APPARATUS AND METHOD FOR OPTIMIZING TREATMENT USING MEDICATION COMPLIANCE PATTERNS AND GLUCOSE SENSOR

- I.D. THERAPEUTICS LLC

Methods and apparatus monitor medication usage data for one patient or a population of patients, which can be processed to determine compliance patterns. Such methods and systems can associate, analyze, organize and present medication usage data, compliance patterns, and correlations between compliance patterns and outcomes data for electronic analysis or analysis by a caretaker. Such methods and apparatus permit analysis of compliance patterns to enable, for example, establishment or adjustment of safe and effective treatment regimens, and may include feedback systems for ensuring authenticity of medication and/or effects of medication on a patient. Such methods and apparatus also permit detection of medication in a biological sample of the patient through a medication monitor having a detection device, such as a skin-prick device, and a glucose sensor that can detect a target agent in the biological sample.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
BACKGROUND

The disclosure relates to methods and apparatus for monitoring, storing, and analyzing patient medication usage data and compliance patterns involving use of a sample testing device, associating compliance patterns with medication properties data and patient history data, and for determining a correlation between medication compliance patterns and other data.

Medications may include potent chemical and/or biological elements designed to induce a specific ameliorative effect on a patient's disease state or medical condition. Medication manufacturers, prescribing physicians, and caretakers have limited information about a given medication that typically includes only basic guidelines for safely and effectively prescribing very powerful substances to patients.

This dearth of information is especially significant with regard to the effects of medication treatment regimen compliance. Medication manufacturers, prescribing physicians and other caretakers struggle to effectively manage the effects on patients of non-compliant medication intake or compliance patterns. For example, without data regarding patient medication usage, caregivers face difficulty discerning whether negative effects of medication intake are the result of precise, over, or under compliance by the patient, or a prescribed treatment regimen that is ineffective or unsafe. There are few or no independent objective measures of a patient's actual compliance with a prescribed treatment regimen, aside from the patients' memory about their own historical medication compliance over time. Further, there are few or no independent objective measures of negative or ameliorative effects attributable to varying degrees of over and/or under compliance with treatment regimens.

Poor or unexpected medication treatment regimen compliance is a medical problem that poses risks to patient health and potentially increases health care costs. By way of example, a patient who has undergone an organ transplant may be prescribed a regimen of immunosuppressive medications to protect the patient's transplanted organ from being rejected by the patient's immune system. If a patient takes too much or too little of these medications, or takes them at incorrect time intervals, then the patient's body may experience a cascade of biochemical reactions that may result in the transplanted organ being rejected or other diseases being acquired, or other complications.

For example, if a patient takes too much of an immunosuppressive medication, even intermittently, the patient's inherent immunological capability may be adversely affected, thereby rendering the patient susceptible to malignancies, bacterial infections and viral, infections. The complications of an over-suppressed immune system can lead to death, severe illness that requires hospitalization, and can compromise the transplanted organ. The medical intervention often expended to redress such adverse consequences can add significant direct and indirect financial costs for the patient and the health care system, in addition to potentially limiting the patient's quality of life.

SUMMARY

Methods and apparatus are needed that provide robust checks and balances for establishing medication regimens, adjusting medication regimens, and caring for patients who are taking medications, especially for those taking immunosuppressive medication. Further, methods and apparatus are needed that permit monitoring, analysis, and recording of medication usage data and regimen compliance patterns for individual patients and patient populations. Further still, methods are needed that permit analysis of medication regimen compliance patterns in view of patient history data to enable, for example, medication manufacturers and/or prescribing caretakers to safely and efficaciously establish and adjust treatment regimens.

Methods and apparatus are also needed that allow a user of a medication, and especially an immunosuppressive medication, to quickly, conveniently and efficiently monitor and manage medication usage data so that such data can be used for the treatment of that patient and/or a population of patients. Such methods and apparatus may include determining medication usage data by means of a detection device that is configured to detect a target agent in a biological sample by converting a substance to glucose and then detecting the presence and/or amount of the detected glucose.

U.S. Patent Application Publication No. 2012/0072231 discloses methods, systems, and apparatus for monitoring patient medication usage, determining medication compliance patterns, and establishing and adjusting medication regimens. The disclosure of U.S. Patent Application Publication No. 2012/0072231 is hereby incorporated by reference in its entirety.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a perspective view of a medication monitor cartridge compartment in accordance with an exemplary embodiment.

FIGS. 2A and 2B are schematic drawings showing exemplary mechanisms of target agent (analyte) detection using a glucose sensor based on the interaction between recognition molecule A, recognition molecule B and the target, agent.

FIGS. 3A and 3B are perspective views of a medication monitor in accordance with an exemplary embodiment.

FIG. 4 is a perspective view of a medication monitor cartridge compartment in accordance with an exemplary embodiment.

DETAILED DESCRIPTION OF EMBODIMENTS

It is desirable to provide a medication monitor that can detect both usage of medication and an amount and/or quantity of a target agent in a biological sample, through the use of a glucose sensor that is operatively connected to the medication monitor to generate medication usage data. It is also desirable to then communicate that medication usage to the patient, a caregiver, and/or a data network to generate and adjust treatment regimens based on the medication usage data.

According to exemplary implementations, a medication monitor for producing medication usage data may include a housing and a detection device operatively associated with the medication monitor. The detection device may include a sensor for detecting a target agent in a biological sample from a user. For example, the biological sample may be body fluid such as one or more of blood, mine and saliva. A preferred detection device may be a skin-prick device such as a finger-prick device. The detection device may be hard wired to the medication monitor or may be wirelessly connected to the medication monitor. The sensor may include a solid support to which is attached a recognition molecule that specifically binds to the target agent in the presence of the target agent but not significantly to other agents. The sensor may also include a substance that can be enzymatic-ally converted to glucose and an enzyme that can catalyze the conversion of the substance to glucose. The enzyme may attach directly or indirectly to the recognition molecule such that in the presence of the target agent the enzyme can convert the substance into glucose. The medication monitor may include a processor that produces medication data based on an amount of glucose sensed and may include a communications port configured to communicate the medication data.

According to some embodiments the housing body may include a housing for containing medication or medication packaging. The housing body may define an opening to accommodate insertion and removal of medication or medication packaging. The housing body may also include a lid that opens (e.g., by sliding, pivoting or removal) to removably cover the opening. A medication sensor may be provided that is configured to sense identity of the medication. A removal sensor may also be provided that senses medication or medication packaging removal or device opening. Hie processor, in addition to producing medication data based on the amount of glucose sensed, may also produce medication usage data based on the sensed removal. The medication sensor and the removal sensor may be provided to monitor patients' compliance with treatment regimens by keeping back of the medication in the medication monitor. Medication data and medication usage data may be correlated by way of one or more processor onboard the medication monitor and/or in an external data network with which the medication monitor can communicate.

According to exemplary embodiments, the target agent may be an immunosuppressant detected in a biological sample, such as the user's blood, such as Tacrolimus, or a metabolite thereof. The enzyme may be attached to a Tacrolimus analogue molecule that competes less strongly than Tacrolimus or a metabolite thereof for binding to the recognition molecule. Alternatively, the enzyme may be attached to a molecule that binds to Tacrolimus or a metabolite thereof that binds to the recognition molecule.

Examples of the solid support may include a bead or a membrane. The recognition molecule may include a nucleic acid molecule, a protein, a polymer, or an antibody that specifically binds to the target agent. The enzyme, for example, may be an invertase, sucrase or sucrase-isomaltase that can convert sucrose to glucose, a maltase that can convert maltose into glucose, a trehalase that can convert trehalose into glucose, an amylase that can convert starch into glucose, or a cellulase that can convert cellulose into glucose. Preferably, the enzyme is invertase. The sensor may include a plurality of sensors with each sensor of the plurality of sensors sensing a target agent specific to that sensor. Different ones of the plurality of sensors may each detect the same target agent or different target agents. An example of a sensor that quantitatively detects a target agent by detecting glucose is disclosed in U.S. Patent Application Publication No. 2012/0315621, which is hereby incorporated by reference in its entirety.

The medication monitor may include a communications port that is configured to wirelessly communicate the medication usage data to a data network to monitor compliance patterns and/or interacting between compliance patterns and sample levels of the target agent(s).

The medication monitor may include a sensor to detect, for example, an origin of manufacture of the pre-filled medication insert. Each pre-filled medication insert may have a barcode, RFID tag or other identifier that relays the origin of manufacture data and/or other data to verify authenticity of the medication. Hie monitor may include a reader such as a barcode reader, an RFID label reader or other information detector that verifies that the medication is not counterfeit or is otherwise a desired mature form and/or dosage of the medication at the time the medication monitor is loaded and/or at the time of use by the patient. The RFID information may also be sent to a receiving system such as a remote server, which verifies, and may send a signal to the monitor and/or another receiver, regarding whether the medication is appropriate. The monitor may also or instead include a sensor to detect a specific shape of a medication, the specific shape of the pre-filled medication insert, a medication made by a unique manufacturing process, or an orientation of the medication within the housing body to determine whether the medicine correctly loaded into the monitor and is not counterfeit or otherwise inappropriate.

Embodiments include methods that may accommodate optimizing treatment of a patient such as a transplant recipient or a population of such patients. Methods may include generating usage data regarding a patient's compliance with a medication treatment regimen with a medication monitor. Methods may include providing the usage data to a central server over a predetermined period of time, and determining the patient's pattern of compliance with the treatment regimen. Methods may include obtaining test results from a detection device, those test results being indicative of a target agent such as the medication or a metabolite of the medication, by collecting a biological sample with a detection device that is operatively connected to the medication monitor, and sensing the target agent with a target agent sensor provided with the detection device.

Target agent presence and/or levels (medication data) can then be correlated in one or more processor in the medication monitor and/or in a data network with which it can communicate. The sensor may comprise a solid support to which may be attached a recognition molecule that permits detection of the target agent, wherein the recognition molecule specifically binds to the target agent in the presence of the target agent but not significantly to other agents. The sensor may also include a substance that can be enzymatically converted to glucose, and an enzyme that can catalyze the conversion of the substance to glucose in the presence of the target agent.

Methods may include detecting the glucose produced from the substance with a glucosimeter to generate the medication data. The detection of glucose may indicate a presence of the target agent in the biological sample, and may also indicate levels of target agent in the biological sample, and an absence of detected glucose may indicate an absence of the target agent in the biological sample. Methods may also include correlating the patient's pattern of compliance with the medication data from the detection device, and outputting at least one recommendation from the above-mentioned processors). The recommendation may include replacing at least one medication used in the treatment, regimen with at least one different medication, changing at least one dosage of at least one medication used in the treatment regimen, changing the timing of recommended usage of at least one medication in the treatment regimen, not changing the treatment regimen, and/or taking action to improve the patient's compliance.

Methods may also include correlating the patient's pattern of compliance and medication data with patterns of compliance and medication data from a population of patients, and outputting the at least one recommendation based on both the patient's pattern of compliance and medication data and the patterns of compliance and medication data from the population of patients. The treatment regimen may be a standardized treatment regimen or an individual patient treatment regimen.

Exemplary methods may include comparing the patient's compliance pattern with the medication data, and, based on the comparison, outputting a recommendation of replacing an immunosuppressant medication used in the treatment with a different medication, changing a dosage amount and/or frequency of an immunosuppressant medication, or not changing the regimen.

According to exemplary implementations, a self-contained medication monitor for producing medication usage data may be provided. The self-contained medication monitor may include a housing body, a first detection device for detecting removal of medication from the self-contained medication monitor and a second detection device operatively connected to the medication monitor and having a target agent sensor for detecting a target agent in a biological sample from a user. The target agent sensor may comprise a solid support to which is attached a recognition molecule that specifically binds to the target agent in the presence of the target agent but not significantly to other agents, a substance that can be enzymatically converted to glucose, and an enzyme that can catalyze the conversion of the substance to glucose. The enzyme may attach directly or indirectly to the recognition molecule, and in the presence of the target agent the enzyme can convert the substance into glucose. The self-contained medication monitor may also include a processor that produces information and/or recommendations based on detected removal of the medication and an amount of glucose sensed, and that outputs the information and/or recommendations to the patient or a caregiver. The processor may also output a recommendation to a health care provider, and may assist the health care provider in determining target drag levels or benchmark chug levels. The processor may output raw glucose concentration data, compare the amount glucose sensed with a pre-determined or other baseline glucose level, and/or quantitatively determine the amount of the target agent present in the biological sample based on a difference between the baseline glucose level and the amount of glucose sensed. The self-contained medication monitor may also include a display device configured to display the information and/or recommendations to the patient. Such recommendations may include, for example, a reminder to take a medication in accordance with a pre-established regimen, a recommendation to expedite or delay a dose of medication, and/or a recommendation to consult with a caregiver. In comparison to current tests, where the patient must visit his or her healthcare provider to have a drag level test conducted, methods using such a device are more convenient, may be performed more often, may be more closely tied to direct feedback from when the drug was taken, and may be measured independently of when the last drag was taken. For example, a drug dose does not have to be delayed while a patient visits the doctor's office to have drug level tests done.

FIG. 1 shows a treatment regimen compliance monitoring system having a medication monitor 401, and a receiving system 410. The receiving system 410 may be in communication with a storage system such as, for example, a server 420. The receiving system 410 may also be in communication with, for example, a reporting system 430. The receiving system 410 may also be in communication with a detection device 440. Any or all of the receiving system 410, the medication monitor 401, the server 420, the detection device 440, and the reporting system 430 may communicate by a wireless connection, wired connection, or a combination thereof over the internet, local area network, PSTN, or the like. Medication monitor 401 may include a lid 405 having a transparent window 408. FIG. 1 shows the receiving system 410 as separate from the medication monitor 401, but they could be combined in a single device. The transparent window 408 may enable viewing of a medication contained by the medication container 401. Further, medication monitor 401 may be constructed to house multiple medications separately or together, and may be configured to separately monitor each of the housed medications. Medication monitor 401 may be constructed to house one or more medications in various dosage forms. For example, medication monitor 401 may be constructed to house and dispense oral suspension, injection, inhalation, gel, cream, capsule and/or solid dosage forms.

The medication monitor 401 may also include a detection device 404, and/or be usable with detection device 440. The detection device 404/440 includes a detection portion 403 and at least one glucose sensor 406 that is used to detect the presence and/or level of a target, such as a target anatyte/agent in a biological sample from the user. Exemplary biological samples may include blood, serum, plasma, mine or saliva. The detection portion 403 may, for example, include a skin-prick device that can collect a sample of the user's blood after a pin prick anywhere on the user's body. The skin prick device could, for example, be a finger-prick device that obtains a sample of the user's blood after a finger-prick. The glucose sensor 406 includes a recognition molecule that is specific for the target agent, preferably attached to a solid support, and an enzyme that can catalyze the conversion of a substance into glucose (for example in the presence of the target agent). The enzyme can attach directly or indirectly to the recognition molecule. Although only one detection device 404/440 is illustrated in FIG. 1, it is understood that the detection device 404/440 may include a plurality of glucose sensors 406 and/or detection portion 403. The glucose sensors 406 may be configured to detect the same target agent or may each detect a different target agent found in the biological sample.

FIGS. 2A and 2B provide an overview of the glucose sensor 406 and exemplary methods of its use by respective competition and sandwich assay methods. In FIGS. 2A and 2B, the recognition molecule A and recognition molecule B (a recognition molecule that can bind to the target agent with high specificity) can be the same or different molecules, wherein both can bind to the analyte (referred to herein as the target agent). The enzyme that can catalyze the conversion of a substance (enzyme substrate) into glucose is conjugated with an analyte analogue (that is, an analogue of the target agent; FIG. 2A) or another recognition molecule B (FIG. 2B) to form enzyme-analyte analogue conjugate (FIG. 2A) or enzyme-recognition molecule B conjugate (FIG. 2B), respectively. A substrate provided in the detection device 404 can be catalytically converted into glucose by the enzyme, and the glucose produced can be detected and/or quantified by a glucose meter. The target agent (analyte) can be any substance that can be recognized by recognition molecule A or recognition molecule B.

FIG. 2A shows, for example, a release-based assay. Initially, enzyme-analyte analogue conjugate binds to the solid support through the interaction between enzyme-analyte analogue conjugate and recognition molecule A. When samples containing the target agent are applied to the solid support, the enzyme-analyte analogue conjugate will be released as a result of the competition between enzyme-analyte analogue conjugate and target agent in binding with recognition molecule A. The concentration of enzyme-analyte analogue conjugate released can be proportional to the target agent concentration in the sample. After removal of fee solid support, enzyme-analyte analogue conjugate remaining in the solution can catalyze the conversion of the substrate into glucose, which is detected by a glucose meter, and the readout can be proportional to the analyte concentration.

FIG. 2B shows, for example, a binding-based assay. Initially, recognition molecule A is immobilized to the solid support. When a sample containing or suspected of containing the target agent (analyte) is applied to solid support the analyte binds to recognition molecule A. Subsequently, enzyme-recognition molecule B conjugate, is added and will bind to analyte that is bound to the recognition molecule A, forming a sandwich structure. The amount of enzyme-recognition molecule B conjugate bound can be proportional to the concentration of analyte in the sample. After applying the substrate (e.g., sucrose) to the solid support, the bound enzyme-recognition molecule B conjugate can convert substrate into glucose, which is detected by a glucose sensor such as a glucose meter, and the readout can be proportional to the analyte concentration. In this way, in the presence of more target agent, more enzyme will be bound to the solid support, and the solid support can convert more sucrose into glucose, giving a larger readout in glucose meter.

The glucose meter may be any medical device for determining the approximate concentration of glucose in a sample. Glucose meters, such as a personal glucose meter (PGM), typically display the level of glucose in mg/dl or mmol/l. This disclosure is not limited to a particular brand of glucose meter, though examples include ACCU-CHEK®, ONETOUCH®, PRODIGY®, ADVOCATE®, AGAMATRIX®, ASCENSIA®, BIONTME®, CLEVERCHEK®, EASYGLUCO®, FREESTYLE®, MAXIMA®, MEDISENSE®PRESTIGE®, TRUEBALANCE®, TRUE-TEST®.

Different types of recognition molecules, enzymes, solid supports, etc. and their different binding configurations are described, for example, in U.S. Patent Application Publication No. 2012/0315621, which is incorporated by reference in its entirety.

The glucose sensor 406 can be used to detect any target agent of interest. Thus, the methods and devices provided herein can be used to detect any target agent of interest, such as the specific examples provided herein. Selecting an appropriate recognition molecule that permits detection of the target agent allows one to develop a sensor that can be used to detect a particular target agent. One example of a target agent as an immunosuppressant is Tacrolimus or a metabolite thereof. Recognition molecules could be antibodies (monoclonal or polyclonal) or aptamer based. The antibodies or aptamers have specificity to the target agent. They can be produced by known methods of antibody or aptamer production or can be purchased from OEM suppliers. However, one skilled in the art will appreciate that other target agents can be detected with the disclosed sensors and devices using the disclosed methods. For example, the other target agents could be any of the substances discussed, for example, in paragraphs [0078]-[1010] of U.S. Patent Application Publication No. 2012/0072231, which is hereby incorporated by reference in its entirety.

As seen in FIG. 1, the medication monitor 401 may include a display 400. The display 400 may be, for example, a liquid crystal display that functions to present data generated or received by the medication monitor 401, or other information. Lid 405 may alternatively or additionally include a display 400. The lid 405 may be constructed to slideably and/or hingedly move between an open state and a closed state to accommodate access to and closure of one or more compartments of the medication monitor 401, thereby enabling a user to view the display while viewing and/or accessing at least one compartment of the medication monitor 401.

The medication container 401 may be battery powered, may include a SIM card, and/or may be GPS enabled. Medication container 401 may be a micro-electronic “smart” pill box mat accepts a unique compartmentalized pill container insert that can either be hand-loaded with individual dosage forms of medications, or alternatively, the pill box can accept a custom designed, pre-filled cartridge. The pre-filled cartridge may include RFID or other labels that the medication container 401 can read to confirm the identity and/or amount of the medication contained in the pre-filled cartridge as an anti-counterfeiting measure to determine whether the pre-filled cartridge is authentic. Medication container 401 may be constructed to contain one or more types of medications that are each compartmentalized for ease of patient identification, dispensing, and refilling. The medication container 401 may, for example, be constructed of aesthetically and economically designed injection molded thermoplastic. The medication container itself and/or a cartridge/magazine for it may be childproof or tamperproof, and/or the monitor may be usable with childproof and/or tamperproof containers. The childproof/tamperproof features may be mechanical, electronic, electromechanical or other. For example, they may involve one or more biometric identification features, such as a fingerprint recognition lock, and/or electronic codes, and may optionally include time lock features to help control untimely or excess access to the contained medication.

FIG. 3A shows a medication monitor 501 having a body 510. The body 510 may include a housing 515. The housing 515 may include one or more compartments 520. The medication monitor 501 may include a lid 502. The lid 502 may include a display 508. The display 508 may be, for example, a liquid crystal display, or any other suitable display now known or later developed. The lid 502 may be attached to the body 510 with a pivot 525 whereby the lid 502 may slideably move to cover or uncover the housing 515 of the body 510. The medication usage monitor 501 may have a detection device 404 that includes a detection portion 403 and at least one glucose sensor 406.

FIG. 3B shows medication monitor 501, which may include a lid 502. The lid 502 may include a display 508. The display may be, for example, a liquid crystal display, or any other suitable display now known or later developed. The lid 502 may be attached to the body 510 by a hinge 528 so that the lid 502 may be lifted upward with respect to the body 510 to provide access to the housing 515 and one or more compartments 520 located therein. The housing 515 may be constructed to receive and retain medication dosage forms, medication containers, medication cartridges that are factory refilled and/or refillable by consumers, and/or blister packs containing medication. One or more sensor may be provided to determine when medication is removed from the housing; such sensors may be specific to opening of or dosage removal from a single compartment, or from any of several compartments. Similar to the FIG. 3A, the detection monitor 501 illustrated may include a detection device 404 that includes a detection portion 403 and at least one glucose sensor 406.

The medication monitor 501 may serve as a periodic dispensing device. The medication monitor 501 may also serve as a monitor for determining medication refill needs and communicating related messages. The medication monitor 501 may be one unit or multiple units, and may include multiple containers or compartments for organizing multiple medications. If multiple units are provided to a single patient, they preferably are capable of communicating, and programmed to communicate, with one another to ensure integrated reporting of usage data. The medication monitor 501 may be sized to fit in a pocket, or a purse, or may be larger. The medication monitor 501 may be constructed to hold and organize portable medication monitors. It may optionally include one or more processors as described above.

The medication container 401 may include on-board micro-processing technology. The on-board micro-processing technology may function to record and/or report at least one of a time, a date, and a location of when a medication is inserted or removed, or a pre-loaded cartridge is inserted or removed, and/or other information as discussed herein. The micro-processing technology may function to record the number of medications or dosage forms in a specific medication compartment at any given time. The micro-processing technology may record a date, a location, and/or a time when the lid 408 is opened, and the date, the location, the time, and/or the amount when specific medication is removed or inserted. The micro-processing technology may function to determine medication compliance patterns and medication data, establish or recommend adjustment of treatment regimens in view of compliance data, medication properties data, medication data, and patient history data, and determine correlations among them, or those functions may be performed at a remote location.

Medication container 401 may include a transmitter 412 that effects communication of medication usage data, medication data, compliance patterns or other information discussed herein generated by the medication container 401, including medication usage data based on an output from the glucose sensor 406. The transmitter may effect communication to at least one of the receiving device 410 and the server 420. The communication may comprise the information generated or recorded by the micro-processing technology of the medication monitor 401, the detection device 440, and or other information input by a patient or caregiver. Medication monitor 401 may include a port for communicating data, for example, wirelessly or by a Universal Serial Bus connection.

Information may be transmitted from the monitor 401 to a receiving system 410. The receiving system 410 may include a communications port such as a transceiver for receiving information and transmitting information to the reporting system 430 and/or the server 420. The receiving system 410 may include, for example, a port for communicating wirelessly or over a Universal Serial Bus connection. The receiving system 410 may include a remote storage system that receives and stores information from at least one of the medication monitor 401 and the receiving system 410. The receiving system 410 and/or remote storage device may implement algorithms to analyze information such as medication usage data, established treatment regimen data, medication properties data, medication data, and patient history data, including outcomes data. Outcomes data may be received by receiving system 410 from, for example, server 420 or a healthcare provider, whether private or publicly accessible. A monitored patient's attending physician, healthcare system representative, or laboratory information system, a data collection center, or the like may electronically provide a patient's outcomes data to the receiving system. Medication properties data such as medication interaction data may be received from a central database or other repository of medication interaction data.

FIG. 4 shows a medication cartridge. The medication cartridge may be constructed to be inserted into and/or removed from the medication monitor. The medication cartridge may be pre-filled with medication and/or refillable. The cartridge may accommodate any dosage form. In one version of the cartridge, 610 is a spring or other mechanism that keeps pushing stacked pills up so that one or more pills can be removed at a time when the cartridge is opened. In another version of the cartridge, 610 is a mechanism with art opening that allow one or more pills to be removed from the medication monitor. In this version, the spring or other mechanism may be contained in the package or magazine of stacked of pills. Either mechanism may also contain a sensor (optic or other) that senses when a pill is removed from the magazine or cartridge. A pre-finable medication cartridge 615 according to embodiments is illustrated in FIG. 4. The pre-fillable medication cartridge 615 may have a specific shape to match a specific shape of a housing of the medication monitor. The monitor may include a sensor to detect, for example, an origin of manufacture and/or identity of contents of the pre-fillable medication cartridge 615. Each pre-fillable medication cartridge 615 may have a barcode, an RFID tag, or other information source that relays the origin of manufacture data and/or other data, for example to verify authenticity and/or identity of the medication. The monitor may also include a sensor to detect a specific shape of a medication, the specific shape of the pre-fillable medication cartridge 615, a medication made by a unique manufacturing process, or an orientation of the medication within the housing body as a way of protecting against counterfeiting and/or misuse of medication. The monitor may also include a barcode reader, an RFID label reader, or another information detector that allows for verifying whether the medication is counterfeit and/or the correct medication at the time of loading of the monitor and/or at the time of use by the patient. In addition to forwarding compliance data, the monitor may also send the information read by the reader to a receiving system such as a remote server, preferably the same remote server as discussed above, to verify whether the medication is authentic and/or the correct medication. The receiving system may compare the information received to the information in a database, and report issues to the patient and/or family, caretakers, support organizations, health care providers, etc.

The medication monitor may have one or more cartridges, which may or may not be separable as shown in FIG. 4. These multiple cartridges may hold the same chug (for example if higher doses or multiple pills are needed at a dose time) or different drugs, hi one version there may be a mechanical mechanism in the medication monitor which slides past 610 of each cartridge, removing one or more pill (the pills can be different thicknesses and/or can be dispensed in groups) from each cartridge; the mechanism may be controlled mechanic ally or electromechanically. The magazine may be packaged with pills of existing shape and design, or pills may be designed with specific dimensions and characteristics allowing for pills to interface properly with packaging magazines, cartridges and/or medication monitors. The medication monitor may be configured to detect whether the pills provided in die magazine are authentic and appropriate, for example, by detecting any one or a combination of size, shape, color, unique manufacturing process of the pills, or unique manufacturing process of the pre-filled cartridge, as a barrier to counterfeiting or improper filling (e.g., with an incorrect medication). The outer housing of the pre-packed magazine of pills may have vapor or other barrier properties necessary to maintain the stability of the medication contained in them.

The algorithms used in methods, apparatus and systems described herein may be designed to determine a medication compliance pattern, or to analyze a medication compliance pattern received from the medication monitor 401. Further, the algorithms may be designed to correlate one or more patient medication compliance patterns with medication data and/or outcomes data, which also may be analyzed by way of the algorithms. Still further, the algorithms may analyze medication interaction data in view of medication data, outcomes data and medication compliance patterns to accommodate treatment regimen establishment and/or adjustment. Algorithms may also organize outcomes data, medication usage data, medication data, treatment regimen compliance patterns, and/or a combination thereof for presentation to a caretaker or other interested party. The organization may be effected by a ranking system in which values are attributed to aspects of the data to signify a level of importance to a caretaker or other interested party. For example, an output of an algorithm executed in accordance with an exemplary embodiment may be a warning that may be sent to at least one of a medication monitor 401, receiving system 410, server 420, and reporting system 430. The presentation may be textual, graphical, auditory, and/or diagrammatic. The data may be presented, for example, on a laptop, desktop or workstation computer display, or may be presented on a handheld device such as reporting system 430.

For example, an exemplary algorithm for methods, systems, and apparatus may include inputting a starting dosing regimen of a medication for treatment of a particular condition. Then, compliance patterns may be input. Medication data, medication properties data and patient history data may also be input. The algorithm may output, based on compliance patterns, medication data, medication properties data, and/or patient history data a new regimen or report.

An exemplary algorithm for methods, systems, and apparatus may include inputting medication data generated by obtaining a biological sample with a detection device, such as a skin-prick device, and detecting a presence and/or quantity of a target agent other than glucose in the biological sample through a glucose sensor.

For example, for a kidney transplant recipient (patient) with a safety risk of malignancy or infection, the algorithm may include the step of inputting a starting dosing regimen of cyclosporin. A at x mg per day to provide post kidney transplant immunosuppression. The compliance pattern may be input, which may indicate that the patient has a pattern of high compliance with very few missed doses. The medication data, medication properties data and patient history data, including patient physical data (e.g., weight, body mass index, gender, etc.), patient cyclosporin A measurements, and other data, may be input. The algorithm may also consider malignancy and infection risk data linked to population compliance patterns. Taking these variables into account, the algorithm may then output a new treatment regimen that includes lowering a dose amount of the medication to y mg per day, for example, if compliance is low and/or target agent levels are high. Alternatively, the treatment regimen may be changed to one that is not correlated with malignancy or infection in view of the duration of the given patient's high, compliance pattern and a desired level of target agent in the sample.

Over time, the database is populated with information from patients who have had malignancies or infections correlatable to variations among their individual compliance patterns. Specific compliance patterns and/or target agent levels that have a high probability of resulting in malignancies or infections are identified and are included in the algorithm. The algorithm routinely assesses each individual's ongoing compliance pattern and target agent levels. When an individual pattern is developing a correlation with a malignancies- or infections-related pattern, the algorithm outputs an appropriate and/or pre-established dose reduction and/or other treatment regimen change.

The algorithm may utilize models or subroutines in addition to assessing direct relationships between compliance patterns, target agent levels and malignancies or infections. For example, pharmacokinetic models (single compartment and others) can be utilized to project the resulting dynamic chug levels for the specific individual based on the specific individual's compliance pattern and medication data, allowing intervention to be engaged prior to a malignancy or infection event when projected drug exposure is too high. This approach can also be utilized with appropriate PK and/or ADME models when the patient is also prescribed other medications that may have ding-drug interactions such as inducing or inhibiting drag metabolism. As the database expands and includes existing and future biomarkers of malignancy or infections, the algorithm may establish relationships between individual compliance patterns and medication data and resulting changes in these biomarkers, allowing for the engagement of interventions (e.g., dose reduction) prior to a malignancy or infection event.

In another example, a compliance pattern may show that a patient occasionally misses doses but takes medication consistently and has stable but low target agent levels. The patient history data may show chronic rejection risk associated with the compliance pattern and/or target agent levels, and also show chronic allograft nephropathy data indicating histological tubulointestinal fibrosis and tubular atrophy. The algorithm may output a new dosing regimen that increases the dose, and or the treatment regimen may be changed to one that is not correlated with chronic rejection with the given patient's specific compliance pattern and/or target agent levels.

Over time, the database is populated with information from patients who have had chronic rejection correlatable to variations among their individual compliance patterns and/or target agent levels. Specific compliance patterns and/or target agent levels that have a high probability of resulting in chronic rejection are identified and are included in the algorithm. The algorithm routinely assesses each individual's ongoing compliance pattern and/or target agent levels. When an individual pattern is developing a correlation with a chronic rejection-related pattern, the algorithm outputs an appropriate and/or pre-established dose increase and/or other treatment regimen change.

The algorithm may utilize models or subroutines in addition to assessing direct relationships between compliance patterns and/or target agent levels and chronic-rejection. For example, pharmacokinetic models (single compartment and others) can be utilized to project the resulting dynamic ding levels for the specific individual based on the specific individual's compliance pattern and/or target agent levels, allowing intervention to be engaged prior to the chronic rejection. The intervention may be a behavioral intervention to change the individual's compliance pattern and/or dose amount and/or other treatment regimen changes (e.g., prohibiting administration of certain types of medications). As the database expands and includes existing and future biomarkers of chronic rejection, the algorithm may establish relationships between individual compliance patterns and resulting changes in these target agent levels, allowing for the engagement of interventions (e.g., dose increase) prior to a rejection event.

In another example, a patient with a risk of acute rejection may be given a starting dosing regimen of a ding for post kidney transplant immunosuppression. A compliance pattern that is input may show that the patient has had many missed doses and periods of missed doses. Medication data may show low or highly variable target agent levels. Medication properties data and patient history data may be input. The medication properties data, patient history data, and compliance pattern may be analyzed to determine and output, an intervention prior to acute rejection of the transplanted kidney. For example, the intervention may be warning messages to the patient, family, support organizations; change of medications; change of dosage timing and/or amounts; and the like.

Over time, the database is populated with information from patients who have undergone acute rejection correlatable to variations among their individual compliance patterns and/or target agent levels. Specific compliance patterns and/or target agent levels that have a high probability of resulting in acute rejection are identified and are included in the algorithm. The algorithm routinely assesses each individual's ongoing compliance pattern and/or target agent levels. When an individual pattern is developing a correlation with an acute rejection-related pattern, the algorithm outputs an appropriate and/or pre-established intervention.

The algorithm may utilize models or subroutines in addition to assessing direct relationships between compliance patterns and/or target agent levels and acute rejection. For example, pharmacokinetic models (single compartment and others) can be utilized to project the resulting dynamic chug levels for the specific individual based on the specific individual's compliance pattern and/or target agent levels, allowing intervention to be engaged prior to the acute rejection event, for example when the drag exposure is too low or too intermittent. As the database expands and includes existing and future biomarkers (target agents) of acute rejection, the algorithm may establish relationships between individual compliance patterns and/or target agent levels and resulting changes in these biomarkers, allowing for the engagement of interventions prior to an acute rejection event.

The medication usage data, medication interaction data, outcomes data, medication data and medication compliance pattern(s) analyzed by the algorithms of the receiving system 410 may be made available to a patient's physician or other interested party by way of a secure website. A healthcare provider may understand a patient's individual medication compliance patterns and medication data and thereby perform informed establishment and/or adjustment of the patient's treatment regimen. Over time, as data builds on compliance patterns and/or target agent levels for populations of patients in specific disease states, such pattern data forms a registry that can provide profound insights into the relationship between patient medication compliance patterns and/or target agent levels and treatment regimens with individual patient medical outcomes or population medical outcomes. For example, such a database of patients with common disease indications can be a profound resource for improving public health and lowering the cost of medicine in specific disease states.

Using immunosuppressants in transplant as an example, over the past 20 years improvements in the kidney transplant and post transplant care process have improved 1-year graft survival rates, yet long term graft survival rates (5 years or greater) have not improved. A significant cause of the long term graft failures is declining medication compliance by individual patients. By utilizing methods, systems, and or apparatus described herein, many of these long term graft failures can be prevented, avoiding the high cost of returning to dialysis and re-transplant.

A treatment regimen may be input to a processor, defined as taking two tablets per day, at 12 hour intervals—i.e., one tablet at 8:00 a.m. and one tablet at 8:00 p.m. The input regimen would include a window for each dose, such as plus or minus 30 minutes. The usage data from a monitor would show when each tablet was taken, for example by noting the time of opening of a single-tablet compartment in the monitor. The usage data could be communicated in real time, or stored and communicated in batches, to a compliance pattern processor. The compliance pattern processor could identify a pattern over time, e.g., one month, of for example, timely, near-miss (e.g., within 30 minutes on one side of the window), distant-miss (e.g., between 30 minutes and two hours on one side of the window), and complete-miss doses. Preferably, the compliance pattern processor would also identify patterns of misses before the window and/or patterns of misses after the window. A treatment regimen processor would receive the compliance pattern. At the same or different intervals, patient fluid samples can be taken, for example, by a finger-prick device using the glucose meter, target agent levels in the sample can be determined. The target agent levels could optionally be correlated with the compliance pattern or considered separately. The processor would optionally also contain previously-stored patient history, medication properties, and other data, and could also continue to receive such data, and correlate it to associated compliance patterns. Upon correlating a new compliance pattern or medication data pattern to outcome data, for example, a pattern of increasingly distant misses and complete misses of the evening window but not of the morning window, and/or reduced morning target agent levels, to reduced efficacy of the tablets, it could generate an adjusted treatment regimen of one larger-dose, controlled release tablet to be taken only during the morning window. Outcomes data associated with this new treatment regimen could be input by, e.g., the patient's healthcare provider or by a diagnostic tool packaged with or part of a medication monitor, preferably tailored to a medication packaged with the monitor.

What has been described and illustrated herein are preferred embodiments of the invention along with some variations. The terms, descriptions and figures used herein are set forth by way of illustration only and are not meant as limitations. Those skilled in the art will recognize that many variations are possible within the spirit and scope of the invention.

Claims

1. A medication monitor comprising:

a housing configured to contain medication;
a detection device having a target agent sensor for detecting a target agent in a biological sample from a user, wherein
the target agent sensor comprises: a solid support to which is attached a recognition molecule that specifically binds to the target agent in the presence of the target agent but not significantly to other agents; a substance that can be enzymatically converted to glucose; and an enzyme that can catalyze the conversion of the substance to glucose in the presence of the target agent;
a processor that produces medication data based on an amount of glucose sensed; and
a communication port configured to communicate the medication data to a data network.

2. The medication monitor according to claim 1, wherein

the detection device includes a sample collection device.

3. The medication monitor according to claim 2, wherein

the sample collection device is a skin-prick device.

4. The medication monitor according to claim 1, wherein

the detection device is wirelessly connected to the medication monitor.

5. The medication monitor according to claim 1, wherein

the detection device is hard wired to the medication monitor.

6. The medication monitor according to claim 1, wherein

the housing includes a section for containing medication packaging, the housing defining an opening to accommodate insertion and removal of the medication packaging.

7. The medication monitor according to claim 6, further comprising:

a lid that opens to removably cover the opening;
a medication sensor that is configured to sense identity of the medication; and
a removal sensor that senses medication or medication packaging removal or device opening, wherein
the processor also produces medication data based on the sensed removal.

8. The medication monitor according to claim 1, wherein the target agent is an immunosuppressant or a metabolite thereof.

9. The medication monitor according to claim 1, wherein the target agent is Tacrolimus or a metabolite thereof.

10. The medication monitor according to claim 9, wherein the enzyme is attached to a Tacrolimus analogue molecule or a Tacrolimus metabolite analogue molecule that competes less strongly than Tacrolimus or the metabolite thereof for binding to the recognition molecule.

11. The medication monitor according to claim 9, wherein the enzyme is attached to a molecule that binds to Tacrolimus or a metabolite thereof that is bound to the recognition molecule.

12. The medication monitor according to claim 1, wherein the solid support comprises a bead or a membrane.

13. The medication monitor according to claim 1, wherein the recognition molecule includes a nucleic acid molecule, a protein, a polymer, or an antibody that specifically binds to the target agent.

14. The medication monitor according to claim 1, wherein the enzyme is

an invertase, sucrase or sucrase-isomaltase that can convert sucrose to glucose,
a maltase that can convert maltose into glucose,
a trehalase that can convert trehalose into glucose,
an amylase that can convert starch into glucose, or
a cellulase that can convert cellulose into glucose.

15. The medication monitor according to claim 1, wherein the target agent sensor includes a plurality of sensors, each sensor of the plurality of sensors sensing a target agent specific to that sensor.

16. The medication monitor according to claim 15, wherein each sensor of the plurality of sensors is configured to detect a different target agent.

17. The medication monitor according to claim 1, wherein the communications port is configured to wirelessly communicate the medication data to a data network.

18. The medication monitor of claim 7, wherein the medication sensor is configured to sense an indicium of origin of manufacture of the medication to verify authenticity of the medication.

19. The medication monitor of claim 18, wherein the indicium of origin is provided by an information source associated with a pre-filled medication cartridge that is insertable into said opening.

20. The medication monitor of claim 19, wherein the information source is at least one member selected from the group consisting of an RFID chip, a barcode, and a shape of the cartridge.

21. The medication monitor of claim 18, wherein the information source is at least one member selected from the group consisting of size, shape and color of the medication.

22. The medication monitor of claim 19, wherein the housing includes a plurality of housings for containing a plurality of pre-filled medication cartridges.

23. The medication monitor of claim 22, wherein a shape of each of the plurality of housings is configured to match a specific shape of each pre-filled medication cartridge.

24. The medication monitor of claim 1, wherein

the communication port is configured to output a recommendation to a health care provider to enable the health care provider to determine target drag levels or benchmark drag levels.

25. The medication monitor of claim 1, wherein

the processor is configured to compare the amount glucose sensed with a baseline glucose level and quantitatively determine an amount of the target agent present in the biological sample based on a difference between the baseline glucose level and the amount of glucose sensed.

26. A method of optimizing treatment of a medication recipient patient, comprising:

generating usage data regarding a patient's compliance with a medication treatment regimen with a medication monitor;
providing the usage data to a central server over a predetermined period of time;
determining the patient's pattern of compliance with the medication treatment regimen;
collecting a biological sample with a detection device that is operatively connected to the medication monitor;
sensing a target agent with a target agent sensor provided with the detection device, the sensor comprising: a solid support to which is attached a recognition molecule that permits detection of the target agent, wherein the recognition molecule specifically binds to the target agent in the presence of fee target agent but not significantly to other agents; a substance that can be enzymatically converted to glucose; an enzyme that can catalyze the conversion of fee substance to glucose in the presence of the target agent; and
detecting the glucose produced from the substance with a glucose meter to generate medication data, wherein
the detection of glucose indicates a presence of the target agent in the biological sample, and an absence of detected glucose indicates an absence of the target agent in the biological sample;
correlating the patient's pattern of compliance with the medication data; and
outputting at least one recommendation selected from the group consisting of:
replacing at least one medication used in the treatment regimen with at least one different medication, changing at least one dosage of at least one medication used in the treatment regimen, changing the tuning of recommended usage of at least one medication in the treatment regimen, and not changing the treatment regimen.

27. The method of claim 26, wherein the medication data relates to a concentration of medication in the patient's bloodstream.

28. The method of claim 26, wherein the medication data is provided to the central server.

29. The method of claim 28, wherein the medication data is provided to the central server by the medication monitor.

30. The method of claim 26, wherein

the detection device includes a skin-prick device.

31. The medication monitor according to claim 26, wherein

the detection device is wirelessly connected to the medication monitor.

32. The method of claim 26, wherein

the detection device is hard wired to the medication monitor.

33. The method of claim 26, wherein

the biological sample includes at least one of blood, urine and saliva.

34. The method of claim 26, wherein the patient is a transplant recipient, and the medication comprises an immunosuppressant.

35. The method of claim 34, wherein the immunosuppressant is Tacrolimus.

36. The method of claim 35, wherein the enzyme is attached to a Tacrolimus analogue molecule or a Tacrolimus metabolite analogue molecule that competes less strongly than Tacrolimus or a metabolite thereof for binding to the recognition molecule.

37. The method of claim 35, wherein the enzyme is attached to a molecule that binds to Tacrolimus or a metabolite thereof that is bound to the recognition molecule.

38. The method of claim 26, further comprising:

correlating the patient's pattern of compliance and medication data with patterns of compliance and medication data from a population of patients; and
outputting the at least one recommendation based on both the patient's pattern of compliance and medication data and the patterns of compliance and medication data from the population of patients.

39. The method of claim 26, wherein the treatment regimen is a standardized treatment regimen.

40. The method of claim 26, wherein the treatment regimen is an individual patient treatment regimen.

41. The method of claim 26, further comprising:

outputting a recommendation to a health care provider to enable the health care provider to determine target drug levels or benchmark drug levels.

42. The method of claim 26, further comprising:

comparing the amount glucose produced with a baseline glucose level and quantitatively determining an amount of the target agent present in the biological sample based on a difference between the baseline glucose level and the amount of glucose sensed.

43. A self-contained medication monitor comprising:

a housing body configured to contain medication;
a first detection device for detecting removal of medication from the self-contained medication monitor;
a second detection device having a target agent sensor for detecting a target agent in a biological sample from a user, wherein
the target agent sensor comprises: a solid support to which is attached a recognition molecule that specifically binds to the target agent in the presence of the target agent but not significantly to other agents; a substance that can be enzymatically converted to glucose; and an enzyme that can catalyze the conversion of the substance to glucose in the presence of the target agent;
a processor that produces medication usage data and medication data based on detected removal of the medication and an amount of glucose sensed and that outputs recommendations to the patient based on the medication usage data and medication data; and
a display device configured to display the recommendations to the patient.

44. The self-contained medication monitor according to claim 39, wherein

the processor is configured to output a recommendation to a health care provider to enable the health care provider to determine target drug levels or benchmark drug levels.

45. The self-contained medication monitor according to claim 39, wherein

the processor is configured to compare the amount glucose sensed with a baseline glucose level and quantitatively determine an amount of the target agent present in the biological sample based on a difference between the baseline glucose level and the amount of glucose sensed.
Patent History
Publication number: 20140278468
Type: Application
Filed: Mar 15, 2013
Publication Date: Sep 18, 2014
Applicant: I.D. THERAPEUTICS LLC (Salem, WI)
Inventors: Steven L. MAYER (Salem, WI), David C. KRAVITZ (Barrington Hills, IL), Tracey H. MAYER (Salem, WI)
Application Number: 13/837,809
Classifications
Current U.S. Class: Health Care Management (e.g., Record Management, Icda Billing) (705/2)
International Classification: G06F 19/00 (20060101);