering NK cells/dendritic cells containing in a patient. The immune cells or T-cells that can attack those viral like particles (VLP). core mutations are identified via a cancer biomarker. The 4. The method of claim 1 where immunotherapy further immune cells or T-cells are then cultured with the nanopar comprises obtaining NK cells/dendritic cells grown in cul ticles coated with thermosensitive particles and VLP and ture under light pulses with a tumor biomarker from blood IL-2. The nanoparticles are injected into the patient, con or a tumor biopsy specimen containing tumor lysate, killed trollably heated using a thermal energy source, and imaged, circulating tumor cells (ct cell), and tumor extracellular for specific patient therapy. vesicles (ECV). 5. The method of claim 1 where thermotherapy includes EXAMPLE 9 exposing the tumor-antibody-coated nanoparticle to a light 0128 Nanoparticles are conjugated with RNA phage pulse at a frequency in the range of 20 HZ-60 Hz to decrease VLP, which is generally stable up to about 50° C. VLPs of proliferation of the tumor cell. the related RNA phage PP7 are crosslinked with inter 6. The method of claim 3 where thermotherapy includes Subunit disulfide bonds, rendering them significantly more using a thermoacoustic unit to control a thermal energy stable. They exhibit high immunogenicity. Such nanopar delivery unit using a processor to maintain the nanoparticles ticles complement the inventive NATTI technology and can at a predetermined temperature as a closed circuit once the be employed in anti-cancer and antibacterial treatment. Lytic nanoparticles have attached to the tumor cells, then control phages attach to receptors on the bacterial Surface, inject lably increasing the temperature to which the nanoparticles their genetic material through the bacterial membrane, and are exposed from 37° C. to 41° C.-43° C. for a predeter overtake the bacterium's transcription and translation mined desired time period resulting in melting a thermo machinery to synthesize new phages. sensitive polymer coating the nanoparticles, releasing under control a conjugated VLP. medication/gene which are EXAMPLE 10 attached to the thermosensitive antibody coated nanopar ticles locally at the desired site. 0129. To prevent a severe autoimmune response after 7. The method of claim 3 where immunotherapy com tumor immunotherapy, one uses after or before extracorpo prises stimulating the patients cellular immune response at real plasmapheresis, the nanoparticle assisted thermotherapy the tumor site by and imaging system to apply heavy thermal energy to a tube (a) the released VLP providing tumor-localized immuno containing blood cells and to achieve a temperature as high therapy, and as 60° C. to kill the sensitized immune cells containing (b) the released antigenic material from the thermo nanoparticles. Blood is then passed through a dielectropho therapy-damaged tumor cells providing localized and resis system to characterize and remove dead or live T-cells, non-localized immunotherapy functioning as an inter sensitized killer cells, and tumor cells prior to re-infusing nal vaccination method. blood in the patient while simultaneously administering 8. The method of claim 7 where (b) additionally provides immunosuppressive agents, including biologics. This immune memory. reduces the severe autoimmune response often seen after 9. The method of claim 7 resulting in a temperature tumor immunotherapy. increase at any localized or non-localized site, the tempera 0130. The embodiments shown and described in the ture increase permitting tumor imaging at the localized or specification are only specific embodiments of the inventor non-localized site by photoacoustic technology. who is skilled in the art and are not limiting in any way. 10. The method of claim 1 where the anti-tumor antibody Therefore, various changes, modifications, or alterations to is specific for at least one tumor biomarker in the patients those embodiments may be made without departing from the blood. spirit of the invention in the scope of the following claims. 11. The method of claim 1 where the antibodies from a The references cited are expressly incorporated by reference patient’s blood are to a tumor biomarker in the patients herein in their entirety. blood. What is claimed is: 12. The method of claim 6 where the polymer contains an 1. A cancer therapeutic method comprising administering inhibitory gene(s) and a CRISPR/cas9 complex to stimulate to a patient having an early stage tumor a combination of or modify the tumor genes upon release from the polymer at thermotherapy and immunotherapy combined with gene a desired temperature that is obtained by incremental therapy, the patient having a non-Surgically accessible early increase from 37° C. to 43° C. permitting precision nano stage tumor, where particle assisted thermotherapy and imaging (NATTI) to US 2017/0172932 A1 Jun. 22, 2017

release the gene(s) and optional medicament(s) and/or immunotherapy comprises systemically administering the checkpoint inhibitor(s) from the nanoparticles. patient’s natural killer (NK) cells/dendritic cells pre 13. The method of claim 1 further comprising conjugating sensitized in vitro to the cellular protein(s); and the nanoparticles with a chimeric receptor on B cells and gene delivery, if used, comprises use of a CRISPR/cas9 with T-cells cultured in vitro and expressing an antigen for and/or CRISPRi to replace or modify at least one the chimeric receptor to target abnormal B cells seen in genetic component in the cell. leukemia. 18. The method of claim 17 where thermotherapy 14. The method of claim 1 where the antibody is against includes in vitro exposing the antibody-coated nanoparticle an amyloid plaque. to a light pulse at a frequency in the range of 1 HZ-20 Hz to 15. The method of claim 1 further comprising performing increase proliferation of cell. precision nanoparticle assisted thermotherapy and generat 19. The method of claim 17 wherein the pathological ing a photoacoustic image of the cells to which the nano cellular protein(s) is an amyloid plaque and the patient has particles bind at an early cellular stage having a size less Alzheimer's disease, and/or is a protein from a microorgan than four millimeters not easily detectable by radiographic ism and the patient is Sub-responsive to therapy for that imaging. microorganism. 16. A method of simultaneous localized thermotherapy 20. The method of claim 17, further comprising perform and vaccination, the method comprising ing plasmaphoresis on the patient post-therapy to purify a administering a plurality of nanoparticles to a patient in patient’s blood from toxins and cellular components gener need thereof, the administered nanoparticles having a ated by the therapy. size less than 10 nm in diameter and coated with an 21. The method of claim 17, further comprising antitumor antibody, the patient provided nanoparticle applying a strong pulse of light energy to a tube contain assisted thermotherapy, ing the patient’s blood cells post-therapy to achieve a the unbound nanoparticles undergoing renal elimination temperature up to 60° C. to kill immune cells contain from the body within a few hours of administration, and ing quantum dots: the tumor-bound nanoparticles remaining in the patient. 17. A therapeutic method to treat a pathology implicating passing the pulsed blood cells through a dielectrophoresis a cellular protein(s), the method comprising administering to system to characterize and remove dead or live T-cells, a patient having a cellular pathology a combination of sensitized killer cells, and tumor cells; and thermotherapy and immunotherapy, optionally with gene re-infusing the dielectrophoresis treated blood in the delivery, where patient while simultaneously administering immuno thermotherapy comprises systemically administering an a Suppressive agents, thus reducing the likelihood of a antibody-coated nanoparticle, optionally also contain severe post-therapy autoimmune response in the ing an agent that facilitates cell penetration and option patient. ally coated with a thermosensitive polymer;