US20190351249A1 - Method and Apparatus for Electromagnetic Treatment of Living Systems - Google Patents

Method and Apparatus for Electromagnetic Treatment of Living Systems Download PDF

Info

Publication number
US20190351249A1
US20190351249A1 US16/431,561 US201916431561A US2019351249A1 US 20190351249 A1 US20190351249 A1 US 20190351249A1 US 201916431561 A US201916431561 A US 201916431561A US 2019351249 A1 US2019351249 A1 US 2019351249A1
Authority
US
United States
Prior art keywords
treatment
electromagnetic field
pemf
inter
pulsed electromagnetic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/431,561
Inventor
Arthur A Pilla
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Endonovo Therapeutics Inc
Original Assignee
Endonovo Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Endonovo Therapeutics Inc filed Critical Endonovo Therapeutics Inc
Priority to US16/431,561 priority Critical patent/US20190351249A1/en
Priority to US16/657,827 priority patent/US20200094068A1/en
Publication of US20190351249A1 publication Critical patent/US20190351249A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N2/00Magnetotherapy
    • A61N2/004Magnetotherapy specially adapted for a specific therapy
    • A61N2/008Magnetotherapy specially adapted for a specific therapy for pain treatment or analgesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N2/00Magnetotherapy
    • A61N2/02Magnetotherapy using magnetic fields produced by coils, including single turn loops or electromagnets

Definitions

  • the disclosure relates generally to the field of therapeutic devices and methods for treating patients.
  • Some embodiments pertain generally to a method and apparatus for therapeutic and prophylactic treatment of animal and human cells and tissues.
  • some embodiments pertain to use of non-thermal time-varying electromagnetic fields configured to accelerate the asymmetrical kinetics of the binding of intracellular ions to their respective binding proteins which regulate the biochemical signaling pathways living systems employ to reduce the inflammatory response to injury, and to enhance healing and well-being.
  • inventions pertain to the non-thermal application of repetitive pulse bursts of sinusoidal, rectangular, chaotic or arbitrary waveform electromagnetic fields to instantaneously accelerate ion-buffer binding in signaling pathways in animal and human cells and tissues using ultra lightweight portable coupling devices such as inductors and electrodes, driven by miniature signal generator circuitry that can be incorporated into an anatomical positioning device such as a dressing, bandage, compression bandage, compression dressing; lumbar or cervical back, shoulder, head, neck and other body portion wraps and supports; garments, hats, caps, helmets, mattress pads, seat cushions, beds, stretchers, and other body supports in cars, motorcycles, buses, trains, airplanes, boats, ships and the like.
  • anatomical positioning device such as a dressing, bandage, compression bandage, compression dressing; lumbar or cervical back, shoulder, head, neck and other body portion wraps and supports; garments, hats, caps, helmets, mattress pads, seat cushions, beds, stretchers, and other body supports in cars, motorcycles, buses, trains
  • the proposed EMF transduction pathway relevant to tissue maintenance, repair and regeneration begins with voltage-dependent Ca2+ binding to CaM, which is favored when cytosolic Ca2+ homeostasis is disrupted by chemical and/or physical insults at the cellular level.
  • Ca/CaM binding produces activated CaM that binds to, and activates, cNOS, which catalyzes the synthesis of the signaling molecule NO from L-arginine.
  • This pathway is shown in its simplest schematic form in FIG. 1A .
  • FIG. 1A is a schematic summary of the body's primary anti-inflammatory cascade and the proposed manner by which PEMF may accelerate postoperative pain relief. Surgical injury increases cytosolic Ca2+, which activates CaM.
  • PEMF accelerates CaM activation thereby enhancing NO/cGMP anti-inflammatory signaling. PEMF also enhances CaM-dependent PDE activation, which accelerates cGMP inhibition. PEMF dosing must take into account the competing dynamics of NO/cGMP signaling and PDE inhibition of cGMP.
  • cNOS* represents activated constitutive nitric oxide synthase (cNOS), which catalyzes the production of NO from L-arginine, which, in turn, activates soluble gyanylyl cyclase, sGC.
  • sGC* refers to activated guanylyl cyclase which catalyzes cyclic guanosine monophosphate (cGMP) formation when NO signaling modulates the tissue repair pathway.
  • AC* refers to activated adenylyl cyclase, which catalyzes cyclic adenosine monophosphate (cAMP) when NO signaling modulates differentiation and survival.
  • an EMF signal can be configured to accelerate cytosolic ion binding to a cytosolic buffer, such as voltage dependent Ca2+ binding to CaM, because the rate constant for binding, kon is much greater than the rate constant for unbinding, koff, imparting rectifier-like properties to ion-buffer binding, such as Ca2+ binding to CaM.
  • a cytosolic buffer such as voltage dependent Ca2+ binding to CaM
  • Yet another embodiment pertains to application of sinusoidal, rectangular, chaotic or arbitrary waveform electromagnetic signals, having frequency components below about 100 GHz, configured to accelerate the binding of intracellular Ca2+ to a buffer, such as CaM, to enhance biochemical signaling pathways in animal and human cells and tissues.
  • Signals configured according to additional embodiments produce a net increase in a bound ion, such as Ca2+, at CaM binding sites because the asymmetrical kinetics of Ca/CaM binding allows such signals to accumulate voltage induced at the ion binding site, thereby accelerating voltage-dependent ion binding.
  • Examples of therapeutic and prophylactic applications are modulation of biochemical signaling in anti-inflammatory pathways, modulation of biochemical signaling in cytokine release pathways, modulation of biochemical signaling in growth factor release pathways; edema and lymph reduction, anti-inflammatory, post-surgical and post-operative pain and edema relief, nerve, bone and organ pain relief, increased local blood flow, microvascular blood perfusion, treatment of tissue and organ ischemia, brain tissue ischemia from stroke or traumatic brain injury, treatment of neurological injury and neurodegenerative diseases such as Alzheimer's and Parkinson's; angiogenesis, neovascularization; enhanced immune response; enhanced effectiveness of pharmacological agents; nerve regeneration; prevention of apoptosis; modulation of heat shock proteins for prophylaxis and response to injury or pathology.
  • the systems, devices and/or methods generally relate to application of electromagnetic fields (EMF), and in particular, pulsed electromagnetic fields (PEMF), including a subset of PEMF in a radio frequency domain (e.g., pulsed radio frequency or PRF), for the treatment of any of the applications disclosed herein in animals and humans, including pain, edema, tissue repair and head, cerebral and neural injury, and neurodegenerative conditions.
  • EMF electromagnetic fields
  • PEMF pulsed electromagnetic fields
  • PRF pulsed radio frequency
  • sGC soluble guanylyl cyclase
  • cGMP cyclic guanosine monophosphate
  • the CaM/NO/cGMP signaling pathway can rapidly modulate blood flow in response to normal physiologic demands, as well as to inflammation. Importantly, this same pathway will also rapidly attenuate expression of cytokines such as interleukin-1beta (IL-1 ⁇ ), and iNOS and stimulate anti-apoptotic pathways in neurons.
  • IL-1 ⁇ interleukin-1beta
  • iNOS stimulate anti-apoptotic pathways in neurons.
  • PEMF can also accelerate the inhibition of cGMP by phosphodiesterase (PDE).
  • PDE phosphodiesterase
  • an apparatus incorporates miniaturized circuitry and light weight coil applicators or electrodes to deliver any of the waveforms described herein thus allowing the apparatus to be low cost, portable and, if desired, disposable.
  • the present invention relates to methods and apparatuses for treating patients with pulsed electromagnetic therapies (PEMF).
  • PEMF waveform and the period between PEMF waveform pulses can be configured to simultaneously increase the rate of cGMP production and to minimize the rate of inhibition of cGMP by compounds such as PDE.
  • a non-invasive, relatively low-energy or very low-energy PEMF treatment depends on the ratio of the duration of the treatment interval and the duration of the inter-treatment interval (also referred to herein as the inter-treatment period).
  • the methods described herein are invented from the new finding that the for externally-applied low-energy or (in some variations) very low-energy PEMF treatments, a ratio of treatment interval to inter-treatment period of greater than about 1:6 (e.g., an inter-treatment period that is greater than six times the treatment interval) results in efficacious treatment, whereas ratios less than 1:6 do not.
  • ratios of treatment interval to inter-treatment period may be greater than about 1:7, greater than about 1:8, greater than about 1:9, greater than about 1:10, greater than about 1:11, greater than about 1:12, greater than about 1:15, greater than about 1:18, etc., and/or between about 1:6 and 1:1000, between about 1:6 and 1:500, between about 1:6 and 1:100, between about 1:6 and 1:75, between about 1:6 and 1:50, between about 1:8 and 1:1000, between about 1:8 and 1:500, between about 1:8 and 1:100, etc.
  • the apparatuses described herein may be generally configured to be worn against the body (e.g., incorporated into a garment, jewelry, hat, bed, chair, etc.), and may be specifically adapted/configured to deliver non-invasive, relatively low-energy or very low-energy PEMF treatment in which the ratio of treatment interval to inter-treatment period is as described herein.
  • methods for treating a patient may generally include: generating a pulsed electromagnetic field (PEMF) from a pulsed electromagnetic field source; applying the pulsed electromagnetic field in proximity to a target region affected by an injury or condition to reduce a physiological response to the injury or condition for a treatment interval that is greater than 10 minutes; discontinuing the application of the pulsed electromagnetic field for an inter-treatment period that is greater than six times the treatment interval; and repeating, for a plurality of times, the steps of generating, applying and discontinuing.
  • PEMF pulsed electromagnetic field
  • a pulsed electromagnetic field source may include any apparatus, including those described herein or otherwise known in the art, that can be used to apply relatively low-energy PEMF signals. Examples of such devices and PEMF signals (including low-energy PEMF signals) are described, for example, in U.S. patent applications: U.S. Pat. Nos.
  • a relatively low-energy PEMF waveform may generally apply milliTesla (mT), e.g., between about 1 and 100 mT, magenitc field strength, or average magnetic field strength.
  • Very low-energy PEMF may apply microTesla ( ⁇ T), e.g., less than 1 mT, less than 100 ⁇ T, less than 50 ⁇ T, less than 20 ⁇ T, less than 10 ⁇ T, less than 5 ⁇ T, etc.
  • the PEMF signal though relatively or very low energy may be applied in the specific pulsed waveforms as described herein to any target region.
  • a target region may be any body region, including surface (e.g., skin) or internal (e.g., brain, organ, etc.) region, particularly those that are more superficially located. Wounds such as surgical wounds are an example of a target region. Nerves, including spinal, peripheral or central (e.g., brain) nervous system regions may also be treated as described herein. The treatments described herein may be used to treat a medical disorder, and may modulate or improve a physiological response to the injury or condition, including but not limited to, swelling/inflammation, necrosis, healing (e.g., tissue growth, cell migration), scarring, etc.
  • a treatment interval may include the period during which treatment (PEMF energy) is actively being applied, for example, as a burst or plurality of bursts of pulses.
  • the waveforms may be applied at a regular, irregular or random duration, period or wave-shape within the burst (envelope) and the amplitude of the envelope may be regular (e.g., sinusoidal, square, etc.), irregular, or random.
  • sinusoidal pulses at a carrier frequency may be applied within a rectangular envelope that had a burst duration (e.g., greater than 200 microseconds, greater than 300 microseconds, greater than 400 microseconds, greater than 500 microseconds, greater than 600 microseconds, between 500 microseconds and 1 second, etc.), and may be repeated at a burst repetition rate.
  • the treatment interval may therefore include quiescent periods, but they are typically part of the inter-pulse or inter-burst periods defining the periodicity of the waveforms or burst of waveforms.
  • a treatment interval may be between 5 and 600 minutes, but more likely between 5 and 50 minutes. As described herein, it may be particularly efficacious to apply treatment for a treatment interval of greater than about 10 minutes, e.g., greater than about 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, etc. minutes.
  • the treatment periods may be divided by off-times (inter-treatment periods) during which no PEMF signals are applied by the apparatus, which may be particularly configured to prevent the application of any PEMF signal during this inter-treatment period.
  • the pulsed electromagnetic field is configured to simultaneously increase the rate of ion-dependent signaling and to minimize the rate of inhibition of such signaling by natural compounds.
  • This period may be referred to as a period during which the PEMF signal application is discontinued, e.g., discontinuing the application of the pulsed electromagnetic field for an inter-treatment period (or “offtime”). Following the discontinuation, another treatment period (and another inter-treatment period, off-time) may be repeated.
  • the waveforms applied during the subsequent treatment periods may be the same or different. For example, the waveform characteristics, such as amplitude, frequency (e.g.
  • burst frequency and/or pulse frequency and/or carrier wave frequency), duration (burst duration, pulse duration), and/or waveform and/or envelope (burst) shape may be different between, or in some variations within, subsequent treatment periods.
  • the duration of the subsequent treatment period may be the same or different, though they may still be greater than some minimum treatment period duration (e.g., greater than 10 min, 11 min, 12 min, 13 min, 14 min, 15 min, 20 min, 25 min, 30 min, 40 min, 50 min, 60 min, etc.) and/or less than a maximum treatment period duration (e.g., less than 50 min, 60 min, 70 min, 80 min, 90 min, 2 hrs, 2.5 hrs, 3 hrs, etc.), where the minimum is always less than the maximum.
  • the method or apparatus may adjust one or more characteristics of the treatment period and/or the treatment period duration based on feedback measured or otherwise received from the patient, including feedback based on a detected level of a biomarker.
  • the pulsed electromagnetic field may be configured to simultaneously increase the rate of cGMP signaling and to minimize the rate of inhibition of such signaling by compounds such as phosphodiesterase (PDE).
  • the pulsed electromagnetic field may be configured to simultaneously increase the rate of cGMP signaling and to minimize the rate of inhibition of such signaling by compounds such as phosphodiesterase (PDE), is applied for a duration consistent with the above.
  • the application treatment may include any appropriate number of repetitions, and may be opend-ended (e.g., stopped manually by the patient and/or a medical professional).
  • repeating may comprise repeating for at least 10 times, 11 times, 12 times, 20 times, 30 times, 40 times, 50 times, etc. or for some minimum time interval (e.g., 20 min, 25 min, 30 min, 35 min, 40 min, 50 min, 60 min, 90 min, 2 hrs, 4 hrs, 8 hrs, 12 hrs, 18 hrs, 24 hrs, 36 hrs, 2 days, 3 days, 4 days, 5 days, 7 days, 14 days, etc.).
  • the pulsed electromagnetic field may consist of a burst having any appropriate relatively or very low PEMF waveform characteristics.
  • the PEMF signal within a treatment period may have a duration of greater than 2 msec of a 27.12 MHz carrier repeating at between 1 and 20 bursts/sec at an amplitude of between 2 and 10 ⁇ T.
  • the pulsed electromagnetic field may include a burst having a duration of between 2 and 10 msec of a carrier wave repeating at between 1 and 10 bursts/sec at an amplitude of between 3 and 8 ⁇ T.
  • the length of the first treatment interval and the length of the inter-treatment period are selected to minimize phosphodiesterase (PDE) production in the patient.
  • PDE phosphodiesterase
  • Any of these methods may also include monitoring the physiological response; and modifying the pulsed electromagnetic field in response to the monitoring step.
  • they may include monitoring the physiological response; and discontinuing treatment once an acceptable level of the physiological response is reached.
  • the methods may also include modulating inflammatory cytokines and growth factors at the target region by applying the pulsed electromagnetic field to simultaneously increase the rate of such modulation and to minimize the rate of inhibition of such modulation by natural compounds. These methods may also include accelerating the healing of the target region by applying the pulsed electromagnetic field to simultaneously increase the rate of healing and to minimize the rate of inhibition of such healing.
  • Applying may include applying the pulsed electromagnetic field in proximity to a target region affected by a neurological injury or condition to reduce a physiological response comprises reducing a concentration of IL-1 ⁇ .
  • these methods may be used to treat any appropriate injury or condition, including a neurodegenerative disease, e.g., Parkinson's disease. Alzheimer's disease, etc.
  • the injury or condition may be a traumatic brain injury (TBI).
  • TBI traumatic brain injury
  • the injury or condition may be a post-operative inflammation and pain.
  • an apparatus for applying pulsed electromagnetic field (PEMF) energy to a subject may include: a generator unit including a signal generator configured to generate a PEMF waveform having configured to simultaneously increase the rate of ion-dependent signaling and to minimize the rate of inhibition of such signaling; a programmable control unit configured to repeatedly provide a signal to the generator unit corresponding to the PEMF waveform for a treatment interval that is 10 minutes or greater followed immediately by an off time having an inter-treatment period that is greater than six times the treatment interval; and an applicator unit configured to be worn by the subject, wherein the generator unit is configured to power the applicator unit to drive transmission of a PEMF signal from the applicator unit based on the PEMF waveform.
  • PEMF pulsed electromagnetic field
  • the programmable control unit may be programmed to provide an inter-treatment period that is between six and 100 times the treatment interval.
  • the programmable control unit may be configured to prevent the generation of a pulsed electromagnetic field during the inter-treatment period.
  • Any of these apparatuses may include a shut off to stop the generator unit during the inter-treatment period.
  • any appropriate applicator may be used, particularly flexible loop applicators, which may be bent or shaped, though remain a coil.
  • the applicator comprises a first loop configured to provide the PEMF waveform and a second loop configured to provide a second PEMF waveform.
  • a programmable control unit may be configured to repeatedly provide the same signal to the generator unit corresponding to the PEMF waveform.
  • FIG. 1A is a schematic representation of the biological EMF transduction pathway which is a representative target pathway of EMF signals configured according to embodiments described herein.
  • FIG. 1B is a flow diagram of a method for treating a patient according to an embodiment of the devices and methods described herein.
  • FIG. 2A is a block diagram of miniaturized circuitry for use with a coil applicator according to some embodiments described.
  • FIG. 2B illustrates a device for application of electromagnetic signals according to an embodiment of the devices and methods described herein.
  • FIG. 2C illustrates a waveform delivered to a target pathway structure of a plant, animal or human, such as a molecule cell, tissue, organ, or partial or entire organism, according to some embodiments described.
  • FIG. 3 is a photograph illustrating a disposable dual coil PEMF device used in some of the embodiments described herein.
  • the disposable dual coil PEMF includes a battery-powered signal generator is at the bottom between the coils.
  • a nonthermal pulse-modulated radio frequency PEMF signal, configured to modulate NO signaling, may be delivered to tissue with a preprogrammed dosing regimen for each cohort.
  • FIG. 4 is a graph showing the effect of different PEMF therapy regimens on the rate of pain reduction following breast reduction surgery in accordance with various embodiments of pain treatments.
  • FIG. 5 is a graph showing the effect of different PEMF therapy regimens in accordance with the embodiments described herein on pain medication usage of patients after surgery.
  • the results show approximately 2-fold more narcotic pills were taken by 24 h postoperatively in the Q4 (sham) and 5/20 (active) cohorts versus the Q4 (active) and Q2 (active) cohorts. There was no significant difference in postoperative narcotic use between the 5/20 (active) and Q4 (sham) cohorts.
  • FIGS. 6A and 6B are graphs showing the effect of signal configuration on NO production from challenged fibroblasts and chondrocytes (respectively) in culture.
  • FIG. 7 is a graph showing the effect of exposure time of a given signal configuration on cGMP production in primary neuronal cells.
  • PEMF Pulsed electromagnetic fields
  • PEMF treatment can also be used to reduce post-operative pain in other surgical procedures.
  • PEMF enhances calmodulin-dependent nitric oxide, which enhances cyclic guanosine monophosphate signaling and phosphodiesterase activity, which blocks cyclic guanosine monophosphate.
  • This invention describes means to configure PEMF dosing to minimize the effect of the competing response of phosphodiesterace activity.
  • FIG. 1B is a flow diagram of a method for treating a subject with a PEMF.
  • one or more (or a range of) waveforms may be determined that target the appropriate pathway for the target tissue.
  • electromagnetic fields are applied to the target location.
  • FIG. 2A illustrates a block diagram of an EMF delivery apparatus as described according to some embodiments.
  • the apparatus may have miniaturized circuitry for use with a coil applicator.
  • the apparatus may include a CPU MODULATOR, a BATTERY MODULE, a POWER SUPPLY, On/Off switch, and an output amplifier, AMP, as illustrated.
  • the CPU MODULATOR may be an 8 bit 4 MHz micro-controller; however, other suitable bit-MHz combination micro-controllers may be used as well.
  • the CPU MODULATOR may be programmed for a given carrier frequency or pulse duration, such as about 27.12 MHz sinusoidal wave.
  • the CPU MODULATOR may be programmed for a given burst duration, for example about 3 msec. In further variations, the CPU MODULATOR may be programmed to provide a given in situ peak electric field, for example 20 V/m; or a given treatment time, for example about 15 minutes; and/or a given treatment regimen, for example about 10 minutes about every hour. The CPU MODULATOR may also be programmed to deliver an EMF waveform to the target ion binding pathway.
  • Some embodiments combine the signal generation and coil or electrode applicator into one portable or disposable unit, such as illustrated in FIG. 2B (which will be described in greater detail below) for the case of an inductively coupled signal.
  • the electrical coils when electrical coils are used as the applicator, the electrical coils can be powered with a time varying magnetic field that induces a time varying electric field in a target pathway structure according to Faraday's law.
  • An electromagnetic field generated by a circuit such as shown in FIG. 2A can also be applied using electrochemical coupling, wherein electrodes are in direct contact with skin or another outer electrochemically conductive boundary of a target pathway structure.
  • the electromagnetic field generated by the generating circuit of FIG. 2A can also be applied using electrostatic coupling wherein an air gap exists between a generating device such as an electrode and a target pathway structure such as a molecule, cell, tissue, and organ of a plant animal or human.
  • a generating device such as an electrode
  • a target pathway structure such as a molecule, cell, tissue, and organ of a plant animal or human.
  • the ultra lightweight coils and miniaturized circuitry allow for use with common physical therapy treatment modalities and at any location on a plant, animal or human for which any therapeutic or prophylactic effect is desired.
  • An advantageous result of application of some embodiments described is that a living organism's wellbeing can be maintained and enhanced.
  • an embodiment according to the present invention of an induced electric field waveform delivered to a target pathway structure is illustrated.
  • burst duration and period are represented by T1 and T2, respectively.
  • the signal within the rectangular box designated at T1 can be, rectangular, sinusoidal, chaotic or random, provided that the waveform duration or carrier period is less than one-half of the target ion bound time.
  • the peak induced electric field is related to the peak induced magnetic field, shown as B in FIG. 2C , via Faraday's Law of Induction.
  • FIG. 2B illustrates an embodiment of an apparatus 200 that may be used.
  • the apparatus is constructed to be self-contained, lightweight, and portable.
  • a circuit control/signal generator 201 may be held within a (optionally wearable) housing and connected to a generating member such as an electrical coil 202 .
  • the circuit control/signal generator 201 is constructed in a manner that given a target pathway within a target tissue, it is possible to choose waveform parameters that satisfy a frequency response of the target pathway within the target tissue.
  • circuit control/signal generator 201 applies mathematical models or results of such models that approximate the kinetics of ion binding in biochemical pathways. Waveforms configured by the circuit control/signal generator 201 are directed to a generating member 202 .
  • the generating member 202 comprises electrical coils that are pliable and comfortable.
  • the generating member 202 is made from one or more turns of electrically conducting wire in a generally circular or oval shape, any other suitable shape.
  • the electrical coil is a circular wire applicator with a diameter that allows encircling of a subject's cranium. In some embodiments, the diameter is between approximately 6-8 inches. In general, the size of the coil may be fixed or adjustable and the circuit control/signal generator may be matched to the material and the size of the applicator to provide the desired treatment.
  • the apparatus 200 may deliver a pulsing magnetic field that can be used to provide treatment.
  • the device 200 may apply a pulsing magnetic field for a prescribed time and can automatically repeat applying the pulsing magnetic field for as many applications as are needed in a given time period, e.g. 6-12 times a day.
  • the device 200 can be configured to apply pulsing magnetic fields for any time repetition sequence. Without being bound to any theory, it is believed that when electrical coils are used as a generating member 202 , the electrical coils can be powered with a time varying magnetic field that induces a biologically and therapeutically effective time varying electric field in a target tissue location.
  • an electromagnetic field generated by the generating member 202 can be applied using electrochemical coupling, wherein electrodes are in direct contact with skin or another outer electrically conductive boundary of the target tissue (e.g. skull or scalp).
  • the electromagnetic field generated by the generating member 202 can also be applied using electrostatic coupling wherein an air gap exists between a generating member 202 such as an electrode and the target tissue.
  • a signal generator and battery is housed in the miniature circuit control/signal generator 201 and the miniature circuit control/signal generator 201 may contain an on/off switch and light indicator.
  • the activation and control of the treatment device may be done via remote control such as by way of a fob that may be programmed to interact with a specific individual device.
  • the treatment device further includes a history feature that records the treatment parameters carried out by the device such that the information is recorded in the device itself and/or can be transmitted to another device such as computer, smart phone, printer, or other medical equipment/device.
  • the treatment device 200 has adjustable dimensions to accommodate fit to a variety of patient sizes and anatomy.
  • the generating member 202 may comprise modular components which can be added or removed by mated attaching members.
  • the treatment device 200 may contain a detachable generating member (e.g. detachable circular coil or other configurations) that can be removed and replaced with configurations that are better suited for the particular patient's needs.
  • a circular coil generating member 202 may be removed and replaced with an elongate generating member such that PEMF treatment can be applied where other medical equipment may obstruct access by a circular generating member 202 .
  • the generating member may be made from Litz wire that allows the generating member to flex and fold to accommodate different target areas or sizes.
  • the PEMF devices disclosed herein can be used to treat patients with post-operative pain.
  • Acute postoperative pain is a significant medical problem. Postoperative pain must be managed effectively to optimize surgical outcomes, reduce morbidity, shorten the duration of hospital stay, and control ever-increasing health-care costs [1].
  • pain mechanisms involve increased sensitivity of nociceptors due to increased presence of proinflammatory cytokines in the wound milieu [2].
  • Narcotics are most commonly used to treat postoperative pain; however, narcotics do not reduce nociceptor sensitivity and cause undesirable side effects and potential addiction.
  • Alternative approaches to decrease post-operative pain involve slowing the appearance of proinflammatory agents at the surgical site [2].
  • NO/cGMP CaM-dependent nitric oxide/cyclic guanosine monophosphate
  • FIG. 1A This mechanism is schematically represented in FIG. 1A .
  • PEMF modulation of angiogenesis via effects on FGF-2 has been reported [10-15].
  • the PEMF effect could be blocked with an FGF-2 inhibitor, consistent with a PEMF effect on NO/cGMP signaling [12, 13].
  • PEMF has been reported to accelerate postoperative pain decrease, with a concomitant reduction in narcotic requirements, in double-blinded, randomized clinical studies on breast reduction (BR) [16], breast augmentation [17, 18], and autologous flap breast reconstruction [19].
  • BR breast reduction
  • the BR study also showed that PEMF reduced inflammation by reducing IL-1 beta more than two-fold in the wound exudate, which correlated with the higher rate of pain reduction from PEMF [16].
  • PEMF can and has been used throughout the body, including after abdominoplasties, major intra-abdominal surgery, extremity procedures, and facial fat grafting [20, 21].
  • a number of patients with post-operative pain were treated with PEMF and studied.
  • two prospective, nonrandomized, active cohorts of breast reduction patients with 15 min PEMF per 2 h; “Q2 (active)”, and 5 min PEMF per 20 min; “5/20 (active)”, dosing regimens were added to a double-blind clinical study wherein 20 min PEMF per 4 h, “Q4 (active)”, dosing was shown to significantly accelerate postoperative pain reduction compared with Q4 shams.
  • Postoperative visual analog scale pain scores and narcotic use were compared.
  • pain at 24 h postoperative was, respectively, 43% and 35% of pain at 1 h postoperative for patients in the Q4 (active) and Q2 (active) cohorts (P ⁇ 0.01).
  • PEMF signal parameters were identical for all patients in active cohorts.
  • the dosing change was treatment regimen.
  • the rate of increase in CaM-dependent NO, and therefore cGMP, from PEMF in tissue for the 5/20 regimen is nearly 2.5-fold higher than that for the Q2 (active) and 4-fold higher than that for the Q4 (active) regimens.
  • the NO/cGMP signaling pathway is a principal anti-inflammatory pathway.
  • CaM-dependent PDE activity regulates this pathway by inhibiting cGMP.
  • the PEMF signal used in this study is known to enhance NO/cGMP signaling, and to enhance CaM-dependent PDE activity [5-7]. It was proposed that the 5/20 regimen caused PDE activity to predominate, thereby inhibiting all the enhanced cGMP produced by PEMF. The result is no effect of PEMF on postoperative pain.
  • PEMF effects depend on signal configuration. The first showed that the PEMF effect on breast cancer cell apoptosis was significant when the same waveform, applied for the same exposure time, repeated at 20 Hz but not at 50 Hz [35]. The second study showed that PEMF significantly reduced the expression of inflammatory markers, tumor necrosis factor, and nuclear factor-kappa beta in challenged macrophages when the same waveform, applied for the same exposure time, was repeated at 5 Hz, but not at 15 or 30 Hz [36].
  • Two PEMF signals with 27.12 MHz sinusoidal carrier were tested.
  • One PEMF signal had a burst width of 2 msec repeating at 2 Hz (Signal I) while the second signal (Signal II) had a burst width of 3 msec repeating at 5 Hz.
  • Both signals were tested at amplitudes ranging from 0.5 to 8 ⁇ T. Exposure time was 15 min for each amplitude condition. Since each waveform was configured to target Ca/CaM binding, Signal II would be expected to produce approximately 4-fold more NO than that produced by Signal I, at all amplitudes studied.
  • a PEMF signal consisting of a 2 msec burst of a 27.12 MHz carrier repeating at 2 Hz and delivering 4 ⁇ T amplitude was tested.
  • Primary neuronal cells were subjected to oxygen glucose deprivation (OGD) which subjects the cells to ischemic conditions such as those which exist in cardiac and brain ischemia.
  • OGD oxygen glucose deprivation
  • the parameters of the PEMF signal were chosen to modulate CaM/NO/cGMP signaling so that exposure to PEMF during OGD would be expected to produce increased cGMP.
  • the PEMF effect on PDE activity also increases.
  • an exposure time of 60 minutes would be expected to produce about 4-fold more NO than the standard effective 15 minute exposure.
  • the results are shown in FIG. 7 wherein it may be seen that 15 minute PEMF exposure maximally restored cGMP production. In contrast, an exposure time of 60 minutes was not effective.
  • any of the PEMF parameters can be selected to minimize the PDE inhibition of cGMP and/or maximize the production of cGMP.
  • the length of the inter-treatment period and the length of the treatment interval are selected to minimize the PDE inhibition of cGMP.
  • Any of the PEMF waveform parameters can be optimized in conjunction with the length of the treatment interval and inter-treatment period to achieve a desired change to the production of PDE and/or to decrease the inhibition of cGMP by PDE.
  • the device applies an envelope of high-frequency waveforms at low amplitude (e.g. less than 50 milliGauss, less than 100 milliGauss, less than 200 milliGauss, etc.) 105 .
  • This envelope of high-frequency pulses is then repeated at a particular frequency (repetition rate) after an appropriate delay.
  • the repetition rate may be varied to minimize PDE inhibition of PDE.
  • the amplitude may be varied to minimize PDE inhibition of PDE.
  • the burst duration may be varied to minimize PDE inhibition of PDE.
  • the initial signal configuration (burst duration, burst repetition and amplitude) can be repeated for a first treatment time and then followed by a delay during which the treatment is “off” 107 .
  • This waiting interval (inter-treatment interval) may last for minutes or hours and then the treatment interval may be repeated again until the treatment regime is complete 109 .
  • the length of the inter-treatment period can be selected to minimize the PDE inhibition of cGMP.
  • the inter-treatment period is greater than about 15 minutes. In some embodiments the inter-treatment period is greater than about 30 minutes. In some embodiments the inter-treatment period is greater than about 60 minutes. In some embodiments the inter-treatment period is greater than about 90 minutes. In some embodiments the inter-treatment period is greater than about 120 minutes. In some embodiments the inter-treatment period is greater than about 180 minutes. In some embodiments the inter-treatment period is greater than about 240 minutes
  • the inter-treatment period can be expressed as a multiple of the PEMF treatment interval. In some embodiments the inter-treatment period is at least three times longer than the treatment interval. In some embodiments the inter-treatment period is at least four times longer than the treatment interval. In some embodiments the inter-treatment period is at least five times longer than the treatment interval. In some embodiments the inter-treatment period is at least six times longer than the treatment interval. In some embodiments the inter-treatment period is at least seven times longer than the treatment interval. In some embodiments the inter-treatment period is at least eight times longer than the treatment interval. In some embodiments the inter-treatment period is at least ten times longer than the treatment interval. In some embodiments the inter-treatment period is at least fifteen times longer than the treatment interval. In some embodiments the inter-treatment period is at least twenty times longer than the treatment interval.
  • any of the PEMF treatment intervals disclosed herein can be used with any of the inter-treatment intervals disclosed herein.
  • the treatment interval is about 5 minutes or longer. In some embodiments the treatment interval is about 10 minutes or longer. In some embodiments the treatment interval is about 15 minutes or longer. In some embodiments the treatment interval is about 20 minutes or longer.
  • the PEMF treatment period is five minutes with a 15 minute inter-treatment period. In some embodiments the PEMF treatment period is 15 minutes with a 105 minute inter-treatment period (e.g. 15 minutes of PEMF treatment per two hours). In some embodiments the PEMF treatment period is 20 minutes with a 160 minute inter-treatment period (e.g. 20 minutes of PEMF treatment per three hours).
  • the treatment device is pre-programmed (or configured to receive pre-programming) to execute the entire treatment regime (including multiple on-periods and/or intra-treatment intervals) punctuated by predetermined off-periods (inter-treatment intervals) when no treatment is applied.
  • the device is pre-programmed to emit a PEMF signal at 27.12 MHz at 2 msec bursts repeating at 2 bursts/sec.
  • the device is pre-programmed to emit a PEMF signal at 27.12 MHz (at about amplitude 250-400 mV/cm) pulsed in 4 msec bursts at 2 Hz.
  • the method may include a pulsed electromagnetic field comprising a 2 msec burst of 27.12 MHz sinusoidal waves repeating at 2 Hz. In other variations, the method may include a pulsed electromagnetic field comprising a 3 msec burst of 27.12 MHz sinusoidal waves repeating at 2 Hz. In further embodiments, the pulsed electromagnetic field may comprise a 4 msec burst of 27.12 MHz sinusoidal waves repeating at 2 Hz.
  • the patient can be monitored during the PEMF treatment regime to determine the physiological response to the PEMF treatment regime.
  • the treatment cycle e.g. treatment period and inter-treatment period
  • the treatment cycle can be repeated until a desired physiological response is achieved.
  • the subsequent treatment cycle parameters can be adjusted by a health professional to achieve a desired physiological response in the patient.
  • references to a structure or feature that is disposed “adjacent” another feature may have portions that overlap or underlie the adjacent feature.
  • spatially relative terms such as “under”, “below”, “lower”, “over”, “upper” and the like, may be used herein for ease of description to describe one element or feature's relationship to another element(s) or feature(s) as illustrated in the figures. It will be understood that the spatially relative terms are intended to encompass different orientations of the device in use or operation in addition to the orientation depicted in the figures. For example, if a device in the figures is inverted, elements described as “under” or “beneath” other elements or features would then be oriented “over” the other elements or features. Thus, the exemplary term “under” can encompass both an orientation of over and under.
  • the device may be otherwise oriented (rotated 90 degrees or at other orientations) and the spatially relative descriptors used herein interpreted accordingly.
  • the terms “upwardly”, “downwardly”, “vertical”, “horizontal” and the like are used herein for the purpose of explanation only unless specifically indicated otherwise.
  • first and second may be used herein to describe various features/elements (including steps), these features/elements should not be limited by these terms, unless the context indicates otherwise. These terms may be used to distinguish one feature/element from another feature/element. Thus, a first feature/element discussed below could be termed a second feature/element, and similarly, a second feature/element discussed below could be termed a first feature/element without departing from the teachings of the present invention.
  • a numeric value may have a value that is +/ ⁇ 0.1% of the stated value (or range of values), +/ ⁇ 1% of the stated value (or range of values), +/ ⁇ 2% of the stated value (or range of values), +/ ⁇ 5% of the stated value (or range of values), +/ ⁇ 10% of the stated value (or range of values), etc.
  • Any numerical values given herein should also be understood to include about or approximately that value, unless the context indicates otherwise. For example, if the value “10” is disclosed, then “about 10” is also disclosed. Any numerical range recited herein is intended to include all sub-ranges subsumed therein.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Radiology & Medical Imaging (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Hospice & Palliative Care (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Neurology (AREA)
  • Magnetic Treatment Devices (AREA)

Abstract

Embodiments of the invention include methods of treating a patient with physiological responses arising from an injury or condition, such as post-operative pain, traumatic brain injury, and cognitive defects. These treatment methods can include the steps of generating a pulsed electromagnetic field from a pulsed electromagnetic field source which is configured to simultaneously increase the rate of ion-dependent signaling, such as CaM/NO/cGMP signaling and to minimize the rate of inhibition of such signaling by natural compounds and applying the pulsed electromagnetic field in proximity to a target region affected by the injury or condition for a first treatment interval followed by an inter-treatment period with no electromagnetic field between treatment intervals to reduce a physiological response to the injury or condition.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. provisional patent application No. 62/075,122, filed on Nov. 4, 2014, and titled “METHOD AND APPARATUS FOR ELECTROMAGNETIC TREATMENT OF LIVING SYSTEMS,” the entirety of which is herein incorporated by reference.
  • INCORPORATION BY REFERENCE
  • All publications and patent applications mentioned in this specification are herein incorporated by reference in their entirety to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
  • FIELD
  • The disclosure relates generally to the field of therapeutic devices and methods for treating patients.
  • BACKGROUND
  • Described herein are electromagnetic treatment devices, systems and methods. Some embodiments pertain generally to a method and apparatus for therapeutic and prophylactic treatment of animal and human cells and tissues. In particular, some embodiments pertain to use of non-thermal time-varying electromagnetic fields configured to accelerate the asymmetrical kinetics of the binding of intracellular ions to their respective binding proteins which regulate the biochemical signaling pathways living systems employ to reduce the inflammatory response to injury, and to enhance healing and well-being. Other embodiments pertain to the non-thermal application of repetitive pulse bursts of sinusoidal, rectangular, chaotic or arbitrary waveform electromagnetic fields to instantaneously accelerate ion-buffer binding in signaling pathways in animal and human cells and tissues using ultra lightweight portable coupling devices such as inductors and electrodes, driven by miniature signal generator circuitry that can be incorporated into an anatomical positioning device such as a dressing, bandage, compression bandage, compression dressing; lumbar or cervical back, shoulder, head, neck and other body portion wraps and supports; garments, hats, caps, helmets, mattress pads, seat cushions, beds, stretchers, and other body supports in cars, motorcycles, buses, trains, airplanes, boats, ships and the like.
  • In some embodiments, the proposed EMF transduction pathway relevant to tissue maintenance, repair and regeneration, begins with voltage-dependent Ca2+ binding to CaM, which is favored when cytosolic Ca2+ homeostasis is disrupted by chemical and/or physical insults at the cellular level. Ca/CaM binding produces activated CaM that binds to, and activates, cNOS, which catalyzes the synthesis of the signaling molecule NO from L-arginine. This pathway is shown in its simplest schematic form in FIG. 1A. FIG. 1A is a schematic summary of the body's primary anti-inflammatory cascade and the proposed manner by which PEMF may accelerate postoperative pain relief. Surgical injury increases cytosolic Ca2+, which activates CaM. PEMF accelerates CaM activation thereby enhancing NO/cGMP anti-inflammatory signaling. PEMF also enhances CaM-dependent PDE activation, which accelerates cGMP inhibition. PEMF dosing must take into account the competing dynamics of NO/cGMP signaling and PDE inhibition of cGMP.
  • As shown in FIG. 1A, cNOS* represents activated constitutive nitric oxide synthase (cNOS), which catalyzes the production of NO from L-arginine, which, in turn, activates soluble gyanylyl cyclase, sGC. The term “sGC*” refers to activated guanylyl cyclase which catalyzes cyclic guanosine monophosphate (cGMP) formation when NO signaling modulates the tissue repair pathway. “AC*” refers to activated adenylyl cyclase, which catalyzes cyclic adenosine monophosphate (cAMP) when NO signaling modulates differentiation and survival.
  • According to some embodiments, an EMF signal can be configured to accelerate cytosolic ion binding to a cytosolic buffer, such as voltage dependent Ca2+ binding to CaM, because the rate constant for binding, kon is much greater than the rate constant for unbinding, koff, imparting rectifier-like properties to ion-buffer binding, such as Ca2+ binding to CaM.
  • Yet another embodiment pertains to application of sinusoidal, rectangular, chaotic or arbitrary waveform electromagnetic signals, having frequency components below about 100 GHz, configured to accelerate the binding of intracellular Ca2+ to a buffer, such as CaM, to enhance biochemical signaling pathways in animal and human cells and tissues. Signals configured according to additional embodiments produce a net increase in a bound ion, such as Ca2+, at CaM binding sites because the asymmetrical kinetics of Ca/CaM binding allows such signals to accumulate voltage induced at the ion binding site, thereby accelerating voltage-dependent ion binding. Examples of therapeutic and prophylactic applications are modulation of biochemical signaling in anti-inflammatory pathways, modulation of biochemical signaling in cytokine release pathways, modulation of biochemical signaling in growth factor release pathways; edema and lymph reduction, anti-inflammatory, post-surgical and post-operative pain and edema relief, nerve, bone and organ pain relief, increased local blood flow, microvascular blood perfusion, treatment of tissue and organ ischemia, brain tissue ischemia from stroke or traumatic brain injury, treatment of neurological injury and neurodegenerative diseases such as Alzheimer's and Parkinson's; angiogenesis, neovascularization; enhanced immune response; enhanced effectiveness of pharmacological agents; nerve regeneration; prevention of apoptosis; modulation of heat shock proteins for prophylaxis and response to injury or pathology.
  • In some variations the systems, devices and/or methods generally relate to application of electromagnetic fields (EMF), and in particular, pulsed electromagnetic fields (PEMF), including a subset of PEMF in a radio frequency domain (e.g., pulsed radio frequency or PRF), for the treatment of any of the applications disclosed herein in animals and humans, including pain, edema, tissue repair and head, cerebral and neural injury, and neurodegenerative conditions.
  • Transient elevations in cytosolic Ca2+, from external stimuli as simple as changes in temperature and receptor activation, or as complex as mechanical disruption of tissue, will activate CaM. Once Ca2+ ions are bound, a conformational change will allow CaM bind to and activate a number of key enzymes involved in cell viability and function, such as the endothelial and neuronal constitutive nitric oxide synthases (cNOS); eNOS and nNOS, respectively. As a consequence, NO is rapidly produced, albeit in lower concentrations than the explosive increases in NO produced by inducible NOS (iNOS), during the inflammatory response. In contrast, these smaller, transient increases in NO produced by Ca/CaM-binding will activate soluble guanylyl cyclase (sGC), which will catalyze the formation of cyclic guanosine monophosphate (cGMP). The CaM/NO/cGMP signaling pathway can rapidly modulate blood flow in response to normal physiologic demands, as well as to inflammation. Importantly, this same pathway will also rapidly attenuate expression of cytokines such as interleukin-1beta (IL-1β), and iNOS and stimulate anti-apoptotic pathways in neurons. All of these effects are mediated by calcium and cyclic nucleotides, which in turn regulate growth factors such as basic fibroblast growth factor (FGF-2) and vascular endothelial growth factor (VEGF), resulting in pleiotrophic effects on cells involved in tissue repair and maintenance. PEMF can also accelerate the inhibition of cGMP by phosphodiesterase (PDE). Improved PEMF signal configurations and treatment regimens are disclosed herein that can minimize the inhibition of cGMP by PDE.
  • Therefore, a need exists for an apparatus and a method that modulates the biochemical pathways that regulate animal and human tissue response to maximize the rate of cGMP production while minimizing the rate of inhibition of cGMP. In some embodiments, an apparatus incorporates miniaturized circuitry and light weight coil applicators or electrodes to deliver any of the waveforms described herein thus allowing the apparatus to be low cost, portable and, if desired, disposable.
  • SUMMARY OF THE DISCLOSURE
  • The present invention relates to methods and apparatuses for treating patients with pulsed electromagnetic therapies (PEMF). The PEMF waveform and the period between PEMF waveform pulses can be configured to simultaneously increase the rate of cGMP production and to minimize the rate of inhibition of cGMP by compounds such as PDE.
  • In particular, described herein are methods of optimizing PEMF treatment based on the surprising finding that the effectiveness of a non-invasive, relatively low-energy or very low-energy PEMF treatment depends on the ratio of the duration of the treatment interval and the duration of the inter-treatment interval (also referred to herein as the inter-treatment period). The methods described herein are invented from the new finding that the for externally-applied low-energy or (in some variations) very low-energy PEMF treatments, a ratio of treatment interval to inter-treatment period of greater than about 1:6 (e.g., an inter-treatment period that is greater than six times the treatment interval) results in efficacious treatment, whereas ratios less than 1:6 do not. In addition, in some variations it may be beneficial to have ratios of treatment interval to inter-treatment period of less than about 1:100. For example, the ratio of treatment interval to inter-treatment period may be greater than about 1:7, greater than about 1:8, greater than about 1:9, greater than about 1:10, greater than about 1:11, greater than about 1:12, greater than about 1:15, greater than about 1:18, etc., and/or between about 1:6 and 1:1000, between about 1:6 and 1:500, between about 1:6 and 1:100, between about 1:6 and 1:75, between about 1:6 and 1:50, between about 1:8 and 1:1000, between about 1:8 and 1:500, between about 1:8 and 1:100, etc.
  • The apparatuses described herein may be generally configured to be worn against the body (e.g., incorporated into a garment, jewelry, hat, bed, chair, etc.), and may be specifically adapted/configured to deliver non-invasive, relatively low-energy or very low-energy PEMF treatment in which the ratio of treatment interval to inter-treatment period is as described herein.
  • For example described herein are methods for treating a patient (e.g., human, animal, etc.) that may generally include: generating a pulsed electromagnetic field (PEMF) from a pulsed electromagnetic field source; applying the pulsed electromagnetic field in proximity to a target region affected by an injury or condition to reduce a physiological response to the injury or condition for a treatment interval that is greater than 10 minutes; discontinuing the application of the pulsed electromagnetic field for an inter-treatment period that is greater than six times the treatment interval; and repeating, for a plurality of times, the steps of generating, applying and discontinuing.
  • A pulsed electromagnetic field source may include any apparatus, including those described herein or otherwise known in the art, that can be used to apply relatively low-energy PEMF signals. Examples of such devices and PEMF signals (including low-energy PEMF signals) are described, for example, in U.S. patent applications: U.S. Pat. Nos. 7,744,524, 7,740,574, 8,415,123, 7,758,490, 7,896,797 and 8,343,027, and pending applications no.: US-2010-0210893, US-2010-0222631, US-2013-0274540, US-2014-0046115, US-2014-0046117, US-2011-0207989, US-2012-0116149, US-2014-0213843, US-2014-0213844, and US-2012-0089201-A1. Each of these patents and pending applications is herein incorporated by reference in its entirety, and in particular for its teaching of PEMF application devices, waveforms, and therapies.
  • A relatively low-energy PEMF waveform may generally apply milliTesla (mT), e.g., between about 1 and 100 mT, magenitc field strength, or average magnetic field strength. Very low-energy PEMF may apply microTesla (μT), e.g., less than 1 mT, less than 100 μT, less than 50 μT, less than 20 μT, less than 10 μT, less than 5 μT, etc. The PEMF signal, though relatively or very low energy may be applied in the specific pulsed waveforms as described herein to any target region.
  • A target region may be any body region, including surface (e.g., skin) or internal (e.g., brain, organ, etc.) region, particularly those that are more superficially located. Wounds such as surgical wounds are an example of a target region. Nerves, including spinal, peripheral or central (e.g., brain) nervous system regions may also be treated as described herein. The treatments described herein may be used to treat a medical disorder, and may modulate or improve a physiological response to the injury or condition, including but not limited to, swelling/inflammation, necrosis, healing (e.g., tissue growth, cell migration), scarring, etc.
  • In general, a treatment interval may include the period during which treatment (PEMF energy) is actively being applied, for example, as a burst or plurality of bursts of pulses. The waveforms may be applied at a regular, irregular or random duration, period or wave-shape within the burst (envelope) and the amplitude of the envelope may be regular (e.g., sinusoidal, square, etc.), irregular, or random. In particular, sinusoidal pulses at a carrier frequency (e.g., of 27.12 or a harmonic thereof) may be applied within a rectangular envelope that had a burst duration (e.g., greater than 200 microseconds, greater than 300 microseconds, greater than 400 microseconds, greater than 500 microseconds, greater than 600 microseconds, between 500 microseconds and 1 second, etc.), and may be repeated at a burst repetition rate. The treatment interval may therefore include quiescent periods, but they are typically part of the inter-pulse or inter-burst periods defining the periodicity of the waveforms or burst of waveforms. A treatment interval may be between 5 and 600 minutes, but more likely between 5 and 50 minutes. As described herein, it may be particularly efficacious to apply treatment for a treatment interval of greater than about 10 minutes, e.g., greater than about 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, etc. minutes.
  • During any of the treatments described herein the treatment periods may be divided by off-times (inter-treatment periods) during which no PEMF signals are applied by the apparatus, which may be particularly configured to prevent the application of any PEMF signal during this inter-treatment period.
  • The method of claim 1, wherein the pulsed electromagnetic field is configured to simultaneously increase the rate of ion-dependent signaling and to minimize the rate of inhibition of such signaling by natural compounds. This period may be referred to as a period during which the PEMF signal application is discontinued, e.g., discontinuing the application of the pulsed electromagnetic field for an inter-treatment period (or “offtime”). Following the discontinuation, another treatment period (and another inter-treatment period, off-time) may be repeated. The waveforms applied during the subsequent treatment periods may be the same or different. For example, the waveform characteristics, such as amplitude, frequency (e.g. burst frequency and/or pulse frequency and/or carrier wave frequency), duration (burst duration, pulse duration), and/or waveform and/or envelope (burst) shape, may be different between, or in some variations within, subsequent treatment periods. The duration of the subsequent treatment period may be the same or different, though they may still be greater than some minimum treatment period duration (e.g., greater than 10 min, 11 min, 12 min, 13 min, 14 min, 15 min, 20 min, 25 min, 30 min, 40 min, 50 min, 60 min, etc.) and/or less than a maximum treatment period duration (e.g., less than 50 min, 60 min, 70 min, 80 min, 90 min, 2 hrs, 2.5 hrs, 3 hrs, etc.), where the minimum is always less than the maximum. In some variations, the method or apparatus may adjust one or more characteristics of the treatment period and/or the treatment period duration based on feedback measured or otherwise received from the patient, including feedback based on a detected level of a biomarker.
  • The pulsed electromagnetic field may be configured to simultaneously increase the rate of cGMP signaling and to minimize the rate of inhibition of such signaling by compounds such as phosphodiesterase (PDE). The pulsed electromagnetic field may be configured to simultaneously increase the rate of cGMP signaling and to minimize the rate of inhibition of such signaling by compounds such as phosphodiesterase (PDE), is applied for a duration consistent with the above.
  • The application treatment may include any appropriate number of repetitions, and may be opend-ended (e.g., stopped manually by the patient and/or a medical professional). For example repeating may comprise repeating for at least 10 times, 11 times, 12 times, 20 times, 30 times, 40 times, 50 times, etc. or for some minimum time interval (e.g., 20 min, 25 min, 30 min, 35 min, 40 min, 50 min, 60 min, 90 min, 2 hrs, 4 hrs, 8 hrs, 12 hrs, 18 hrs, 24 hrs, 36 hrs, 2 days, 3 days, 4 days, 5 days, 7 days, 14 days, etc.).
  • As mentioned, the pulsed electromagnetic field may consist of a burst having any appropriate relatively or very low PEMF waveform characteristics. For example, the PEMF signal within a treatment period may have a duration of greater than 2 msec of a 27.12 MHz carrier repeating at between 1 and 20 bursts/sec at an amplitude of between 2 and 10 μT. The pulsed electromagnetic field may include a burst having a duration of between 2 and 10 msec of a carrier wave repeating at between 1 and 10 bursts/sec at an amplitude of between 3 and 8 μT.
  • The length of the first treatment interval and the length of the inter-treatment period are selected to minimize phosphodiesterase (PDE) production in the patient.
  • Any of these methods may also include monitoring the physiological response; and modifying the pulsed electromagnetic field in response to the monitoring step. For example, they may include monitoring the physiological response; and discontinuing treatment once an acceptable level of the physiological response is reached.
  • The methods may also include modulating inflammatory cytokines and growth factors at the target region by applying the pulsed electromagnetic field to simultaneously increase the rate of such modulation and to minimize the rate of inhibition of such modulation by natural compounds. These methods may also include accelerating the healing of the target region by applying the pulsed electromagnetic field to simultaneously increase the rate of healing and to minimize the rate of inhibition of such healing.
  • Applying may include applying the pulsed electromagnetic field in proximity to a target region affected by a neurological injury or condition to reduce a physiological response comprises reducing a concentration of IL-1β.
  • As mentioned above, these methods may be used to treat any appropriate injury or condition, including a neurodegenerative disease, e.g., Parkinson's disease. Alzheimer's disease, etc. The injury or condition may be a traumatic brain injury (TBI). The injury or condition may be a post-operative inflammation and pain.
  • Also described herein are apparatuses configured to perform any of these methods. For example, an apparatus for applying pulsed electromagnetic field (PEMF) energy to a subject may include: a generator unit including a signal generator configured to generate a PEMF waveform having configured to simultaneously increase the rate of ion-dependent signaling and to minimize the rate of inhibition of such signaling; a programmable control unit configured to repeatedly provide a signal to the generator unit corresponding to the PEMF waveform for a treatment interval that is 10 minutes or greater followed immediately by an off time having an inter-treatment period that is greater than six times the treatment interval; and an applicator unit configured to be worn by the subject, wherein the generator unit is configured to power the applicator unit to drive transmission of a PEMF signal from the applicator unit based on the PEMF waveform.
  • The programmable control unit may be programmed to provide an inter-treatment period that is between six and 100 times the treatment interval. The programmable control unit may be configured to prevent the generation of a pulsed electromagnetic field during the inter-treatment period.
  • Any of these apparatuses may include a shut off to stop the generator unit during the inter-treatment period.
  • Any appropriate applicator may be used, particularly flexible loop applicators, which may be bent or shaped, though remain a coil. In some variations, the applicator comprises a first loop configured to provide the PEMF waveform and a second loop configured to provide a second PEMF waveform.
  • A programmable control unit may be configured to repeatedly provide the same signal to the generator unit corresponding to the PEMF waveform.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The novel features of the invention are set forth with particularity in the claims that follow. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
  • FIG. 1A is a schematic representation of the biological EMF transduction pathway which is a representative target pathway of EMF signals configured according to embodiments described herein.
  • FIG. 1B is a flow diagram of a method for treating a patient according to an embodiment of the devices and methods described herein.
  • FIG. 2A is a block diagram of miniaturized circuitry for use with a coil applicator according to some embodiments described.
  • FIG. 2B illustrates a device for application of electromagnetic signals according to an embodiment of the devices and methods described herein.
  • FIG. 2C illustrates a waveform delivered to a target pathway structure of a plant, animal or human, such as a molecule cell, tissue, organ, or partial or entire organism, according to some embodiments described.
  • FIG. 3 is a photograph illustrating a disposable dual coil PEMF device used in some of the embodiments described herein. The disposable dual coil PEMF includes a battery-powered signal generator is at the bottom between the coils. A nonthermal pulse-modulated radio frequency PEMF signal, configured to modulate NO signaling, may be delivered to tissue with a preprogrammed dosing regimen for each cohort.
  • FIG. 4 is a graph showing the effect of different PEMF therapy regimens on the rate of pain reduction following breast reduction surgery in accordance with various embodiments of pain treatments.
  • FIG. 5 is a graph showing the effect of different PEMF therapy regimens in accordance with the embodiments described herein on pain medication usage of patients after surgery. The results show approximately 2-fold more narcotic pills were taken by 24 h postoperatively in the Q4 (sham) and 5/20 (active) cohorts versus the Q4 (active) and Q2 (active) cohorts. There was no significant difference in postoperative narcotic use between the 5/20 (active) and Q4 (sham) cohorts.
  • FIGS. 6A and 6B are graphs showing the effect of signal configuration on NO production from challenged fibroblasts and chondrocytes (respectively) in culture.
  • FIG. 7 is a graph showing the effect of exposure time of a given signal configuration on cGMP production in primary neuronal cells.
  • DETAILED DESCRIPTION
  • Pulsed electromagnetic fields (PEMF) reduce postoperative pain and narcotic requirements in breast augmentation, reduction, and reconstruction patients. PEMF treatment can also be used to reduce post-operative pain in other surgical procedures. PEMF enhances calmodulin-dependent nitric oxide, which enhances cyclic guanosine monophosphate signaling and phosphodiesterase activity, which blocks cyclic guanosine monophosphate. This invention describes means to configure PEMF dosing to minimize the effect of the competing response of phosphodiesterace activity.
  • FIG. 1B is a flow diagram of a method for treating a subject with a PEMF. In some variations, before beginning the treatment, one or more (or a range of) waveforms may be determined that target the appropriate pathway for the target tissue. In such embodiments, once this determination is made, electromagnetic fields are applied to the target location.
  • FIG. 2A illustrates a block diagram of an EMF delivery apparatus as described according to some embodiments. As shown in FIG. 2A, the apparatus may have miniaturized circuitry for use with a coil applicator. In some embodiments, the apparatus may include a CPU MODULATOR, a BATTERY MODULE, a POWER SUPPLY, On/Off switch, and an output amplifier, AMP, as illustrated. In further variations, the CPU MODULATOR may be an 8 bit 4 MHz micro-controller; however, other suitable bit-MHz combination micro-controllers may be used as well. For example, in some embodiments, the CPU MODULATOR may be programmed for a given carrier frequency or pulse duration, such as about 27.12 MHz sinusoidal wave. Moreover, the CPU MODULATOR may be programmed for a given burst duration, for example about 3 msec. In further variations, the CPU MODULATOR may be programmed to provide a given in situ peak electric field, for example 20 V/m; or a given treatment time, for example about 15 minutes; and/or a given treatment regimen, for example about 10 minutes about every hour. The CPU MODULATOR may also be programmed to deliver an EMF waveform to the target ion binding pathway.
  • Some embodiments combine the signal generation and coil or electrode applicator into one portable or disposable unit, such as illustrated in FIG. 2B (which will be described in greater detail below) for the case of an inductively coupled signal. In some variations, when electrical coils are used as the applicator, the electrical coils can be powered with a time varying magnetic field that induces a time varying electric field in a target pathway structure according to Faraday's law. An electromagnetic field generated by a circuit such as shown in FIG. 2A can also be applied using electrochemical coupling, wherein electrodes are in direct contact with skin or another outer electrochemically conductive boundary of a target pathway structure.
  • In yet another embodiment, the electromagnetic field generated by the generating circuit of FIG. 2A (or FIG. 2B) can also be applied using electrostatic coupling wherein an air gap exists between a generating device such as an electrode and a target pathway structure such as a molecule, cell, tissue, and organ of a plant animal or human. Advantageously, the ultra lightweight coils and miniaturized circuitry, according to some embodiments, allow for use with common physical therapy treatment modalities and at any location on a plant, animal or human for which any therapeutic or prophylactic effect is desired. An advantageous result of application of some embodiments described is that a living organism's wellbeing can be maintained and enhanced.
  • Referring to FIG. 2C, an embodiment according to the present invention of an induced electric field waveform delivered to a target pathway structure is illustrated. As shown in FIG. 2C, burst duration and period are represented by T1 and T2, respectively. In some embodiments, the signal within the rectangular box designated at T1 can be, rectangular, sinusoidal, chaotic or random, provided that the waveform duration or carrier period is less than one-half of the target ion bound time. The peak induced electric field is related to the peak induced magnetic field, shown as B in FIG. 2C, via Faraday's Law of Induction.
  • FIG. 2B illustrates an embodiment of an apparatus 200 that may be used. The apparatus is constructed to be self-contained, lightweight, and portable. A circuit control/signal generator 201 may be held within a (optionally wearable) housing and connected to a generating member such as an electrical coil 202. In some embodiments, the circuit control/signal generator 201 is constructed in a manner that given a target pathway within a target tissue, it is possible to choose waveform parameters that satisfy a frequency response of the target pathway within the target tissue. For some embodiments, circuit control/signal generator 201 applies mathematical models or results of such models that approximate the kinetics of ion binding in biochemical pathways. Waveforms configured by the circuit control/signal generator 201 are directed to a generating member 202. In some variations, the generating member 202 comprises electrical coils that are pliable and comfortable. In further embodiments, the generating member 202 is made from one or more turns of electrically conducting wire in a generally circular or oval shape, any other suitable shape. In further variations, the electrical coil is a circular wire applicator with a diameter that allows encircling of a subject's cranium. In some embodiments, the diameter is between approximately 6-8 inches. In general, the size of the coil may be fixed or adjustable and the circuit control/signal generator may be matched to the material and the size of the applicator to provide the desired treatment.
  • The apparatus 200 may deliver a pulsing magnetic field that can be used to provide treatment. In some embodiments, the device 200 may apply a pulsing magnetic field for a prescribed time and can automatically repeat applying the pulsing magnetic field for as many applications as are needed in a given time period, e.g. 6-12 times a day. The device 200 can be configured to apply pulsing magnetic fields for any time repetition sequence. Without being bound to any theory, it is believed that when electrical coils are used as a generating member 202, the electrical coils can be powered with a time varying magnetic field that induces a biologically and therapeutically effective time varying electric field in a target tissue location.
  • In other embodiments, an electromagnetic field generated by the generating member 202 can be applied using electrochemical coupling, wherein electrodes are in direct contact with skin or another outer electrically conductive boundary of the target tissue (e.g. skull or scalp). In other variations, the electromagnetic field generated by the generating member 202 can also be applied using electrostatic coupling wherein an air gap exists between a generating member 202 such as an electrode and the target tissue. In further examples, a signal generator and battery is housed in the miniature circuit control/signal generator 201 and the miniature circuit control/signal generator 201 may contain an on/off switch and light indicator. In further embodiments, the activation and control of the treatment device may be done via remote control such as by way of a fob that may be programmed to interact with a specific individual device. In other variations, the treatment device further includes a history feature that records the treatment parameters carried out by the device such that the information is recorded in the device itself and/or can be transmitted to another device such as computer, smart phone, printer, or other medical equipment/device.
  • In other variations, the treatment device 200 has adjustable dimensions to accommodate fit to a variety of patient sizes and anatomy. For example, the generating member 202 may comprise modular components which can be added or removed by mated attaching members. Alternatively, the treatment device 200 may contain a detachable generating member (e.g. detachable circular coil or other configurations) that can be removed and replaced with configurations that are better suited for the particular patient's needs. A circular coil generating member 202 may be removed and replaced with an elongate generating member such that PEMF treatment can be applied where other medical equipment may obstruct access by a circular generating member 202. In other variations, the generating member may be made from Litz wire that allows the generating member to flex and fold to accommodate different target areas or sizes.
  • The PEMF devices disclosed herein can be used to treat patients with post-operative pain. Acute postoperative pain is a significant medical problem. Postoperative pain must be managed effectively to optimize surgical outcomes, reduce morbidity, shorten the duration of hospital stay, and control ever-increasing health-care costs [1]. For the vast majority of surgical procedures, pain mechanisms involve increased sensitivity of nociceptors due to increased presence of proinflammatory cytokines in the wound milieu [2]. Narcotics are most commonly used to treat postoperative pain; however, narcotics do not reduce nociceptor sensitivity and cause undesirable side effects and potential addiction. Alternative approaches to decrease post-operative pain involve slowing the appearance of proinflammatory agents at the surgical site [2].
  • To this end, a new modality, nonthermal, nonpharmacologic radio frequency pulsed electromagnetic field (PEMF) therapy has been reported to instantaneously enhance calmodulin (CaM)-dependent nitric oxide (NO) release in challenged cells and tissues. This, in turn, enhances the body's primary anti-inflammatory pathway, CaM-dependent nitric oxide/cyclic guanosine monophosphate (NO/cGMP) signaling [3e7]. In the surgical context, NO/cGMP signaling decreases the rate of release of proinflammatory cytokines, such as interleukin-1 beta (IL [interleukin]-1b) [8], and increases the release of growth factors, such as fibroblast growth factor-2 (FGF-2) [9], in the wound milieu. This mechanism is schematically represented in FIG. 1A. PEMF modulation of angiogenesis via effects on FGF-2 has been reported [10-15]. In some studies, the PEMF effect could be blocked with an FGF-2 inhibitor, consistent with a PEMF effect on NO/cGMP signaling [12, 13].
  • In the clinical setting, PEMF has been reported to accelerate postoperative pain decrease, with a concomitant reduction in narcotic requirements, in double-blinded, randomized clinical studies on breast reduction (BR) [16], breast augmentation [17, 18], and autologous flap breast reconstruction [19]. The BR study also showed that PEMF reduced inflammation by reducing IL-1 beta more than two-fold in the wound exudate, which correlated with the higher rate of pain reduction from PEMF [16]. PEMF can and has been used throughout the body, including after abdominoplasties, major intra-abdominal surgery, extremity procedures, and facial fat grafting [20, 21].
  • Taken together, preclinical and clinical results support an anti-inflammatory mechanism for PEMF based on modulation of CaM-dependent NO/cGMP signaling. However, the NO/cGMP cascade is dynamic [22] and regulated, in part, by phosphodiesterase (PDE) inhibition of cyclic guanosine monophosphate (cGMP) [23]. This inhibition is particularly important for PEMF therapy because PDE isoenzymes are also CaM-dependent, meaning the timing of PDE activity is modulated by the same PEMF signal that modulates the timing of NO/cGMP signaling [24]. Thus, although the dynamics of NO/cGMP signaling in challenged tissue can be modulated by PEMF, the effect of PEMF dosing on the competing dynamics of CaM-dependent NO/cGMP signaling and PDE inhibition of cGMP on pain outcome must be taken into account.
  • For example, a number of patients with post-operative pain were treated with PEMF and studied. Specifically, two prospective, nonrandomized, active cohorts of breast reduction patients, with 15 min PEMF per 2 h; “Q2 (active)”, and 5 min PEMF per 20 min; “5/20 (active)”, dosing regimens were added to a double-blind clinical study wherein 20 min PEMF per 4 h, “Q4 (active)”, dosing was shown to significantly accelerate postoperative pain reduction compared with Q4 shams. Postoperative visual analog scale pain scores and narcotic use were compared.
  • Data from 50 patients were available for analysis. The change in VAS scores normalized to 1 h for each cohort is summarized in FIG. 4. The rate of postoperative pain decrease in the first 24 h postoperative for patients in the Q4 (active) and Q2 (active) cohorts was not significantly different (P=0.485), but was nearly 3-fold faster than that for patients in the 5/20 (active) and Q4 (sham) cohorts (P<0.01). In contrast, the rate of pain decrease for patients treated with the 5/20 (active) regimen was not significantly different from those receiving no PEMF treatment in the Q4 (sham) cohort (P=0.271). Specifically, pain at 24 h postoperative was, respectively, 43% and 35% of pain at 1 h postoperative for patients in the Q4 (active) and Q2 (active) cohorts (P<0.01). In contrast, pain at 24 h for patients in the 5/20 (active) cohort was 87% of pain at 1 h, compared with 74% for patients in the Q4 (sham) cohort (P=0.451). These results can be seen in FIG. 4. A similar pattern of results was found in narcotic usage. Postoperative narcotic usage by 24 h postoperative for patients in the 5/20 (active) cohort was not significantly different from that in the Q4 (sham) cohort (P=0.478), and both were approximately 2-fold higher compared with narcotic usage for patients in the Q4 (active) and Q2 (active) cohorts (P<0.02). Narcotic usage for patients in the Q2 (active) and Q4 (active) cohorts was not significantly different (P=0.246). These results can be seen in FIG. 5.
  • The identical PEMF signal configuration was used for all active cohorts; however, the dosing regimen was different. Entry criteria were identical for all patients. Surgery was performed by the same surgeon on all patients. The results clearly suggest that the effectiveness of PEMF therapy on the rate of postoperative pain decrease and post-operative narcotic requirements in BR patients depends on PEMF dosing regimen. A 5/20 (active) regimen was no different than the Q4 (sham) regimen for pain reduction, whereas a Q2 (active) regimen was as effective as the Q4 (active) regimen.
  • The findings revealed that the regimen of PEMF can significantly impact its effect on postoperative pain. It was expected that the most frequent dosing at 5 min every 20 min would have the greatest effect on pain reduction, but this was not the case. Mean VAS pain scores for patients in the 5/20 (active) cohort were not significantly different from those for patients in the Q4 (sham) cohort, which were more than two-fold higher at 24 h postoperatively than VAS scores for patients in the Q4 (active) and Q2 (active) cohorts. Similar comparative results were obtained for postoperative narcotic usage for patients in each of the active cohorts.
  • PEMF signal parameters, including repetition rate, were identical for all patients in active cohorts. The dosing change was treatment regimen. The rate of increase in CaM-dependent NO, and therefore cGMP, from PEMF in tissue for the 5/20 regimen is nearly 2.5-fold higher than that for the Q2 (active) and 4-fold higher than that for the Q4 (active) regimens. The NO/cGMP signaling pathway is a principal anti-inflammatory pathway. CaM-dependent PDE activity regulates this pathway by inhibiting cGMP. The PEMF signal used in this study is known to enhance NO/cGMP signaling, and to enhance CaM-dependent PDE activity [5-7]. It was proposed that the 5/20 regimen caused PDE activity to predominate, thereby inhibiting all the enhanced cGMP produced by PEMF. The result is no effect of PEMF on postoperative pain.
  • Two recent publications illustrate that PEMF effects depend on signal configuration. The first showed that the PEMF effect on breast cancer cell apoptosis was significant when the same waveform, applied for the same exposure time, repeated at 20 Hz but not at 50 Hz [35]. The second study showed that PEMF significantly reduced the expression of inflammatory markers, tumor necrosis factor, and nuclear factor-kappa beta in challenged macrophages when the same waveform, applied for the same exposure time, was repeated at 5 Hz, but not at 15 or 30 Hz [36]. In both studies, CaM-dependent NO/cGMP signaling modulates the expressions of these inflammatory markers [37,38], suggesting the effect of increased repetition rate is consistent with increased production of NO at a rate high enough for PDE inhibition of cGMP isoforms to predominate, thus blocking the PEMF effect. It is interesting to note that similar dosing effects have been observed in studies using low-level laser therapy, wherein the mechanism of action also involves NO/cGMP signaling [39]. Comparable with our study, low-level laser therapy improvement of neurologic performance in a mouse traumatic brain injury model depended on treatment regimen [40].
  • This study provides evidence that nonthermal radio frequency PEMF therapy can accelerate pain reduction and decrease pain medication requirements in the immediate postoperative period. The effect of PEMF regimen has been elucidated and effective regimens defined. Every 2 or 4 h dosing significantly decreases postoperative pain, whereas every 20-min dosing has no effect compared with placebo. The results of this study confirm dosing by which a given PEMF signal, configured to enhance the body's primary anti-inflammatory signaling pathway, CaM-dependent NO/cGMP, can accelerate postoperative pain relief.
  • At the cellular level the effect of PEMF signal configuration on PDE inhibition of cGMP was examined in cell cultures. Cells were plated in DMEM containing low concentration (challenge) of fetal calf-serum in 24-well plates. Two cell types were tested; human fibroblasts (HFC) and human chondrocytes (HCC). Cells were grown for 24-hours to allow for attachment and repair after initial plating. Cells were exposed to PEMF signals for 15 minutes. Immediately after treatment conditioned media (CM) was collected and assayed for NO levels using the Griess reaction combined with vanadium. The Griess assay tests for nitrite (NO2-). Vanadium was used to reduce nitrate (NO3-) to NO2- since NO immediately reacts with water to form NO3- and NO2- and the Griess assay only measures NO2-.
  • Two PEMF signals with 27.12 MHz sinusoidal carrier were tested. One PEMF signal had a burst width of 2 msec repeating at 2 Hz (Signal I) while the second signal (Signal II) had a burst width of 3 msec repeating at 5 Hz. Both signals were tested at amplitudes ranging from 0.5 to 8 μT. Exposure time was 15 min for each amplitude condition. Since each waveform was configured to target Ca/CaM binding, Signal II would be expected to produce approximately 4-fold more NO than that produced by Signal I, at all amplitudes studied.
  • The results are shown in FIGS. 6A and 6B, wherein it may be seen that Signal I produced significant increases in NO over a range of amplitudes. In contrast, Signal II did not produce increased NO at any amplitude tested. As for the clinical example CaM-dependent PDE activity regulates NO/cGMP signaling by inhibiting cGMP. It was proposed that Signal II with increased burst duration and repetition rate compared to Signal I increased NO too rapidly causing the PEMF effect on PDE activity to predominate, thereby inhibiting all the enhanced NO produced by PEMF. The result is no effect of PEMF on NO release in challenged fibroblasts and chondrocytes for Signal II at any amplitude tested.
  • In another cellular study the effect of exposure time of a PEMF signal consisting of a 2 msec burst of a 27.12 MHz carrier repeating at 2 Hz and delivering 4 μT amplitude was tested. Primary neuronal cells were subjected to oxygen glucose deprivation (OGD) which subjects the cells to ischemic conditions such as those which exist in cardiac and brain ischemia. OGD is expected to reduce NO and therefore cGMP. The parameters of the PEMF signal were chosen to modulate CaM/NO/cGMP signaling so that exposure to PEMF during OGD would be expected to produce increased cGMP. However, as exposure time increases, the PEMF effect on PDE activity also increases. Referring to the results given in the previous clinical and cellular examples, an exposure time of 60 minutes would be expected to produce about 4-fold more NO than the standard effective 15 minute exposure. The results are shown in FIG. 7 wherein it may be seen that 15 minute PEMF exposure maximally restored cGMP production. In contrast, an exposure time of 60 minutes was not effective.
  • Therefore, in some embodiments any of the PEMF parameters can be selected to minimize the PDE inhibition of cGMP and/or maximize the production of cGMP. In some embodiments the length of the inter-treatment period and the length of the treatment interval are selected to minimize the PDE inhibition of cGMP. Any of the PEMF waveform parameters can be optimized in conjunction with the length of the treatment interval and inter-treatment period to achieve a desired change to the production of PDE and/or to decrease the inhibition of cGMP by PDE.
  • In the example shown in FIG. 1B, once treatment begins 103, the device, in some variations, applies an envelope of high-frequency waveforms at low amplitude (e.g. less than 50 milliGauss, less than 100 milliGauss, less than 200 milliGauss, etc.) 105. This envelope of high-frequency pulses is then repeated at a particular frequency (repetition rate) after an appropriate delay. The repetition rate may be varied to minimize PDE inhibition of PDE. The amplitude may be varied to minimize PDE inhibition of PDE. The burst duration may be varied to minimize PDE inhibition of PDE.
  • The initial signal configuration (burst duration, burst repetition and amplitude) can be repeated for a first treatment time and then followed by a delay during which the treatment is “off” 107. This waiting interval (inter-treatment interval) may last for minutes or hours and then the treatment interval may be repeated again until the treatment regime is complete 109.
  • In some embodiments the length of the inter-treatment period can be selected to minimize the PDE inhibition of cGMP. In some embodiments the inter-treatment period is greater than about 15 minutes. In some embodiments the inter-treatment period is greater than about 30 minutes. In some embodiments the inter-treatment period is greater than about 60 minutes. In some embodiments the inter-treatment period is greater than about 90 minutes. In some embodiments the inter-treatment period is greater than about 120 minutes. In some embodiments the inter-treatment period is greater than about 180 minutes. In some embodiments the inter-treatment period is greater than about 240 minutes
  • In some embodiments the inter-treatment period can be expressed as a multiple of the PEMF treatment interval. In some embodiments the inter-treatment period is at least three times longer than the treatment interval. In some embodiments the inter-treatment period is at least four times longer than the treatment interval. In some embodiments the inter-treatment period is at least five times longer than the treatment interval. In some embodiments the inter-treatment period is at least six times longer than the treatment interval. In some embodiments the inter-treatment period is at least seven times longer than the treatment interval. In some embodiments the inter-treatment period is at least eight times longer than the treatment interval. In some embodiments the inter-treatment period is at least ten times longer than the treatment interval. In some embodiments the inter-treatment period is at least fifteen times longer than the treatment interval. In some embodiments the inter-treatment period is at least twenty times longer than the treatment interval.
  • Any of the PEMF treatment intervals disclosed herein can be used with any of the inter-treatment intervals disclosed herein. In some embodiments the treatment interval is about 5 minutes or longer. In some embodiments the treatment interval is about 10 minutes or longer. In some embodiments the treatment interval is about 15 minutes or longer. In some embodiments the treatment interval is about 20 minutes or longer.
  • In some embodiments the PEMF treatment period is five minutes with a 15 minute inter-treatment period. In some embodiments the PEMF treatment period is 15 minutes with a 105 minute inter-treatment period (e.g. 15 minutes of PEMF treatment per two hours). In some embodiments the PEMF treatment period is 20 minutes with a 160 minute inter-treatment period (e.g. 20 minutes of PEMF treatment per three hours).
  • In some variations, the treatment device is pre-programmed (or configured to receive pre-programming) to execute the entire treatment regime (including multiple on-periods and/or intra-treatment intervals) punctuated by predetermined off-periods (inter-treatment intervals) when no treatment is applied. In further variations, the device is pre-programmed to emit a PEMF signal at 27.12 MHz at 2 msec bursts repeating at 2 bursts/sec. In other embodiments, the device is pre-programmed to emit a PEMF signal at 27.12 MHz (at about amplitude 250-400 mV/cm) pulsed in 4 msec bursts at 2 Hz.
  • In further variations, the method may include a pulsed electromagnetic field comprising a 2 msec burst of 27.12 MHz sinusoidal waves repeating at 2 Hz. In other variations, the method may include a pulsed electromagnetic field comprising a 3 msec burst of 27.12 MHz sinusoidal waves repeating at 2 Hz. In further embodiments, the pulsed electromagnetic field may comprise a 4 msec burst of 27.12 MHz sinusoidal waves repeating at 2 Hz.
  • The patient can be monitored during the PEMF treatment regime to determine the physiological response to the PEMF treatment regime. The treatment cycle (e.g. treatment period and inter-treatment period) can be repeated until a desired physiological response is achieved. Depending on the patient's response to the treatment, the subsequent treatment cycle parameters can be adjusted by a health professional to achieve a desired physiological response in the patient.
  • LIST OF REFERENCES CITED HEREIN
    • [1] Kehlet H, Wilmore D. Evidence-based surgical care and the evolution of fast-track surgery. Ann Surg 2008; 248:189.
    • [2] Binshtok A M, Wang H, Zimmermann K, et al. Nociceptors are interleukin-1beta sensors. J Neurosci 2008; 28:14062.
    • [3] Pilla A A. Mechanisms and therapeutic applications of time varying and staticmagnetic fields. In: Barnes F, GreenebaumB, editors. Biological and Medical Aspects of Electromagnetic Fields. Boca Raton Fla.: CRC Press; 2007. p. 351.
    • [4] Pilla A A, Muehsam D J, Markov M S, Sisken B F. EMF signals and ion/ligand binding kinetics: prediction of bioeffective waveform parameters. Bioelectrochem Bioenerg 1999; 48:27.
    • [5] Pilla A, Fitzsimmons R, Muehsam D, Wu J, Rohde C, Casper D. Electromagnetic fields as first messenger in biological signaling: application to calmodulin-dependent signaling in tissue repair. Biochim Biophys Acta 2011; 1810:1236.
    • [6] Pilla A A. Electromagnetic fields instantaneously modulate nitric oxide signaling in challenged biological systems. Biochem Biophys Res Commun 2012; 426:330.
    • [7] Pilla A A. Nonthermal electromagnetic fields: from first messenger to therapeutic applications. Electromagn Biol Med 2013; 32:123.
    • [8] Ren K, Torres R. Role of interleukin-beta during pain and inflammation. Brain Res Rev 2009; 60:57.
    • [9] Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev 2003; 83:835.
    • [10] Yen-Patton G P, Patton W F, Beer D M, et al. Endothelial cell response to pulsed electromagnetic fields: stimulation of growth rate and angiogenesis in vitro. J Cell Physiol 1988; 134:37.
    • [11] Tepper O M, Callaghan M J, Chang E I, et al. Electromagnetic fields increase in vitro and in vivo angiogenesis through endothelial release of FGF-2. FASEB J 2004; 18:1231.
    • [12] Callaghan M J, Chang E I, Seiser N, et al. Pulsed electromagnetic fields accelerate normal and diabetic wound healing by increasing endogenous FGF-2 release. Plast Reconstr Surg 2008; 121:130.
    • [13] Roland D, Ferder M S, Kothuru R, Faierman T, Strauch B. Effects of pulsed magnetic energy on a microsurgically transferred vessel. Plast Reconstr Surg 2000; 105:1371.
    • [14] Weber R V, Navarro A, Wu J K, Yu H L, Strauch B. Pulsed magnetic fields applied to a transferred arterial loop support the rat groin composite flap. Plast Reconstr Surg 2004; 114:1185.
    • [15] Delle Monache S, Alessandro R, Iorio R, Gualtieri G, Colonna R. Extremely low frequency electromagnetic fields (ELF-EMFs) induce in vitro angiogenesis process in human endothelial cells. Bioelectromagnetics 2008; 29:640.
    • [16] Rohde C, Chiang A, Adipoju O, Casper D, Pilla A A. Effects of pulsed electromagnetic fields on interleukin-1 beta and postoperative pain: a double-blind, placebo-controlled, pilot study in breast reduction patients. Plast Reconstr Surg 2010; 125:1620.
    • [17] Hede{acute over ( )}n P, Pilla A A. Effects of pulsed electromagnetic fields on postoperative pain: a double-blind randomized pilot study in breast augmentation patients. Aesthet Plast Surg 2008; 32:660.
    • [18] Rawe I M, Lowenstein A, Barcelo C R, Genecov D G. Control of postoperative pain with a wearable continuously operating pulsed radiofrequency energy device: a preliminary study. Aesthet Plast Surg 2012; 36:458.
    • [19] Rohde C, Hardy K, Asherman J, Taylor E, Pilla A A. PEMF therapy rapidly reduces post-operative pain in TRAM flap patients. Plast Reconstr Surg 2012; 130:91.
    • [20] Strauch B, Herman C, Dabb R, Ignarro L J, Pilla A A. Evidencebased use of pulsed electromagnetic field therapy in clinical plastic surgery. Aesthet Surg J 2009; 29:135.
    • [21] Guo L, Kubat N J, Nelson T R, Isenberg R A. Meta-analysis of clinical efficacy of pulsed radio frequency energy treatment. Ann Surg 2012; 255:457.
    • [22] Batchelor A M, Bartus K, Reynell C, et al. Exquisite sensitivity to subsecond, picomolar nitric oxide transients conferred on cells by guanylyl cyclase-coupled receptors. Proc Natl Acad Sci USA 2010; 107:22060.
    • [23] Mo E, Amin H, Bianco I H, Garthwaite J. Kinetics of a cellular nitric oxide/cGMP/phosphodiesterase-5 pathway. J Biol Chem 2004; 279:26149.
    • [24] Miller C L, Oikawa M, Cai Y, et al. Role of Ca2b/calmodulinstimulated cyclic nucleotide phosphodiesterase 1 in mediating cardiomyocyte hypertrophy. Circ Res 2009; 105: 956.
    • [25] Li Wan Po A, Petersen B. How high should total pain-relief score be to obviate the need for analgesic remedication in acute pain? Estimation using signal detection theory and individual-patient meta-analysis. J Clin Pharm Ther 2006; 31:161.
    • [26] Wise R J. A preliminary report on a method of planning the mammaplasty. Plast Reconstr Surg 1956; 17:367.
    • [27] Karp N S. Medial pedicle/vertical breast reduction made easy: the importance of complete inferior glandular resection. Ann Plast Surg 2004; 52:458.
    • [28] Davison S P, Mesbahi A N, Ducic I, Sarcia M, Dayan J, Spear S L. The versatility of the superomedial pedicle with various skin reduction patterns. Plast Reconstr Surg 2007; 120:1466.
    • [29] Rasouli J, Lekhraj R, White N M, et al. Attenuation of interleukin-1beta by pulsed electromagnetic fields after traumatic brain injury. Neurosci Lett 2012; 519:4.
    • [30] McLeod B R, Pilla A A, Sampsel M W. Electromagnetic fields induced by Helmholtz aiding coils inside saline-filled boundaries. Bioelectromagnetics 1983; 4:357.
    • [31] Panagopoulos D J, Johansson O, Carlo G L. Evaluation of specific absorption rate as a dosimetric quantity for electromagnetic fields bioeffects. PLoS One 2013; 8: e62663.
    • [32] Coll A M, Ameen J R, Mead D. Postoperative pain assessment tools in day surgery: literature review. J Adv Nurs 2004; 46:124.
    • [33] Bodian C A, Freedman G, Hossain S, Eisenkraft J B, Beilin Y. The visual analog scale for pain: clinical significance in postoperative patients. Anesthesiology 2001; 95:1356.
    • [34] Lacy C F, Armstrong L L, Goldman M P, et al. Drug Information Handbook. 15th ed. Hudson, O H: Lexicomp; 2007.
    • [35] Crocetti S, Beyer C, Schade G, Egli M, Fro{umlaut over ( )}hlich J, Franco-Obrego{acute over ( )}n A. Low intensity and frequency pulsed electromagnetic fields selectively impair breast cancer cell viability. PLoS One 2013; 8:e72944.
    • [36] Ross C L, Harrison B S. Effect of pulsed electromagnetic field on inflammatory pathway markers in RAW 264.7 murine macrophages. J Inflamm Res 2013; 6:45.
    • [37] Yurdagul A Jr, Chen J, Funk S D, Albert P, Kevil C G, Orr A W. Altered nitric oxide production mediates matrix-specific PAK2 and NF-kB activation by flow. Mol Biol Cell 2013; 24:398.
    • [38] Ha K S, Kim K M, Kwon Y G, et al. Nitric oxide prevents 6-hydroxydopamine-induced apoptosis in PC 12 cells through cGMP-dependent PI3 kinase/Akt activation. FASEB J 2003; 17:1036.
    • [39] Chung H, Dai T, Sharma S K, Huang Y Y, Carroll J D, Hamblin M R. The nuts and bolts of low-level laser (light) therapy. Ann Biomed Eng 2012; 40:516.
    • [40] Xuan W, Vatansever F, Huang L, et al. Transcranial low-level laser therapy improves neurological performance in traumatic brain injury in mice: effect of treatment repetition regimen. PLoS One 2013; 8:e53454.
    • [41] Liu S S, Richman J M, Thirlby R C, Wu C L. Efficacy of continuous wound catheters delivering local anesthetic for postoperative analgesia: a quantitative and qualitative systematic review of randomized controlled trials. J Am Coil Surg 2006; 203:914.
    • [42] Taylor, Hardy, Alonso, Pilla, and Rhode, “Pulsed Electromagnetic Fields Dosing Impacts Postoperative Pain in Breast Reduction Patients”, Journal of Surgical Research (2014) Oct. 9, 2014.
  • When a feature or element is herein referred to as being “on” another feature or element, it can be directly on the other feature or element or intervening features and/or elements may also be present. In contrast, when a feature or element is referred to as being “directly on” another feature or element, there are no intervening features or elements present. It will also be understood that, when a feature or element is referred to as being “connected”, “attached” or “coupled” to another feature or element, it can be directly connected, attached or coupled to the other feature or element or intervening features or elements may be present. In contrast, when a feature or element is referred to as being “directly connected”, “directly attached” or “directly coupled” to another feature or element, there are no intervening features or elements present. Although described or shown with respect to one embodiment, the features and elements so described or shown can apply to other embodiments. It will also be appreciated by those of skill in the art that references to a structure or feature that is disposed “adjacent” another feature may have portions that overlap or underlie the adjacent feature.
  • Terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. For example, as used herein, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “comprises” and/or “comprising,” when used in this specification, specify the presence of stated features, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, steps, operations, elements, components, and/or groups thereof. As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items and may be abbreviated as “/”.
  • Spatially relative terms, such as “under”, “below”, “lower”, “over”, “upper” and the like, may be used herein for ease of description to describe one element or feature's relationship to another element(s) or feature(s) as illustrated in the figures. It will be understood that the spatially relative terms are intended to encompass different orientations of the device in use or operation in addition to the orientation depicted in the figures. For example, if a device in the figures is inverted, elements described as “under” or “beneath” other elements or features would then be oriented “over” the other elements or features. Thus, the exemplary term “under” can encompass both an orientation of over and under. The device may be otherwise oriented (rotated 90 degrees or at other orientations) and the spatially relative descriptors used herein interpreted accordingly. Similarly, the terms “upwardly”, “downwardly”, “vertical”, “horizontal” and the like are used herein for the purpose of explanation only unless specifically indicated otherwise.
  • Although the terms “first” and “second” may be used herein to describe various features/elements (including steps), these features/elements should not be limited by these terms, unless the context indicates otherwise. These terms may be used to distinguish one feature/element from another feature/element. Thus, a first feature/element discussed below could be termed a second feature/element, and similarly, a second feature/element discussed below could be termed a first feature/element without departing from the teachings of the present invention.
  • Throughout this specification and the claims which follow, unless the context requires otherwise, the word “comprise”, and variations such as “comprises” and “comprising” means various components can be co-jointly employed in the methods and articles (e.g., compositions and apparatuses including device and methods). For example, the term “comprising” will be understood to imply the inclusion of any stated elements or steps but not the exclusion of any other elements or steps.
  • As used herein in the specification and claims, including as used in the examples and unless otherwise expressly specified, all numbers may be read as if prefaced by the word “about” or “approximately,” even if the term does not expressly appear. The phrase “about” or “approximately” may be used when describing magnitude and/or position to indicate that the value and/or position described is within a reasonable expected range of values and/or positions. For example, a numeric value may have a value that is +/−0.1% of the stated value (or range of values), +/−1% of the stated value (or range of values), +/−2% of the stated value (or range of values), +/−5% of the stated value (or range of values), +/−10% of the stated value (or range of values), etc. Any numerical values given herein should also be understood to include about or approximately that value, unless the context indicates otherwise. For example, if the value “10” is disclosed, then “about 10” is also disclosed. Any numerical range recited herein is intended to include all sub-ranges subsumed therein. It is also understood that when a value is disclosed that “less than or equal to” the value, “greater than or equal to the value” and possible ranges between values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value “X” is disclosed the “less than or equal to X” as well as “greater than or equal to X” (e.g., where X is a numerical value) is also disclosed. It is also understood that the throughout the application, data is provided in a number of different formats, and that this data, represents endpoints and starting points, and ranges for any combination of the data points. For example, if a particular data point “10” and a particular data point “15” are disclosed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between 10 and 15. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.
  • Although various illustrative embodiments are described above, any of a number of changes may be made to various embodiments without departing from the scope of the invention as described by the claims. For example, the order in which various described method steps are performed may often be changed in alternative embodiments, and in other alternative embodiments one or more method steps may be skipped altogether. Optional features of various device and system embodiments may be included in some embodiments and not in others. Therefore, the foregoing description is provided primarily for exemplary purposes and should not be interpreted to limit the scope of the invention as it is set forth in the claims.
  • The examples and illustrations included herein show, by way of illustration and not of limitation, specific embodiments in which the subject matter may be practiced. As mentioned, other embodiments may be utilized and derived there from, such that structural and logical substitutions and changes may be made without departing from the scope of this disclosure. Such embodiments of the inventive subject matter may be referred to herein individually or collectively by the term “invention” merely for convenience and without intending to voluntarily limit the scope of this application to any single invention or inventive concept, if more than one is, in fact, disclosed. Thus, although specific embodiments have been illustrated and described herein, any arrangement calculated to achieve the same purpose may be substituted for the specific embodiments shown. This disclosure is intended to cover any and all adaptations or variations of various embodiments. Combinations of the above embodiments, and other embodiments not specifically described herein, will be apparent to those of skill in the art upon reviewing the above description.

Claims (24)

1. A method for treating a patient, the method comprising:
generating a pulsed electromagnetic field from a pulsed electromagnetic field source;
applying the pulsed electromagnetic field in proximity to a target region affected by an injury or condition to reduce a physiological response to the injury or condition for a treatment interval that is greater than 10 minutes;
discontinuing the application of the pulsed electromagnetic field for an inter-treatment period that is greater than six times the treatment interval; and
repeating, for a plurality of times, the steps of generating, applying and discontinuing.
2. The method of claim 1 further comprising increasing a rate of ion-dependent signaling and minimizing a rate of inhibition of such signaling by natural compounds.
3. The method of claim 1, further comprising increasing a rate of cyclic guanosine monophosphate (cGMP) signaling and minimizing a rate of inhibition of said cGMP signaling by compounds including phosphodiesterase (PDE).
4. (canceled)
5. The method of claim 1, wherein said repeating comprises repeating for at least 10 times.
6. The method of claim 1, wherein said repeating comprises repeating the steps of generating, applying and discontinuing, further wherein the pulsed electromagnetic field generated with each repetition is the same.
7. (canceled)
8. (canceled)
9. The method of claim 1, wherein the treatment interval is between 10 and 30 minutes.
10. The method of claim 1, wherein the inter-treatment period is greater than seven times the treatment interval.
11. The method of claim 1, wherein the inter-treatment period is between six and 100 times the treatment interval.
12. The method of claim 1, wherein the inter-treatment period is greater than or equal to about 90 minutes.
13. The method of claim 1, wherein the inter-treatment period is greater than or equal to about 180 minutes.
14. The method of claim 1, wherein the length of a first treatment interval and the length of the inter-treatment period are selected to minimize phosphodiesterase (PDE) production in the patient.
15. The method of claim 1, further comprising
monitoring the physiological response; and
modifying the pulsed electromagnetic field in response to the monitoring step.
16. The method of claim 1, further comprising monitoring the physiological response; and discontinuing treatment once an acceptable level of the physiological response is reached.
17. The method of claim 1, further comprising modulating inflammatory cytokines and growth factors at the target region by applying the pulsed electromagnetic field to simultaneously increase the rate of said modulation and to minimize the rate of inhibition of said modulation by natural compounds.
18. The method of claim 1, further comprising accelerating healing of the target region by applying the pulsed electromagnetic field to simultaneously increase a rate of healing and to minimize a rate of inhibition of such healing.
19. The method of claim 1, wherein applying the pulsed electromagnetic field in proximity to the target region affected by the neurological injury or condition to reduce the physiological response comprises reducing a concentration of interleukin-1 beta (IL-1β).
20. The method of claim 1, wherein the injury or condition is a neurodegenerative disease.
21. The method of claim 19, wherein the neurodegenerative disease is Parkinson's disease or Alzheimer's disease.
22. The method of claim 1, wherein the injury or condition is traumatic brain injury (TBI).
23. The method of claim 1, wherein the injury or condition is post-operative inflammation and pain.
24-30. (canceled)
US16/431,561 2003-12-05 2019-06-04 Method and Apparatus for Electromagnetic Treatment of Living Systems Abandoned US20190351249A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/431,561 US20190351249A1 (en) 2014-11-04 2019-06-04 Method and Apparatus for Electromagnetic Treatment of Living Systems
US16/657,827 US20200094068A1 (en) 2003-12-05 2019-10-18 Method for treatment of non-alcoholic steatohepatitis using pulsed electromagnetic field therapy

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462075122P 2014-11-04 2014-11-04
US14/932,928 US10350428B2 (en) 2014-11-04 2015-11-04 Method and apparatus for electromagnetic treatment of living systems
US16/431,561 US20190351249A1 (en) 2014-11-04 2019-06-04 Method and Apparatus for Electromagnetic Treatment of Living Systems

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/932,928 Continuation US10350428B2 (en) 2003-12-05 2015-11-04 Method and apparatus for electromagnetic treatment of living systems

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/977,043 Continuation-In-Part US20130218235A9 (en) 2003-12-05 2007-10-22 Excessive fibrous capsule formation and capsular contracture apparatus and method for using same

Publications (1)

Publication Number Publication Date
US20190351249A1 true US20190351249A1 (en) 2019-11-21

Family

ID=55851513

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/932,928 Expired - Fee Related US10350428B2 (en) 2003-12-05 2015-11-04 Method and apparatus for electromagnetic treatment of living systems
US15/920,248 Abandoned US20180200531A1 (en) 2014-11-04 2018-03-13 Method and Apparatus for Electromagnetic Treatment of Living Systems
US16/431,561 Abandoned US20190351249A1 (en) 2003-12-05 2019-06-04 Method and Apparatus for Electromagnetic Treatment of Living Systems

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/932,928 Expired - Fee Related US10350428B2 (en) 2003-12-05 2015-11-04 Method and apparatus for electromagnetic treatment of living systems
US15/920,248 Abandoned US20180200531A1 (en) 2014-11-04 2018-03-13 Method and Apparatus for Electromagnetic Treatment of Living Systems

Country Status (1)

Country Link
US (3) US10350428B2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11020603B2 (en) 2019-05-06 2021-06-01 Kamran Ansari Systems and methods of modulating electrical impulses in an animal brain using arrays of planar coils configured to generate pulsed electromagnetic fields and integrated into clothing
US11517760B2 (en) 2019-05-06 2022-12-06 Kamran Ansari Systems and methods of treating medical conditions using arrays of planar coils configured to generate pulsed electromagnetic fields and integrated into clothing

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9656096B2 (en) 2003-12-05 2017-05-23 Rio Grande Neurosciences, Inc. Method and apparatus for electromagnetic enhancement of biochemical signaling pathways for therapeutics and prophylaxis in plants, animals and humans
US10806942B2 (en) 2016-11-10 2020-10-20 Qoravita LLC System and method for applying a low frequency magnetic field to biological tissues
EP3621686A4 (en) 2017-05-08 2021-01-27 AAH Holdings, LLC Multi-coil electromagnetic apparatus
EP3873593B1 (en) 2018-10-18 2024-02-21 Aah Holdings Llc Inductive applicator coil arrangement for therapeutically treating human and animal bodies
AU2019394862B2 (en) 2018-12-03 2022-09-29 Aah Holdings Llc Apparatus and method for treatment of mental and behavioral conditions and disorders with electromagnetic fields

Family Cites Families (232)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA608693A (en) 1960-11-15 Batrow John Electro-physiotherapy apparatus
US1233841A (en) 1914-05-12 1917-07-17 Marconi Wireless Telegraph Co America Means for receiving electrical oscillations.
FR748828A (en) 1932-01-13 1933-07-10 Siemens Ag Electrode arrangement for medical treatment in the capacitor field of electric oscillations
US2130758A (en) 1935-06-01 1938-09-20 E J Rose Mfg Company Of Califo Electrode for diathermy treatment and the like
GB604107A (en) 1940-06-21 1948-06-29 Dansk Ind Syndikat Cie Madsen Thermionic valve circuits for generating modulated pulse trains
US2276996A (en) 1940-11-30 1942-03-17 A J Ginsberg Non-radio-interfering therapeutic apparatus
US2648727A (en) 1949-10-04 1953-08-11 Crosley Broadeasting Corp Push-pull wide band amplifier
DE970276C (en) 1951-11-29 1958-09-04 Hermann Staehle Fa Dipl Ing Electromedical treatment device for combating pain through periodically varying currents
US3181535A (en) 1957-10-04 1965-05-04 Diapulse Mfg Corp Of America Athermapeutic apparatus
US3043310A (en) 1959-04-24 1962-07-10 Diapulse Mfg Corp Of America Treatment head for athermapeutic apparatus
US3270746A (en) 1963-08-26 1966-09-06 Dynapower Systems Corp High-performance electrotherapeutic treatment head
US3329149A (en) 1964-10-28 1967-07-04 Dynapower Systems Corp Of Cali Supporting arm for electrotherapeutic treatment head
US3329148A (en) 1965-09-21 1967-07-04 Dynapower Systems Corp Of Cali Control of electrotherapeutic apparatus
US3978864A (en) 1965-10-11 1976-09-07 L. B. Smith Estates, Inc. Electrotherapeutic treatment head
AT316729B (en) 1971-04-06 1974-07-25 Kraus Werner Device to support the formation of bone substance
US3800802A (en) 1972-01-07 1974-04-02 Int Medical Electronics Ltd Short-wave therapy apparatus
DE2314573C2 (en) 1973-03-23 1986-12-18 Werner Dipl.-Ing. 8000 München Kraus Device for promoting healing processes
LU70345A1 (en) 1974-06-18 1976-05-31
US3952751A (en) 1975-01-08 1976-04-27 W. Denis Kendall High-performance electrotherapeutic apparatus
US4315503A (en) 1976-11-17 1982-02-16 Electro-Biology, Inc. Modification of the growth, repair and maintenance behavior of living tissues and cells by a specific and selective change in electrical environment
US4105017A (en) 1976-11-17 1978-08-08 Electro-Biology, Inc. Modification of the growth repair and maintenance behavior of living tissue and cells by a specific and selective change in electrical environment
US4266532A (en) 1976-11-17 1981-05-12 Electro-Biology, Inc. Modification of the growth, repair and maintenance behavior of living tissues and cells by a specific and selective change in electrical environment
US4197851A (en) 1977-04-14 1980-04-15 Fellus Victor M Apparatus for emitting high-frequency electromagnetic waves
JPS54119792A (en) 1978-03-03 1979-09-17 Iriyou Kougaku Kenkiyuushiyo K Electric stimulation device for removing pain
US4305115A (en) 1979-03-14 1981-12-08 Harry H. Leveen Electrostatic shield
US4340063A (en) 1980-01-02 1982-07-20 Empi, Inc. Stimulation device
US4374482A (en) 1980-12-23 1983-02-22 Norlin Industries, Inc. Vocal effect for musical instrument
US4428366A (en) 1981-05-06 1984-01-31 Alfred B. Kurtz Electromagnetic apparatus and method for the reduction of serum glucose levels
JPS5821387A (en) 1981-07-29 1983-02-08 Olympus Optical Co Ltd Irradiator for laser
NL8105297A (en) 1981-11-23 1983-06-16 Ver Wetens Onderwijs Gereform METHOD FOR GENERATING NERVE OR MUSCLE STIMULATION SIGNALS
US4556051A (en) 1982-11-05 1985-12-03 Empi, Inc. Method and apparatus for healing tissue
US4550714A (en) 1983-03-09 1985-11-05 Electro-Biology, Inc. Electromagnetic coil insert for an orthopedic cast or the like
IT1159024B (en) 1983-06-02 1987-02-25 Ruggero Cadossi METHOD AND DEVICE FOR THE TREATMENT OF FABRICS AND LIVING CELLS THROUGH ELECTROMAGNETIC FIELDS BUTTONS
US4654574A (en) 1983-06-29 1987-03-31 Sheldon Thaler Apparatus for reactively applying electrical energy pulses to a living body
US4829984A (en) 1983-12-15 1989-05-16 Gordon Robert T Method for the improvement of transplantation techniques and for the preservation of tissue
US4627438A (en) 1984-01-09 1986-12-09 Pain Suppression Labs, Inc. Electronic migraine modulator apparatus and methodology
US4548208A (en) 1984-06-27 1985-10-22 Medtronic, Inc. Automatic adjusting induction coil treatment device
DE3427243C1 (en) 1984-07-24 1985-11-28 Mela GmbH Elektromedizin, 8000 München Analgesia circuitry
US4889526A (en) 1984-08-27 1989-12-26 Magtech Laboratories, Inc. Non-invasive method and apparatus for modulating brain signals through an external magnetic or electric field to reduce pain
US4674482A (en) 1984-09-12 1987-06-23 Irt, Inc. Pulse electro-magnetic field therapy device with auto bias circuit
US4616629A (en) 1985-05-24 1986-10-14 Electro-Biology, Inc. Coil construction for electromagnetic treatment of an afflicted body region
US4765310A (en) 1985-11-01 1988-08-23 Dynatens Research Corporation Electrical and magnetic pain treatment device
US4672951A (en) 1985-12-30 1987-06-16 Bio-Electric, Inc. Method and apparatus for treatment of biological tissue
US5000178A (en) 1986-05-23 1991-03-19 Lti Biomedical, Inc. Shielded electromagnetic transducer
US4998532A (en) 1986-05-23 1991-03-12 Lti Biomedical, Inc. Portable electro-therapy system
US5014699A (en) 1986-05-23 1991-05-14 Trustees Of The University Of Pennsylvania Electromagnetic method and apparatus for healing living tissue
US4940453A (en) 1987-01-28 1990-07-10 Cadwell Industries, Inc. Method and apparatus for magnetically stimulating neurons
US5116304A (en) 1987-01-28 1992-05-26 Cadwell Industries, Inc. Magnetic stimulator with skullcap-shaped coil
CN87208158U (en) 1987-05-20 1988-10-19 张雪珊 Dual-functional domestic lamp
JP2810665B2 (en) 1987-06-15 1998-10-15 松下電工株式会社 Blood circulation promotion device
US4850372A (en) 1987-08-25 1989-07-25 The Johns Hopkins University Electromagnetic non-invasive measurement and monitoring system for osteoporosis
EP0308700A3 (en) 1987-09-08 1990-12-19 Gilson Woo Treatment of ailments, afflictions and diseases
US5318561A (en) 1988-03-23 1994-06-07 Life Resonances Inc. Deformable magnetic field aiding coils for use in controlling tissue growth
US5123898A (en) 1988-03-23 1992-06-23 Life Resonances, Inc. Method and apparatus for controlling tissue growth with an applied fluctuating magnetic field
US5224922A (en) 1988-05-19 1993-07-06 Kurtz Warren H Quasistatic biological cell and tissue modifier
US4993413A (en) 1988-09-22 1991-02-19 The Research Foundation Of State University Of New York Method and apparatus for inducing a current and voltage in living tissue
RU1799577C (en) 1989-08-17 1993-03-07 Межотраслевой научно-технический комплекс "Микрохирургия глаза" Method for improving vision function affected by ophthalmic nerve and retina disease
CA2021506A1 (en) 1989-08-17 1991-02-18 Abraham R. Liboff Electromagnetic treatment therapy for stroke victims
US5181902A (en) 1990-09-21 1993-01-26 American Medical Electronics, Inc. Double-transducer system for PEMF Therapy
CN1052053A (en) 1990-10-28 1991-06-12 郑五星 Orthogonal infra-red electromagnetic therapy instrument
ATE125455T1 (en) 1991-02-28 1995-08-15 Medi Line Gmbh RADIATION DEVICE FOR TREATING LIVING TISSUE USING ELECTROMAGNETIC WAVES.
US5314401A (en) 1991-03-29 1994-05-24 Amei Technologies Inc. Conformable PEMF transducer
DE4221739A1 (en) 1991-07-09 1993-01-14 Fischer Ag LF pulse generator and transmission antenna for proton transport in blood - has coil which induces higher energy in electrolyte fluid than thermal energy, and within cell-specific amplitude window
SU1831343A3 (en) 1991-10-17 1993-07-30 Stanislav N Darovskikh Device for stimulation of functional state of biological object
US5370680A (en) 1992-05-27 1994-12-06 Magnetic Resonance Therapeutics, Inc. Athermapeutic apparatus employing electro-magnetic fields
US5478303A (en) 1992-09-18 1995-12-26 Foley-Nolan; Darragh Electromagnetic apparatus for use in therapy
US5338286A (en) 1992-12-08 1994-08-16 Electro-Biology, Inc. Electromagnetic bioresponse by selective spectral suppression in pulsed field stimulation
US5386837A (en) 1993-02-01 1995-02-07 Mmtc, Inc. Method for enhancing delivery of chemotherapy employing high-frequency force fields
US5584863A (en) 1993-06-24 1996-12-17 Electropharmacology, Inc. Pulsed radio frequency electrotherapeutic system
DE4335102A1 (en) 1993-10-14 1995-04-20 Fischer Ag Device for determining the effect of pulsed magnetic fields on an organism
IT1265231B1 (en) 1993-11-26 1996-10-31 Biofarm Srl DEVICE TO SLOW DOWN HAIR LOSS AND STIMULATE REGROWTH
WO1995019808A1 (en) 1994-01-21 1995-07-27 Erik Larsen Device for stimulating body cells by electromagnetic radiation
AU2209195A (en) 1994-04-12 1995-10-30 Australasian Medical Technology (Nz) Limited Orthotic devices incorporating pulsed electromagnetic field therapy
US5407421A (en) 1994-05-18 1995-04-18 Goldsmith; Seth Compressive brace
US6004257A (en) 1994-05-25 1999-12-21 Jacobson; Jerry I. Method for ameliorating the aging process and the effects thereof utilizing electromagnetic energy
WO1995033514A1 (en) 1994-06-09 1995-12-14 Magnetic Resonance Therapeutics, Inc. Electro-therapeutic method
WO1996011723A1 (en) 1994-10-17 1996-04-25 Australasian Medical Technology Limited Devices and methods for implementation of pulsed electromagnetic field therapy
US6086525A (en) 1994-11-28 2000-07-11 Neotonus, Inc. Magnetic nerve stimulator for exciting peripheral nerves
US6213934B1 (en) 1995-06-01 2001-04-10 Hyper3D Corp. Electromagnetic bone-assessment and treatment: apparatus and method
US5707334A (en) 1995-08-21 1998-01-13 Young; Robert B. Method of treating amygdala related transitory disorders
US5718246A (en) 1996-01-03 1998-02-17 Preferential, Inc. Preferential induction of electrically mediated cell death from applied pulses
US5792209A (en) 1996-04-01 1998-08-11 Varner; Lawrence Norman Osteoporosis-relief device
US5778894A (en) 1996-04-18 1998-07-14 Elizabeth Arden Co. Method for reducing human body cellulite by treatment with pulsed electromagnetic energy
US6234953B1 (en) 1996-06-06 2001-05-22 Lawson Research Institute Electrotherapy device using low frequency magnetic pulses
US6246912B1 (en) 1996-06-27 2001-06-12 Sherwood Services Ag Modulated high frequency tissue modification
ES2245465T3 (en) 1996-09-18 2006-01-01 Applied Genetics Incorporated Dermatics NORBORNENE AND NORBORNAN COMPOUND DIOLS FOR THE TREATMENT OF PIGMENTATION DISORDERS, NEURODEGENERATIVE DISEASES OR PROLIFERATIVE SKIN DISEASES.
US5743844A (en) 1996-11-01 1998-04-28 Amei Technologies, Inc. High efficiency pulsed electromagnetic field (PEMF) stimulation therapy method and system
US6132362A (en) 1996-11-01 2000-10-17 Amei Technologies, Inc. Pulsed electromagnetic field (PEMF) stimulation therapy system with bi-phasic coil
US6261221B1 (en) 1996-11-01 2001-07-17 Amei Technologies Inc. Flexible coil pulsed electromagnetic field (PEMF) stimulation therapy system
US5718721A (en) 1996-12-23 1998-02-17 Ross; Jesse Method of relieving migraine headache pain
US6321120B1 (en) 1997-12-29 2001-11-20 Indnjc, Inc. RF therapeutic cancer apparatus and method
US6684108B2 (en) 1996-12-30 2004-01-27 Indnjc, Inc. Therapeutic and diagnostic apparatus and method
US5968527A (en) 1997-02-27 1999-10-19 Catholic University Of America, The Protection of living systems from the adverse effects of stress
US5908444A (en) 1997-06-19 1999-06-01 Healing Machines, Inc. Complex frequency pulsed electromagnetic generator and method of use
US6458157B1 (en) 1997-08-04 2002-10-01 Suaning Gregg Joergen Retinal stimulator
US5951459A (en) 1997-08-29 1999-09-14 Orthosoft, L.L.C. Magnetic coil for pulsed electromagnetic field
US5997464A (en) 1997-08-29 1999-12-07 Orthosoft, L.L.C. Magnetic coil for pulsed electromagnetic field
KR100396055B1 (en) 1997-09-26 2003-08-27 우길선 Treatment of afflictions, ailments and diseases
US6083149A (en) 1997-10-22 2000-07-04 Emf Therapeutics, Inc. Magnetic field device and method for inhibiting angiogenesis and retarding growth rates of tumors in mammals
DE69941557D1 (en) 1998-01-15 2009-12-03 Regenesis Biomedical Inc IMPROVED DEVICE FOR TREATMENT BY PULSED ELECTROMAGNETIC ENERGY
US6348070B1 (en) 1998-04-17 2002-02-19 Med-El Elektromedizinische Gerate Ges.M.B.H Magnetic-interference-free surgical prostheses
NL1009568C2 (en) 1998-07-06 2000-01-10 Raitec B V I O Device for treating patients using electromagnetic radiation.
JP2002522125A (en) 1998-08-03 2002-07-23 エイエムイーアイ テクノロジーズ インコーポレイテッド PEMF device for osteoporosis and stimulation of tissue growth
US7209787B2 (en) 1998-08-05 2007-04-24 Bioneuronics Corporation Apparatus and method for closed-loop intracranial stimulation for optimal control of neurological disease
US6099459A (en) 1998-09-04 2000-08-08 Jacobson; Jerry I. Magnetic field generating device and method of generating and applying a magnetic field for treatment of specified conditions
US6190893B1 (en) 1998-09-18 2001-02-20 Massachusetts Institute Of Technology Electroactive materials for stimulation of biological activity of bone marrow stromal cells
US6569654B2 (en) 1998-09-18 2003-05-27 Massachusetts Institute Of Technology Electroactive materials for stimulation of biological activity of stem cells
US6155966A (en) 1998-11-17 2000-12-05 Parker; Lloyd S. Apparatus and method for toning tissue with a focused, coherent electromagnetic field
US6283956B1 (en) 1998-11-30 2001-09-04 David H. McDaniels Reduction, elimination, or stimulation of hair growth
US6936044B2 (en) 1998-11-30 2005-08-30 Light Bioscience, Llc Method and apparatus for the stimulation of hair growth
US6193763B1 (en) 1998-12-17 2001-02-27 Robert A. Mackin Apparatus and method for contemporaneous treatment and fluoroscopic mapping of body tissue
US6231528B1 (en) 1999-01-15 2001-05-15 Jonathan J. Kaufman Ultrasonic and growth factor bone-therapy: apparatus and method
CA2298515A1 (en) 1999-02-11 2001-08-10 Queen's University At Kingston Method and apparatus for detecting eye movement
US6149577A (en) 1999-03-18 2000-11-21 Emf Therapeutics, Inc. Apparatus and method for creating a substantially contained, finite magnetic field useful for relieving the symptoms pain and discomfort associated with degenerative diseases and disorders in mammals
WO2000057951A1 (en) 1999-03-26 2000-10-05 Flock Stephen T Delivery of pharmaceutical compounds and collection of biomolecules using electromagnetic energy and uses thereof
US6261831B1 (en) 1999-03-26 2001-07-17 The United States Of America As Represented By The Secretary Of The Air Force Ultra-wide band RF-enhanced chemotherapy for cancer treatmeat
US6463336B1 (en) 1999-04-01 2002-10-08 Mmtc, Inc Active bandage suitable for applying pulsed radio-frequencies or microwaves to the skin for medical purposes
US6200259B1 (en) 1999-06-03 2001-03-13 Keith L. March Method of treating cardiovascular disease by angiogenesis
IL146976A0 (en) 1999-06-08 2002-08-14 Medical Bracing Systems Ltd Pemf biophysical stimulation field generator device and method
US7177696B1 (en) 1999-06-09 2007-02-13 H & P Medical Research, Inc. Multiple selectable field/current-voltage pads having individually powered and controlled cells
RU2164424C1 (en) 1999-06-28 2001-03-27 Коноплев Сергей Петрович Method and device for applying low frequency electromagnetic therapy
US6556872B2 (en) 1999-08-24 2003-04-29 Ev Vascular, Inc. Therapeutic device and method for treating diseases of cardiac muscle
US6560489B2 (en) 1999-08-24 2003-05-06 Em Vascular, Inc. Therapeutic device and method for treating diseases of cardiac muscle
US6561968B1 (en) 1999-08-31 2003-05-13 Biofields Aps Method and an apparatus for stimulating/ modulating biochemical processes using pulsed electromagnetic fields
US6458151B1 (en) 1999-09-10 2002-10-01 Frank S. Saltiel Ostial stent positioning device and method
US6678562B1 (en) 2000-01-12 2004-01-13 Amei Technologies Inc. Combined tissue/bone growth stimulator and external fixation device
US6853864B2 (en) 2000-02-02 2005-02-08 Catholic University Of America, The Use of electromagnetic fields in cancer and other therapies
US7022506B2 (en) 2000-02-23 2006-04-04 The Trustees Of The University Of Pennsylvania Method and device for treating osteoarthritis, cartilage disease, defects and injuries in the human knee
US7465546B2 (en) 2000-02-23 2008-12-16 The Trustees Of The University Of Pennsylvania Regulation of transforming growth factor-beta (TGF-β) gene expression in living cells via the application of specific and selective electric and electromagnetic fields
US6919205B2 (en) 2000-02-23 2005-07-19 The Trustees Of The University Of Pennsylvania Regulation of type II collagen gene expression using specific and selective electrical and electromagnetic signals
US7374916B2 (en) 2000-02-23 2008-05-20 The Trustees Of The University Of Pennsylvania Regulation of aggrecan gene expression using specific and selective electrical and electromagnetic signals
US7130692B2 (en) 2000-02-23 2006-10-31 The Trustees Of The University Of Pennsylvania Portable electrotherapy device for treating osteoarthritis and other diseases, defects and injuries of the knee joint
US7465566B2 (en) 2000-02-23 2008-12-16 The Trustees Of The University Of Pennsylvania Regulation of genes via application of specific and selective electrical and electromagnetic signals
US7429471B2 (en) 2000-02-23 2008-09-30 The Trustees Of The University Of Pennsylvania Regulation of matrix metalloproteinase gene expression using specific and selective electrical and electromagnetic signals
JP2001293098A (en) 2000-04-14 2001-10-23 Nippon Koden Corp Coil device and coil driving device
US6434426B1 (en) 2000-04-27 2002-08-13 Medtronic Inc. Method and system for determining a noise floor in a cardiac pacing system
US20020035358A1 (en) 2000-05-09 2002-03-21 Ming Wang Pulsed electromagnetic field therapy for treatment of corneal disorders and injuries
ATE287799T1 (en) 2000-06-21 2005-02-15 Top Q Ag HAND WRITING DEVICE WITH INTEGRATED TOUCH PEN ELEMENT AND LASER POINTER
US6421562B1 (en) 2000-07-17 2002-07-16 Jesse Ross Alternative treatment of a nonsurgically treatable intracranial occlusion
US6591138B1 (en) 2000-08-31 2003-07-08 Neuropace, Inc. Low frequency neurostimulator for the treatment of neurological disorders
US7160241B1 (en) 2000-10-06 2007-01-09 Ewa Herbst Treatment of living tissues using electromagnetic fields
US7089060B1 (en) 2001-02-23 2006-08-08 Amei Technologies Inc. Methods of stimulating cell receptor activity using electromagnetic fields
US6648812B2 (en) 2001-02-28 2003-11-18 Nu-Magnetics, Inc. Bi-axial rotating magnetic therapeutic device
US6589159B2 (en) 2001-04-12 2003-07-08 Sumathi Paturu Magnetic therapy devices and methods
US8047979B2 (en) 2001-04-20 2011-11-01 Mclean Hospital Corporation Magnetic field treatment techniques
DE10126607A1 (en) 2001-05-31 2002-12-05 Richard Markoll Portable applicator for pulsed signal therapy
WO2003014395A1 (en) 2001-08-07 2003-02-20 Genox Research, Inc. Method of examining bronchial asthma
US6839589B2 (en) 2001-09-25 2005-01-04 Jiri Joseph Petlan Method and apparatus for treatment of living matter using pulsed radio frequency electromagnetic radiation
RU2203702C1 (en) 2001-10-29 2003-05-10 Ширяев Вячеслав Михайлович Method and device for applying physiotherapeutic treatment
US20030093028A1 (en) 2001-11-09 2003-05-15 Michael Spiegel Appararus and method for magnetic induction of therapeutic electric fields
US7288062B2 (en) 2001-11-09 2007-10-30 Michael Spiegel Apparatus for creating therapeutic charge transfer in tissue
US7177695B2 (en) 2001-12-17 2007-02-13 Cefamoptec Industries, Inc. Early stage wound healing using electromagnetic radiation
US6844378B1 (en) 2002-01-04 2005-01-18 Sandia Corporation Method of using triaxial magnetic fields for making particle structures
US7010353B2 (en) 2002-01-07 2006-03-07 Ebi, L.P. Non-invasive capacitively coupled electrical stimulation device for treatment of soft tissue wounds
US6701185B2 (en) 2002-02-19 2004-03-02 Daniel Burnett Method and apparatus for electromagnetic stimulation of nerve, muscle, and body tissues
US20030158585A1 (en) * 2002-02-19 2003-08-21 Burnett Daniel R. Method and apparatus for electromagnetic stimulation of nerve, muscle, and body tissues
US6733435B2 (en) 2002-03-08 2004-05-11 Canedo Luis Electromagnetic method of treatment of lesions associated with inadequate blood perfusion, partial denervation, tissue loss, pain, edema, inflammation and infection
US6934580B1 (en) 2002-07-20 2005-08-23 Flint Hills Scientific, L.L.C. Stimulation methodologies and apparatus for control of brain states
CN1408448A (en) 2002-09-16 2003-04-09 周明非 Multifunctional hair growing helmet
US7819794B2 (en) 2002-10-21 2010-10-26 Becker Paul F Method and apparatus for the treatment of physical and mental disorders with low frequency, low flux density magnetic fields
US6955642B1 (en) 2002-11-26 2005-10-18 Ebi, Lp Pulsed electromagnetic field stimulation method and apparatus with improved dosing
US7175587B2 (en) 2002-12-31 2007-02-13 Em-Probe, Inc. Method and apparatus for pulsed electromagnetic therapy
US20050059153A1 (en) 2003-01-22 2005-03-17 George Frank R. Electromagnetic activation of gene expression and cell growth
US7551957B2 (en) 2003-03-06 2009-06-23 Bioelectronics Corp. Electromagnetic therapy device and methods
GB2400316A (en) 2003-04-10 2004-10-13 Richard Markoll Electromagnetic stimulation in patients with osteoporosis
US20050049640A1 (en) 2003-05-12 2005-03-03 Gurtner Geoffrey C. Electromagnetic fields increase in vitro and in vivo angiogenesis through endothelial release of FGF-2
WO2004108208A2 (en) 2003-06-06 2004-12-16 Wayne State University, Technology Transfer Office Methods and apparatus for conversion of eukaryotic cells by application of electric and magnetic fields
EA200600093A1 (en) 2003-06-24 2006-08-25 Хелтоникс, Инк. DEVICE AND METHOD OF BIOELECTRIC STIMULATION, ACCELERATION OF HEALING, FACILITATION OF PAIN OR DEVITALIZATION OF PATHOGENS
US7456189B2 (en) 2003-09-30 2008-11-25 Boehringer Ingelheim International Gmbh Bicyclic heterocycles, medicaments containing these compounds, their use and processes for their preparation
ITTO20030893A1 (en) 2003-11-11 2005-05-12 Igea Srl ELECTROMAGNETIC FIELD STIMULATOR DEVICE FOR ANATOMICAL BIOPHYSICAL PROTECTION.
CN1893999A (en) 2003-11-14 2007-01-10 宾夕法尼亚大学理事会 Method and device for treating osteoarthritis and cartilage disease, defects, and injuries in the human hip
US7744524B2 (en) 2003-12-05 2010-06-29 Ivivi Health Sciences, Llc Apparatus and method for electromagnetic treatment of plant, animal, and human tissue, organs, cells, and molecules
US20050251229A1 (en) 2004-04-19 2005-11-10 Pilla Arthur A Electromagnetic treatment apparatus and method for angiogensis modulation of living tissues and cells
US20130218235A9 (en) 2005-03-07 2013-08-22 Arthur A. Pilla Excessive fibrous capsule formation and capsular contracture apparatus and method for using same
US20070173904A1 (en) 2006-01-25 2007-07-26 Pilla Arthur A Self-contained electromagnetic apparatus for treatment of molecules, cells, tissues, and organs within a cerebrofacial area and method for using same
US9415233B2 (en) 2003-12-05 2016-08-16 Rio Grande Neurosciences, Inc. Apparatus and method for electromagnetic treatment of neurological pain
US7758490B2 (en) 2005-09-10 2010-07-20 Ivivi Health Sciences, Llc Integrated coil apparatus for therapeutically treating human and animal cells, tissues and organs with electromagnetic fields and method for using same
US20110112352A1 (en) 2003-12-05 2011-05-12 Pilla Arthur A Apparatus and method for electromagnetic treatment
US20080132971A1 (en) 2006-09-20 2008-06-05 Pille Arthur A Electromagnetic apparatus for respiratory disease and method for using same
US20060212077A1 (en) 2005-03-07 2006-09-21 Pilla Arthur A Electromagnetic treatment apparatus for augmenting wound repair and method for using same
US20070026514A1 (en) 2005-03-07 2007-02-01 Pilla Arthur A Electromagnetic treatment apparatus for enhancing pharmacological, chemical, and topical agent effectiveness and method for using same
US9656096B2 (en) 2003-12-05 2017-05-23 Rio Grande Neurosciences, Inc. Method and apparatus for electromagnetic enhancement of biochemical signaling pathways for therapeutics and prophylaxis in plants, animals and humans
US20080058793A1 (en) 2006-06-12 2008-03-06 Pilla Arthur A Electromagnetic apparatus for prophylaxis and repair of ophthalmic tissue and method for using same
US9440089B2 (en) 2003-12-05 2016-09-13 Rio Grande Neurosciences, Inc. Apparatus and method for electromagnetic treatment of neurological injury or condition caused by a stroke
US8961385B2 (en) 2003-12-05 2015-02-24 Ivivi Health Sciences, Llc Devices and method for treatment of degenerative joint diseases with electromagnetic fields
US7611453B2 (en) 2003-12-12 2009-11-03 Pilla Arthur A Apparatus and method for static magnetic field treatment of tissue, organs, cells, and molecules
NZ548447A (en) 2004-01-12 2010-04-30 Univ Pennsylvania Up-regulation of bone morphogenetic protein (BMP) gene expression in bone cells by electromagnetic signals
US20050222625A1 (en) 2004-03-30 2005-10-06 Shlomo Laniado Method and apparatus for non-invasive therapy of cardiovascular ailments using weak pulsed electromagnetic radiation
US7333858B2 (en) 2004-03-31 2008-02-19 Cochlear Limited Pulse burst electrical stimulation of nerve or tissue fibers
MX358779B (en) 2004-04-26 2018-09-04 Ivivi Health Sciences Llc Electromagnetic treatment induction apparatus and method.
NL1026431C1 (en) 2004-06-16 2005-12-19 Umc Utrecht Holding Bv Device for generating electric current fields in a human body and method for the use thereof.
US20090099623A1 (en) 2004-09-13 2009-04-16 Neuronix Ltd. Systems and methods for treatment of medical conditions related to the central nervous system and for enhancing cognitive functions
US7520849B1 (en) 2004-09-20 2009-04-21 Ebi, Lp Pulsed electromagnetic field method of treating soft tissue wounds
US20060161226A1 (en) 2005-01-18 2006-07-20 Mcmickle George R Apparatus and method for reducing follicular cell apoptosis
US8088057B2 (en) 2005-02-01 2012-01-03 James David Honeycutt Apparatus and methods to improve sleep, reduce pain and promote natural healing
US20070060954A1 (en) 2005-02-25 2007-03-15 Tracy Cameron Method of using spinal cord stimulation to treat neurological disorders or conditions
US20080039901A1 (en) 2005-06-03 2008-02-14 Kronberg James W Methods for modulating chondrocyte proliferation using pulsing electric fields
US7840272B2 (en) 2005-06-03 2010-11-23 Medrelief Inc. Methods for modulating osteochondral development using bioelectrical stimulation
CN1879906A (en) 2005-06-15 2006-12-20 郑云峰 Magnetic stimulating device for nervous centralis system and its usage method
US20090216068A1 (en) 2005-06-15 2009-08-27 Thomas Alex W Therapeutic low frequency pulsed magnetic fields and devices therefor
US20070043254A1 (en) 2005-08-18 2007-02-22 Demarco Jill R Underwear Having Detachable Magnetic Devices
US20070149901A1 (en) 2005-12-08 2007-06-28 Em-Probe, Inc. Methods and apparatus for pulsed electromagnetic therapy
US7419474B2 (en) 2006-01-03 2008-09-02 Richard Lee Non-linear therapy signal synthesizer
US20070179558A1 (en) 2006-01-30 2007-08-02 Gliner Bradford E Systems and methods for varying electromagnetic and adjunctive neural therapies
GB0604107D0 (en) 2006-03-01 2006-04-12 Barak Steven P Alarm system
US8926676B2 (en) 2006-04-11 2015-01-06 Advanced Neuromodulation Systems, Inc. Systems and methods for applying signals, including contralesional signals, to neural populations
US20090018613A1 (en) 2006-08-02 2009-01-15 Genestim, Llc Regulation of vascular endothelial growth factor (vegf) gene expression in tissue via the application of electric and/or electromagnetic fields
WO2008056414A1 (en) 2006-11-08 2008-05-15 Medical Appliance Co., Ltd. Neurotrophic factor production accelerating apparatus
US20100210894A1 (en) 2006-12-01 2010-08-19 Beth Israel Deaconess Medical Center, Inc. Transcranial magnetic stimulation (TMS) methods and apparatus
US7949403B2 (en) 2007-02-27 2011-05-24 Accelerated Care Plus Corp. Electrical stimulation device and method for the treatment of neurological disorders
US8034014B2 (en) 2007-03-06 2011-10-11 Biomet Biologics, Llc Angiogenesis initation and growth
CN101815555A (en) 2007-04-12 2010-08-25 Ivivi健康科学有限责任公司 Electromagnetic field treatment apparatus and using method thereof
US7783348B2 (en) 2007-05-03 2010-08-24 Orthocor Medical, Inc. Stimulation device for treating osteoarthritis
CA2733081C (en) 2007-08-06 2015-12-15 Great Lakes Biosciences, Llc Methods and apparatus for electrical stimulation of tissues using signals that minimize the effects of tissue impedance
US8062229B2 (en) 2007-08-10 2011-11-22 Rauscher Elizabeth A Methods and devices for measurement and treatment of pain and the treatment of inflammation and osteoporosis
US20090105781A1 (en) 2007-09-26 2009-04-23 Genestim Nascent Enterprises Llc Regulation of matrix metalloproteinase (mmp) gene expression in tumor cells via the application of electric and/or electromagnetic fields
US8167784B1 (en) 2007-12-03 2012-05-01 James David Honeycutt Apparatus and methods to generate circadian rhythm based pulsed electromagnetic fields using micro-watts of electrical energy
GB2457077A (en) 2008-02-01 2009-08-05 Julian Joshua Preston-Powers Cooling system for headwear
DE102008021575A1 (en) 2008-04-30 2009-11-05 Neue Magnetodyn Gmbh Device for stimulating a healing process
US8972024B2 (en) 2008-06-20 2015-03-03 Medrelief Inc. Systems, apparatuses, and methods for providing non-transcranial electrotherapy
US8984664B2 (en) 2008-07-08 2015-03-24 Lawrence Livermore National Security, Llc Helmet blastometer
US8801589B2 (en) 2008-08-04 2014-08-12 The Trustees Of Columbia University In The City Of New York Methods, apparatus, and systems for magnetic stimulation
WO2010056632A1 (en) 2008-11-13 2010-05-20 The Rockefeller University Neuromodulation having non-linear dynamics
US9180305B2 (en) 2008-12-11 2015-11-10 Yeda Research & Development Co. Ltd. At The Weizmann Institute Of Science Systems and methods for controlling electric field pulse parameters using transcranial magnetic stimulation
JP2013508119A (en) 2009-10-26 2013-03-07 エムキネティクス, インコーポレイテッド Method and apparatus for electromagnetic stimulation of nerves, muscles and body tissues
WO2011079309A2 (en) 2009-12-23 2011-06-30 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
CA2813036A1 (en) 2010-10-01 2012-04-05 Ivivi Health Sciences, Llc Method and apparatus for electromagnetic treatment of head, cerebral and neural injury in animals and humans
CN102151362B (en) 2011-04-12 2013-10-09 苏州悦安医疗电子有限公司 Intelligent footdrop corrector
US20130035539A1 (en) 2011-08-05 2013-02-07 Andrew Kornstein System and method for treating hair loss
WO2013067512A1 (en) 2011-11-04 2013-05-10 Ivivi Health Sciences, Llc Method and apparatus for electromagnetic treatment of cognition and neurological injury
US8343027B1 (en) 2012-01-30 2013-01-01 Ivivi Health Sciences, Llc Methods and devices for providing electromagnetic treatment in the presence of a metal-containing implant
WO2015161063A1 (en) 2014-04-16 2015-10-22 Iviv Health Sciences, Llc A two-part pulsed electromagnetic field applicator for application of therapeutic energy

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11020603B2 (en) 2019-05-06 2021-06-01 Kamran Ansari Systems and methods of modulating electrical impulses in an animal brain using arrays of planar coils configured to generate pulsed electromagnetic fields and integrated into clothing
US11517760B2 (en) 2019-05-06 2022-12-06 Kamran Ansari Systems and methods of treating medical conditions using arrays of planar coils configured to generate pulsed electromagnetic fields and integrated into clothing

Also Published As

Publication number Publication date
US10350428B2 (en) 2019-07-16
US20180200531A1 (en) 2018-07-19
US20160121135A1 (en) 2016-05-05

Similar Documents

Publication Publication Date Title
US20190351249A1 (en) Method and Apparatus for Electromagnetic Treatment of Living Systems
US10226640B2 (en) Devices and method for treatment of degenerative joint diseases with electromagnetic fields
US10426967B2 (en) Apparatus and method for electromagnetic treatment of neurological injury or condition caused by a stroke
US7758490B2 (en) Integrated coil apparatus for therapeutically treating human and animal cells, tissues and organs with electromagnetic fields and method for using same
US9433797B2 (en) Apparatus and method for electromagnetic treatment of neurodegenerative conditions
US10207122B2 (en) Method and apparatus for electromagnetic enhancement of biochemical signaling pathways for therapeutics and prophylaxis in plants, animals and humans
US9415233B2 (en) Apparatus and method for electromagnetic treatment of neurological pain
US20180104505A1 (en) Apparatus and method for electromagnetic treatment
US8343027B1 (en) Methods and devices for providing electromagnetic treatment in the presence of a metal-containing implant
US9427598B2 (en) Method and apparatus for electromagnetic treatment of head, cerebral and neural injury in animals and humans
US20140303425A1 (en) Method and apparatus for electromagnetic treatment of cognition and neurological injury
US20200094068A1 (en) Method for treatment of non-alcoholic steatohepatitis using pulsed electromagnetic field therapy
US20070173904A1 (en) Self-contained electromagnetic apparatus for treatment of molecules, cells, tissues, and organs within a cerebrofacial area and method for using same
WO2018089795A1 (en) System and method for applying a low frequency magnetic field to biological tissues
WO2007146342A9 (en) Electromagnetism for prophylaxis and opthalmic tissue repair
AU2006220660A1 (en) Pharmacological, chemical, and topical agent enhancement apparatus and method for using same
US20200044318A1 (en) Devices and Methods for the Delivery of Non-Thermal Therapeutic and Prophylactic Electromagnetic Fields from a Mobile Device
Yao et al. Effects of different frequencies of electro-acupuncture at shuigou (GV 26) on recovery of motor function in rats with focal cerebral ischemic injury
MX2008003378A (en) Integrated coil apparatus and method for using same
AU2005336126A1 (en) Integrated coil apparatus and method for using same

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION