专利摘要:
system for modeling the progression of a disease within a population the present invention provides an integrated health care surveillance system and monitoring system that provides real-time sampling, modeling, analysis and recommended interventions. the system can be used to monitor infectious and chronic diseases. when faced with an outbreak of an infectious disease agent, for example, influenza virus, the system can identify cases of assets through proactive sampling in high-risk locations, such as crowded schools and commercial areas. the system can notify competent authorities, for example, local, regional and national governments, when an event is detected, thus allowing proactive management of an eventual outbreak. the system also provides the best response for the deployment of scarce resources.
公开号:BR112012009196B1
申请号:R112012009196-4
申请日:2010-10-18
公开日:2021-03-30
发明作者:Seth G. Michelson;Daniel L. Young;Elizabeth A. Holmes;Ian Gibbons
申请人:Labrador Diagnostics Llc;
IPC主号:
专利说明:

Cross-reference
This deposit claims the benefit of U.S. provisional deposit 61 / 253,015, deposited on October 19, 2009, the contents of which are hereby incorporated by reference in their entirety. State of the art
An epidemic of infectious diseases, capable of spreading across a large region, for example, a continent or across the world, can be very costly for societies. Such incidences include influenza pandemics, smallpox, tuberculosis, human immunodeficiency virus (HIV) and Severe Acute Respiratory Syndrome (SARS). The World Bank estimated in 2008 that an influenza pandemic could cost $ 3 trillion and result in a drop of almost 5% in the world gross domestic product (GDP). The World Bank further estimated that more than 70 million people could die worldwide in a serious pandemic. Others estimated that a flu pandemic could cause an economic recession in the United States, costing the country at least $ 500 billion to $ 675 billion in the short term. In 2003, SARS disrupted trade, travel and work in the Asia-Pacific region and cost the region around US $ 40 billion. The SARS pandemic lasted six months, killing at least 1,000 of the 8,000 infected people in 25 countries. The city of Toronto, CA has been closed to air traffic for several weeks and has suffered significant financial losses.
In 2009, the spring flu season cost billions of dollars, even though it only lasted a few weeks. The 2009-2010 winter flu season is expected to start in late August and could run until April 2010. Even if operative vaccines are available, supplies are expected to be limited and will not be able to stop the flu for several months. Economic losses can be minimized if the flu can be contained through proactive screening that allows for effective antiviral administration and narrows quarantine targets.
The economic loss due to "avoidance behaviors" is even greater than the cost of treating flu victims. The cost includes reducing air travel, avoiding travel to infected destinations and reducing consumption of services, such as mass transportation, dining out, shopping, etc. According to the World Bank, if an epidemic of influenza approaches mortality rates of 2.5% similar to that of influenza918 - 19, avoidance behaviors would cost a region five times greater in mortality losses or absence from work. Summary of the invention
There is an urgent need for an infrastructure to mitigate the spread of infectious diseases such as influenza, when this occurs. The present invention addresses this need through an integrated system that provides real-time sampling, modeling, analysis and / or recommended interventions. The system can identify active cases in an outbreak through proactive sampling in high-risk locations, such as schools and busy commercial areas, and can allow sampling and quarantine of surrounding cases to help eradicate the epidemic. The system can also suggest an appropriate response to the deployment of scarce resources and predict the impact of such mitigation, both in terms of reducing mortality and morbidity and economic impact. In addition, the systems of the present invention can help the government to provide accurate, more reliable and timely information, which can reduce unnecessary avoidance behavior and save billions of dollars.
In one aspect, the present invention provides a system for modeling a disease progression within a population, comprising: a static database component comprising static data related to the disease and / or the population; a dynamic component of the database comprising dynamic data about the population and individual subjects, and a computer modeling component that is configured to model the data in the static component of the database and dynamic component of the database, thus modeling the disease within the population. The disease can be an infectious disease or a chronic disease. In some embodiments, the infectious disease agent or analyte thereof comprises an adenovirus, Bordella pertussis, Chlamydia pneumoiea, Chlamydia trachomatis, Cholera Toxin, Cholera Toxin β, Campylobacter jejuni, Cytomegalovirus, Diphtheria Toxin, Epstein-Barr -Barr EA, Epstein-Barr VCA, Helicobacter Pylori, Hepatitis B Virus Core (HBV), Hepatitis B Virus (HBV) envelope, Hepatitis B Virus Surface (HBV) (Ay), Hepatitis Virus Core C (HCV), Hepatitis C Virus (HCV) NS3, Hepatitis C Virus (HCV) NS4, Hepatitis C Virus (HCV) NS5, Hepatitis A, Hepatitis D, Hepatitis E Virus (HEV) orf2 3 KD, Virus Hepatitis E (HEV) orf2 6 KD, Hepatitis E (HEV) Virus orf3 3KD, Human immunosuppression virus (HIV) -1 p24, Human immunosuppression virus (HIV) -1 gp41, Human immunosuppression virus (HIV) - 1 gp120, Human papilloma virus (HPV), Herpes simplex virus HSV-1/2, Herpes simplex virus HSV-1 gD, Herpes simplex virus HSV-2 gG, Human T cell leukemia (HTLV) -1/2, Influenza A, Influenza A H3N2, Influenza B, Leishmania donovani, Lyme disease, Mumps, M. pneumoniae, M. tuberculosis, Parainfluenza 1, Parainfluenza 2, Parainfluenza 3, Virus Polio, Respiratory syncytial virus (RSV), rubella, Rubella, Streptolysin O, Tetanus toxin, T. pallidum 15 kd, T. pallidum p47, T. cruzi, Toxoplasma, or Varicella Zoster.
In other embodiments, the disease is an infectious disease comprising a microorganism, a microbe, a virus, bacteria, an archaeum, a protozoan, a protist, a fungus or a microscopic plant. The virus can comprise flu or HIV. The bacterium can comprise Mycobacterium tuberculosis. The protozoan can understand malaria.
In still other embodiments, the disease is a chronic disease or condition comprising diabetes, pre-diabetes, insulin resistance, metabolic disorder, obesity or cardiovascular disease.
The static database component of the invention can include information about individuals in the population. Information about individuals in the population may include one or more of age, race, sex, location, genetic factors, single nucleotide polymorphisms (SNPs), family history, history of illness or therapeutic history.
The static component of the database can also comprise information about the disease. Information about the disease may include one or more of virulence, contagiousness, mode of transmission, availability of treatment, availability of vaccine, rate of death, recovery time, cost of treatment, infectiousness, rate of spread, mutation rate and past outbreak.
In some embodiments, the data in the dynamic component of the database is updated in real time. In some embodiments, the data in the dynamic component of the database comprises an indication of the disease status of individuals in the population. The indication of an individual's state of illness can be determined by measuring a biomarker, a physiological parameter, or a combination of these.
When the disease accompanied by the invention is influenza, the biomarker (s) may include hemagglutinin and / or neuraminidase. Hemagglutinin can be selected from the group consisting of H1, H2, H3, H4, H5, H6, H7, H8, H9, H10, H11, H12, H13, H14, H15, and H16, and neuraminidase can be selected from group consisting of N1, N2, N3, N4 and N5. In some embodiments, hemagglutinin comprises H1 and neuraminidase comprises N1. In some embodiments, hemagglutinin comprises H5 and neuraminidase comprises N1.
The biomarker measured by the invention can be an antibody to the host. For example, the biomarker can be an IgM antibody, an IgG antibody or an IgA antibody against a disease marker.
In some embodiments, the biomarker comprises an inflammation marker. Such an inflammation marker may be a cytokine or C-reactive protein. The inflammation marker may also be 1L-1 □, IL-6, IL-8, IL-10, or TNFD.
In some embodiments, the biomarker is measured in a sample of the individual's body fluid. Exemplary body fluids include, without limitation, blood, plasma, serum, saliva, urine, feces, semen, mucus, lymph, saliva, or nasal lavage.
In some embodiments, the physiological parameter measured by the invention comprises one or more of body weight, temperature, heart rate, blood pressure, mobility, hydration, ECG, or the use of alcohol.
The biomarker or physiological parameter can be determined using a “point-of-care” device. Point-of-care devices can be implanted according to the instructions determined by the computer modeling component. The point-of-care device can perform, without limitation, one or more of the cartridge tests, real-time PCR, rapid antigen tests, viral culture and immunoassays. The point-of-care device can measure more than one biomarker with an accuracy greater than 30% and / or better accuracy than standard methods. In some embodiments, the system comprises a plurality of point-of-care devices. Point-of-care devices can be positioned in one or more of: a school, a workplace, a shopping center, a community center, a religious institution, a hospital, a health clinic, a mobile unit, or a house.
The point-of-care device may include a portable instrument. For example, the point-of-care device may include a portable cartridge. In some embodiments, the cartridge is configured to accept reagents for measuring biomarkers. Biomarkers can be measured according to a protocol communicated by the computer modeling component. In some embodiments, the cartridge is configured to measure a set of biomarkers from a plurality of body fluid samples.
The "point of care" device of the invention can include a graphical user interface configured for data entry.
In some embodiments, the “point of care” device is configured to communicate the measurements of physiological parameters or biomarkers to the computer modeling component. Communication can include "wireless" communication, wired communication, or a combination of the above. “Wireless” communication includes, without limitation, WiFi, Bluetooth, Zigbee, cellular, satellite, and / or WWAN. Communication can also be carried out via secure internet communication. In some embodiments, the “point of care” device is configured to perform two-way communications with the computer modeling component.
In some embodiments of the system of the invention, the modeling results are updated in real time when updated dynamic data becomes available, for example, after the computer modeling component receives the updated information from a point of care device ”.
The computer modeling component can be configured to present the modeling results to one or more of the healthcare professionals, government agencies and individual humans. The computer modeling component can also be configured to predict one or more courses of action based on the modeling results. The one or more courses of action are classified according to a classification parameter, including, without limitation, classification by financial considerations, number of affected individuals, year of quality adjusted life (QALY), and / or years of quality life adjusted (QALY) per economic unit of cost.
The one or more courses of action comprise a strategy to control the spread of the disease. The strategy to control the spread of the disease may include one or more of: home quarantine, individual quarantine, geographic quarantine, social detachment, hospitalization, school closure, workplace closure, travel restrictions, public transport closure, treatment or therapeutic intervention, prophylactic treatment or intervention, vaccination, provision of protective clothing, provision of masks and additional point-of-care tests. The strategy to control the spread of the disease may also include one or more of: counseling individuals at risk or affected for behavior modification, biomarker and / or repeated physiological measurements and reward for the individual. Furthermore, the strategy to control the spread of the disease may include one or more of the recommendations of: patient screening, resource management, the effectiveness rate of each strategy, the costs of each strategy, the return on investment for each strategy. The strategy to control the spread of the disease can be one or more of: target prophylaxis, blanket prophylaxis, antibiotic-oriented prophylaxis, antibiotic blanket prophylaxis, antiviral target prophylaxis, antiviral blanket prophylaxis, specific vaccination and blanket vaccination. Targeted prophylaxis or vaccination can be aimed at the prophylaxis or vaccination of children between 1-4 years of age, children between 5-14 years of age, pregnant women, young adults between 15-30 years of age, first-line medical workers response, individuals identified as having a high risk of mortality, or geriatric individuals.
In some embodiments of the invention, the computer modeling component is configured to estimate a surveillance strategy based on the modeling results. The surveillance strategy may include determining the disease status of an individual or group of people using a point of care device. The surveillance strategy can be updated when a sick individual is detected. In some embodiments, the updated strategy comprises one or more of the tests of a group comprising the sick individual, testing a school comprising the sick individual, and testing a workplace comprising the sick individual. The update strategy can also be one or more of: prophylaxis, quarantine or hospitalization.
In some embodiments, the computer modeling component comprises a graphical interface for displaying the modeling results to a user.
The computer modeling component can include a plurality of non-linear differential equations and / or a plurality of parameters. In some embodiments, the computer modeling component comprises a learning machine that updates the plurality of parameters when static data and / or dynamic data are updated.
The data model can be configured to include a plurality of states. In some embodiments, the plurality of states comprises one or more of: susceptible individuals, individuals exposed early, individuals exposed late, the first infected individuals, individuals infected late, individuals recovered, individuals who died due to infection and / or complications associates, asymptomatic individuals, individuals who have received therapeutic treatment, individuals who receive therapeutic treatment and are quarantined, individuals prophylactically treated, vaccinated individuals, people protected due to vaccination, the first infected individuals who are hospitalized, late infected individuals who are hospitalized, susceptible individuals who are at home in quarantine, individuals exposed early who are in the final home, individuals exposed late who are quarantined at home, the first infected individuals who are quarantined at home, individuals infected late who are quarantined at home, asymptomatic individuals who are at home in quarantine, susceptible individuals in quarantine throughout the neighborhood, individuals exposed early in quarantine throughout the neighborhood, people exposed late in quarantine throughout the neighborhood, the first infected individuals in quarantine throughout the neighborhood, the infected individuals in quarantine delay throughout the neighborhood, asymptomatic individuals quarantined throughout the neighborhood, the amount of therapeutic doses available, the amount of antivirals and / or antibiotics available to the target population, individuals at home quarantine that are vaccinated at home, quarantined individuals that are protected due to vaccination, individuals that have recovered in quarantine at home, susceptible individuals destined for mitigation policies for action, individuals exposed early destined for mitigation policies for action, individuals exposed late intended by mitigation policies for action o, asymptomatic individuals destined for action mitigation policies, first infected individuals destined for action mitigation policies, late infected individuals destined for action mitigation policies, prophylactically treated individuals destined for action mitigation policies , vaccinated individuals destined for action mitigation policies, protected individuals destined for action mitigation policies, recovered individuals destined for action mitigation policies, susceptible individuals destined for therapeutic treatment, individuals exposed early destined for therapeutic treatment, individuals exposed late for therapeutic treatment, asymptomatic individuals for therapeutic treatment, first infected individuals for therapeutic treatment, infected individuals with delay for therapeutic treatment, individuals susceptible wastes destined for surveillance, individuals exposed early for surveillance, individuals exposed late for surveillance, asymptomatic individuals intended for surveillance, the first infected individuals intended for surveillance, o infected individuals delayed for surveillance, prophylactic individuals intended for surveillance surveillance, vaccinated individuals for surveillance, protected individuals for surveillance, susceptible individuals in the entire quarantine neighborhood targeted by mitigation policies for action, individuals initially exposed in the entire quarantine neighborhood targeted by mitigation policies for action , individuals exposed late in every quarantine neighborhood destined for action mitigation policies, asymptomatic individuals in the entire quarantine neighborhood destined for action mitigation policies, the first infected individuals in every neighborhood quarantine des based on mitigation policies for action, infected individuals delayed throughout the quarantine neighborhood designated by mitigation policies for action, individuals treated with prophylaxis in the entire quarantined neighborhood designated by mitigation policies for action, the cumulative number of therapeutic doses cumulative number of antivirals and / or antibiotics administered, cumulative number of asymptomatic individuals quarantined at home, cumulative number of symptomatic individuals quarantined at home, total cumulative number of infected individuals, cumulative number of infected individuals who are not quarantined , the cumulative number of individuals infected with some measures taken, the cumulative number of hospitalized individuals and cumulative number of deaths.
In another aspect, the present invention provides a system for controlling the spread of influenza within a population, which comprises: a static database component comprising static data related to influenza and / or the population; a dynamic component of the database comprising dynamic population data; and a computer modeling component that is configured to model the data in the static component of the database and dynamic component of the database, thus modeling the incidence of influenza within the population.
In yet another aspect, the present invention provides a system for controlling the spread of the human immunodeficiency virus (HIV) within a population, comprising: a static database component comprising static data related to HIV and / or the population ; a dynamic database component comprising dynamic population data; a computer modeling component that is configured to model the data in the static and dynamic components of the database, thus modeling the incidence of HIV within the population.
In yet another aspect, the present invention provides a system for controlling the spread of hepatitis within a population, comprising: a static database component comprising static data related to hepatitis and / or the population; a dynamic component of the database comprising dynamic data about the population: and a computer modeling component that is configured to model the data in the static components of the database and dynamic component of the database, thus modeling the incidence of hepatitis within the population.
In one aspect, the present invention provides a system for controlling the spread of diabetes within a population, which comprises: a static database component comprising static data related to diabetes and / or the population; a dynamic component of the database comprising dynamic population data; and a computer modeling component that is configured to model the static and dynamic components in the database, thus modeling the incidence of diabetes within the population. Incorporation by reference
All publications, patents and patent applications mentioned in this description are hereby incorporated by reference to the same extent as if each individual publication, patent filing or patent is specifically and individually indicated to be incorporated by reference. Brief description of the drawings
The innovative features of the invention are presented with particularity in the appended claims. A better understanding of the characteristics and advantages of the present invention will be obtained by reference to the following detailed description, which establishes illustrative embodiments, in which the principles of the invention are used, and the accompanying drawings, in which: Figure 1 illustrates a representation simplified model.
Figure 2 illustrates a model representation taking into account various states and transition between states.
Figure 3 illustrates an assay for H1N1 antigen using complex sandwiches in four different configurations.
Figure 4A illustrates an assay for host anti-virus antibodies. The Figure illustrates a peak recovery assay for host anti-H1N1 antibodies. A version is shown using a-H1 / a-N configuration. Figure 4B illustrates direct assays for a-H1N1 antibodies illustrating sandwich complexes.
Figure 5 illustrates an example of a device that can be used in the present invention. Said device comprises test units, reagent units and other modular components.
Figure 6 illustrates sectional views on two sides of the example device that can be used in the present invention. The exemplary device comprises cavities in said device housing shaped to accommodate a test unit, a reagent unit and a test tip.
Figure 7A demonstrates an example of a test unit comprising a small tip or tubular formation. Figure 7B demonstrates an example of a sampling tip as described here.
Figures 8A and 8B illustrate two examples of reagent units comprising a cup.
Figure 9 illustrates a thin film, for example, contamination, inside the tip when one liquid is expelled and another liquid is aspirated.
Figure 10 demonstrates an example of a system comprising a fluid transfer device and device.
Figure 11 illustrates an exemplary system of the invention comprising a heating block for temperature control and a detector.
Figure 12 demonstrates an exemplary system where a patient delivers blood to a device and then the device is inserted into a reader.
Figure 13 illustrates the process flow of building a system to assess a patient's medical condition.
Figures 14A through 14E demonstrate an example of a plasma separation method where an entire blood sample was aspirated into a sample tip and a magnetic reagent was mixed and suspended with the sample, so a magnetic field is applied to the entire mixture. blood sample and magnetic reagent. The plasma sample separated from the blood can then be distributed into the well of a device.
Figure 15 demonstrates an exemplary control assay method as described herein comprising a known amount of control analyte.
Figure 16 illustrates an exemplary embodiment of a health shield user interface.
Figure 17 illustrates another exemplary embodiment of a health shield user interface.
Figure 18 illustrates a simulation of the La Gloria 2009 epidemic with and without Health Shield mitigation policies.
Figure 19 illustrates diabetes risk prediction visualization.
Figure 20A illustrates the detection of H1N1 viral particles using a point of care device. Figure 20B illustrates the detection of H1N1 viral particles using a “point of care” device in clinical samples.
Figure 21 illustrates the detection of host antibodies using a point of care device.
Figure 22A illustrates the detection of host antibodies using a point of care device.
Figure 22B illustrates the host's dynamic antibody detection range using a point care device.
Figure 23 illustrates the detection of human cytokine IL-6 using a point of care device.
Figure 24 illustrates the detection of C-protein and C-reactive protein (CRP) using a point of care device in a patient undergoing chemotherapy.
Figure 25 illustrates the detection of glucagon-like peptides 1 (GLP-1) using a point of care device.
Figure 26 illustrates the detection of C-peptide, an insulin precursor, using a point of care device.
Figure 27 illustrates the detection of C peptide using a cartridge point of care device compared to the reference detection system (Linco).
Figure 28A illustrates the measurement of GLP-1 in three human patients after feeding. Figure 28B illustrates the measurement of C-peptide over the same experiment.
Figure 29 illustrates a calibration curve correlating a test unit and reagent unit to conduct a test for VEGFR2.
Figure 30 illustrates CRP concentration as a function of the test signal (photon counts) and the data adapted from a fifth degree polynomial function to generate a calibration function.
Figure 31 shows an adjustment obtained between a model and the values of the parameters Smax, CO.5 and D as described here.
Figure 32 shows the data according to the dilution used to obtain the final concentration in a test tip.
Figure 33 illustrates the normalized assay response (B / Bmax) as a function of the normalized log concentration (C / C0.5) for relative dilutions: 1: 1 (continuous line), 5: 1 (dashed line), and 25: 1 (dotted line).
Figures 34 and 35 illustrate an example similar to Figure 33 in different normalized concentrations.
Figure 36A shows a peak in septic IL-6 individuals. Figure 36B shows a decline in protein C in septic individuals.
Figure 37 shows an increase in ll-6 and TNF-D (right panel) in an individual as the H1N1 influenza burden increases in the patient (left panel) Detailed description of the invention
In one embodiment, the present invention provides an integrated health data capture, analysis and pandemic mitigation solution, referred to herein as the Health Shield (ES). ES can be used for infection caused by the flu virus and other pathogens prone to endemic or pandemic. Flu outbreaks cost billions of dollars and cannot currently be completely contained by vaccination. Economic losses can be minimized if the flu can be contained through proactive screening that allows for the administration of effective antiviral agents and narrows quarantine targets. Based on epidemic models, activating the ES of the invention can reduce the spread of the virus, for example, by at least 50%, through proactive sampling and containment. ES can also reduce unnecessary escape behavior by tracking the spread of the virus in real time. When desired, test results can be relayed wirelessly to an ES operational software server. Thus, competent authorities (for example, local, regional and national governments) can be notified with alerts when an event is detected, thus allowing proactive management of an eventual outbreak.
In other embodiments, the Health Shield infrastructure provides industrial and commercial strategies as "safe zones", which allow economically important activities to continue. As a result, fewer workers will be infected with the virus and schools and businesses will be less disturbed. Pandemic mitigation strategies will maintain productivity to boost economic growth and prevent panic-driven actions. The system can comprise an integrated sampling and set of modeling technology incorporated in a real-time computer infrastructure. The ability to sample, model and learn from data as it is acquired longitudinally allows the development of an optimal strategy for the care and treatment of the disease, both on an individual and population basis. Custom applications can be built for many diseases and therapeutic areas. The ES infrastructure can also be used to protect a region from a broad spectrum of threats in addition to infectious diseases, including chronic diseases and threats from bioterrorism. 1. Health Shield Infrastructure
The health shield provides a system to contain the spread of infectious diseases through integrated, automated and real-time sampling, modeling, analysis and recommended interventions. For example, ES can identify active cases in an outbreak (through proactive sampling in high-risk locations, such as crowded schools or commercial areas) and target sampling and defensive measures, for example, quarantine, from surrounding cases to mitigate or eradicate the outbreak. ES algorithms characterize the spread of the epidemic in a similar way to the case of a forest fire, where the THS models mitigation policy aims to eradicate "hot spots" before the "fire" can take over and spread and / or create a cut-off. fire around a critical hot spot.
In some embodiments, the ES comprises two technological components - a Field System (SC) and an Operating System (SO) - that can be adapted to manage chronic diseases to improve health outcomes and decrease health costs. (a) Field System (SC)
The components of the ES field system can be deployed at various points of care, including, without limitation, a clinic, a community location (for example, school, community center), a hospital, a doctor's office or at the home of a individual. SC can also use any number of platforms to monitor the disease, for example, immunoassays, PCR assays, real-time PCR, microorganism plates, etc. The SC also includes standard medical equipment, for example, scales for determining weight, blood pressure devices, thermometers for measuring temperature, rulers for measuring height, etc. In some embodiments, SC devices comprise personalized, single-use, portable cartridges, as described herein. The SC collects relevant data in the field and transmits said data to the operating system.
In some modalities, the field system comprises a measuring device intended to be implanted in an area to be monitored. In some embodiments, the SC analyzes samples of body fluids, for example, blood from a finger prick, in real time. The system analyzes body fluids for evidence of infection or disease by detecting, for example, markers for a pathogen, nucleic acids, proteins, glycoproteins, lipids, or a combination of these indicative of a disease condition. In some embodiments, SC simultaneously measures multiple markers, including one or more of the selected antigens or pathogens, antibodies to pathogenic organisms, cell or intracellular surface proteins or glycoproteins and cytokines indicative of the response of an individual infected with a given pathogen, (for example, a viral strain or other microorganism). The system can also collect environmental, demographic, personal and physiological information (for example, temperature, blood pressure). In some modalities, such information is collected through a graphical touch interface. Individualized content can be analyzed by a remote system to facilitate real-time mitigation strategies.
In some embodiments, SC includes cartridges to perform tests on body fluids. The devices include without limitation devices of no significant risk and the assays can be validated under appropriate guidelines, for example, those provided by the United States Federal Drug Administration (FDA) and / or International Harmonization Conference (ICH). Cartridges used by the present invention are described in US 11 / 389,409 entitled “POINT-OF-CARE-FLUIDIC SYSTEMS AND USES THEREOF,” American patent filing US11 / 746,535 entitled “REAL-TIME DETECTION OF INFLUENZA VIRUS,” American patent filing US. 12 / 244,723 entitled “MODULAR POINT-OF-CARE DEVICES, SYSTEMS, AND USES THEREOF” which are described in more detail below. The measurement systems can be self-sufficient and little or no extra material is needed to operate them. In some embodiments, the only requirement for a SC system is a power source for the instruments. In other embodiments, the power source is provided with the SC in the form of a battery, solar generator, or other portable power source. Cartridges can be preloaded with the desired assays and require little or no preparation before use. For example, some or all of the test components can be stored in a refrigerator (for example, at about 4 ° C) prior to implantation.
The SC platform can run any appropriate assay that is currently performed on the conventional laboratory infrastructure. New tests can be downloaded quickly and fully validated. In some embodiments, assays that are entirely new to the SC system can be customized and validated in less than about three months, two months, one month, 3 weeks, 2 weeks or less than about 1 week. In some embodiments, tests performed on SC systems are validated under FDA ICH guidelines.
Field systems can be placed at any desired point of care, for example, a suspected or known area at risk of infection or disease. Point of care (POCT) tests are defined by a test system close to the patient. Examples of "point of care" may include, but are not limited to, home, clinic, schools, or shopping centers. In some modalities, the SC is deployed in mobile units. Thus, it must be understood that medical experts are not necessarily required for the test. To enable this, the SC can be designed to be simple to use and offer all indications for use in a simple user interface with a touch screen. In some embodiments, the systems are designed for individuals not computer savvy to test themselves in their own homes. In this scenario, data can be sent to a remote system, for example, the operating system as described below, where public officials or others monitoring the tests can learn from the results of the positive tests. In some embodiments, testing and loading of data / analysis is carried out in real time, so that containment measures can be started immediately.
In some modalities, the systems are implemented in public places. If desired, standard public health officials can be trained to take the test. In some embodiments, the systems are designed so that the total training time is minimized at a given location. For example, the current deployment demonstrates that training should not require more than half an hour of time per location, although supplementary and advanced training can be carried out as appropriate. In some embodiments, trained individuals can in turn train others in the use of the systems. The SC can be used successfully at home by patients who have no medical training - as the test was designed to be fully automated and uses a graphical touch screen interface on the instrument to guide users through the testing process. In some embodiments, the only necessary steps for a user are: 1) placing a sample inside the cartridge, for example, sputum or a finger prick, which can be performed by the user himself using a disposable lancet for single use, such as used in diabetes management for glucose monitoring, and 2) insert the cartridge into the monitoring instrument, as described in more detail below.
Non-limiting custom cartridge devices for use in the SC of the invention are described in US patent filing US 11 / 389,409 entitled “POINT-OF-CARE-FLUIDIC SYSTEMS AND USES THEREOF,” US patent filing US 11 / 746,535 entitled “REAL- TIME DETECTION OF INFLUENZA VIRUS, ”US patent filing US 12 / 244,723 entitled“ MODULAR POINT-OF-CARE DEVICES, SYSTEMS, AND USES THEREOF. ” Such devices are further described below. (b) Operating System (OS)
The data collected from each SC device can be safely transmitted to the operating system in real time via the network connection, for example, over a broadband, wireless, satellite or cellular network. A person skilled in the art will appreciate that network communications often include multiple hops, for example, an SC device can connect to a wireless local area network (WLAN) that is connected to the World Wide Web via fixed-band phones. wide.
In some embodiments, the operating system includes one or more servers as they are known in the art and commercially available. Such servers can provide load balancing, task management and backup capability in the event of failure of one or more of the servers or other system components, to improve OS availability. A server can also be implemented in a distributed network of storage and processing units, as known in the art, in which the data processing according to the present invention resides on workstations, such as computers. An OS component server can include a database and system processor. A database can reside on the server, or it can reside on another server system that is accessible to the server. Since the information in a database can contain sensitive information, a security system can be implemented to prevent unauthorized users from gaining access to the database.
In some embodiments, the operating system comprises a data engine that imports data from a desired source to provide guidance for mitigating the epidemic or pandemic. The operating system can translate the source data into a standardized format for analysis. In some embodiments, the data engine is self-learning and dynamically models a plurality of data sets integrated in real time. This OS modeling approach offers several benefits. For example, models can be trained to perform a variety of calculations, including, but not limited to: 1) predicting outcomes for individuals and populations; 2) consider the effectiveness of the proposed intervention strategies for individuals and populations, and 3) quantify the socioeconomic effect of the recommended interventions. In some embodiments, the operating system is available to remote users through a remote interface. For example, users can access the operating system through a secure online web portal or similar.
The OS software portal incorporates automatic modeling in a system that is constantly learning from each new data point that is transmitted to the software portal. The system thus becomes more and more predictable over time. In some embodiments, Monte Carlo modeling approaches are used. Monte Carlo approaches depend on repeated random sampling to calculate the results. Monte Carlo simulation considers random sampling of probability distribution functions as an input model to produce hundreds or thousands of possible results instead of some discrete scenarios. The results provide probabilities of different results occurring. In some embodiments, the solution and readaptation / refining of the model's parameter sets is achieved through the use of reverse research and integrated parameter estimation techniques. See, for example, Sheela, 1979-COMPUTER METHODS IN APPLIED MECHANICS AND ENGINEERING 19 (1979) 99-106; Moles, et al. 2003 - Genome Res. 2003 13: 2467-2474; Rodriguez-Fernandez, et al. BMC Bioinformatics 2006, 7: 483-500; Barthelmann, et al. 2000 - Advances in Computational Mathematics 12; 273-288.
There is a rich literature on the modeling and simulation of epidemiological data. The basis of the McKendrick model is a stochastic process (birth process) that produces a series of differential equations that can be parameterized, explored and, eventually, optimized in relation to the control and spread of the disease. A relatively simple analysis of the process is given by Chiang, CL 1978. An Introduction to Stochastic Processes and Their Applications. Robert E. Kreiger Publishing Co, Inc. Huntington, NY. p517, Once the process is established in a stochastic space and properly parameterized, explicit expressions for the moments of population and or probabilities of extinction can be derived. If the process is simple, these expressions can be modeled and estimated, either in closed form or numerically.
If the populations are large enough that the stochastic variation is small compared to the size of the global population and the dynamics of systems, the spread and growth of a disease state can be modeled using systems of differential equations. For example, a simple SIR model (Susceptible, Infected, removed) of SARS was explored by Choi and Pak, J Epidemiol Community Health. 2003 Oct; 57 (10): 831-5. More complex models taking into account exposure, the SEIR model was explored by d'Onofrio, Mathematical Biosciences 179 (2002) 57-72, especially regarding the optimization of vaccination strategies. For influenza in particular, Stilianakis, et al., J Infect Dis. 1998 Apr; 177 (4): 863-73, looked at particular aspects of drug resistance in the growth and spread of disease. Other aspects of disease modeling, including propagation and diffusion kinetics (FitzGibbon, et al., MATHEMATICAL BIOSCIENCES 128: 131-155 (1995)), mathematical and numerical stability (Dwyer, et al., The American Naturalist, 150 ( 6): 685-707; Inaba, J. Math. Biol. (1990) 28: 411-434).
Simulation is a valuable tool in solving these complex systems. There are many models that take the simulation solution. See, for example, Longini, et al., 1984, Int J. Epidemiology. 13: 496-501; O'Neill, 2002. A Tutorial Introduction to Bayesian Inference for Stochastic Models Using Markov Chain Monte Carlo Methods. Math Biosci. 180: 103-114; Gibson, G.J. 1997. Investigating mechanisms of Spatiotemporal Epidemic Spread Using Stochastic Models. Am Phytopathological Society. 87: 139-146. In particular, see Timpka, et al. (2005) AMIA 2005 Symposium Proceedings. 729-733, with regards to simulating influenza. In some embodiments, the epidemic growth and proliferation model and incumbent screening and containment strategies are incorporated into a cost-effective health economics model. See, for example, Brandeau, et al. Journal of Health Economics 22 (2003) 575-598.
An example representation of a simplified model according to the invention is shown in Figure 1. The model can be configured to describe the spread, surveillance and reduction, with its cost-effectiveness for political management of the epidemic / pandemic. Briefly, a risk population is segmented into several states or conditions (represented by the circles in the Figure), with the flow components between each state modified by a variety of configurable parameters, including, but not limited to, the rate of infection, the means and granularity of the surveillance mechanism, and political decision at hand. To assist the political director in the decision-making process, both own and social costs, for example, QALYs, can be calculated by the model and displayed to the political director.
The model illustrated in Figure 1 includes a system of ordinary non-linear deterministic differential equations. Each node (or state) represents a population of individuals with similar phenotypic characteristics and disease, such as their state of infectivity. Several states may also represent individuals in different locations, such as schools, workplaces, during hospitalization, isolated quarantine, or home quarantine. A plurality of age groups, for example, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50 or more age groups are represented by a modular structure, thus allowing the specification of specific age characteristics. In some embodiments, the model takes age into account in a continuum, as opposed to within discrete groups. The arrows shown connecting the nodes in the Figure indicate the flow from one state to another. As described here, the model parameters come from a variety of sources, for example, literature reports, patient data, previous outbreaks, and can be estimated based on data, as desired. The model projections capture a range of possibilities based on the quantified uncertainties. As the model forecasts are implemented, the parameters can be continuously adjusted in real time according to the actual results in the field. For example, the effectiveness of the various mitigation policies can be re-evaluated and adjusted according to real-world results applied to current and specific affected populations.
Those skilled in the art will appreciate that the model shown in Figure 1 can be expanded to take into account any number of relevant states and parameters. Figure 2 shows a larger model representation. Each circle represents a class of individual and each arrow represents a transition from one state to the next. Transitions from one state to another can take into account changes in natural causes, or interventions, for example, treatment, therapeutic. The model can also take into account transitions that do not involve a state of illness, for example, changing social interaction with different groups. For example, an individual in quarantine may change from involvement in the community to involvement with a limited number of individuals, for example, limited contact with health professionals or other caregivers. The model parameters at the beginning of an epidemic can be derived from data from the nearest applicable previous disease outbreak with the closest demographic data and type of location (for example, a city, a rural area). The model can be continually improved through the application of data collected in the context of the epidemic currently underway! to become progressively better.
At the top of Figure 2, a flow from left to right is highlighted by the line Pi, Si, E1i, E2i, 11 i, i2i, Ri, and Di. These states represent a model of disease spread that comprises the states of prophylactically treated, for example, with antivirals (Pi), susceptible individuals (Si), individuals exposed early (E1i), individuals exposed (E2i late) , the first symptomatic individuals infected (11 i), symptomatic individuals infected late (i2i), recovered and, therefore, potentially immune individuals (IR), and the deceased (Di). An individual can transition from E2i state to Ai state, which represents the asymptomatic infectious subpopulation in the community in question. An individual can also transition to the Vi state, which represents vaccination. From the vaccinated state, an individual can make the transition, either to a clean and immunological state, Ci, or to the ineffective and exposed state, E1i. By taking into account any number of individuals, i, the model can capture a representation of the population spreading the epidemic. The delay criteria, E2i and I2I, accommodate the time-dependent spread of the disease. The segment above the disease spread model represents the impact of a treatment policy and its effects on population well-being and spread of the disease, while the segment below the spread of the specific disease represents a quarantine mitigation strategy. The model integrates a user-defined surveillance strategy and a user-defined mitigation strategy with a cost-effectiveness matrix to assist in decision making. In some embodiments, the model takes into account sub-optimal disease attenuation. For example, even when a hot spot of disease development has been located, there may be logistical delays in obtaining therapeutic agents for the area and in implementing quarantine. These delays can allow the epidemic to progress without mitigation. The model can take sub-optimal mitigation into account.
The model equations form a system of ordinary differential equations (ODE) with flow coefficients appropriately parameterized, as defined by the arrows in Figure 2. The basic shape of the model is given by the ODE vector: dX / dt = f (X, t ) where X is a scaled vector and the function f (x, t) is represented by an array of mixing parameters and functional interactions, as defined in the Figure. In the model in the Figure, there are more than 80 dimensions for the scaled vector. One skilled in the art will appreciate that the format and components of the matrix for the function f are derivable from Figure 2 and the explanation here.
The equation adjustments shown above are duplicated for each of a variety of age groups, as described herein. Consider an example with seven age groups. In the example, the seven-cluster conglomerate model is replicated for each geopolitical region in a given geographic region. The model can then be generalized to take into account the wider spread of the disease in a larger region. For example, by parameterizing the mix matrices and resources / cost tables, it is possible to explain national and international surveillance strategies and mitigation of interregional trips.
A variety of states modeled by the SO and shown in figures 1 and 2 are shown in table 1: Table 1: Description of states and nomenclature for the states used to describe the outbreak



The model of the invention can be configured to take into account many characteristics of the individuals, populations and disease to be monitored. In some embodiments, the strength of infection is taken into account in the model. The strength of infection, also called the rate of transmission, refers to the rate at which existing infected individuals transmit the disease to susceptible individuals. In some embodiments, each infectious individual is given two attributes: an age group j, based on the age of the individual, and a mixing group k, based on the individual's mixing pattern in society. Mixing patterns include, without limitation, mixing freely with others in society, for example, at school or at work, reducing mixing on sick days, etc. The force of infection exerted on the population of age group i by all populations of age groups j can be calculated as follows: A. ^ ∑∑JΔ ^ + CI- ^
Where, β is Transmission rate (per day per infectious individual per susceptible individual) θ is Parameters defining mixing randomness between groups of different ages: if θ = 1 the interactions are perfectly associative, if θ = 0, the interactions are perfectly random pi is Relative susceptibility of individuals in the age group / (pj is Relative infectivity of infectious individuals in the age group j Δ *. is a weight factor that takes into account differences in the relative extent of interactions potentially causing transmission between individuals in the group age ie those of age groups j and mixing groups k Ik is The number of infectious individuals in the age group j Nk is Total number of individuals in the age group j and mixing group k in the population Nt is Total number of individuals of all age groups in the population In the infection strength equation, the interaction weights Δk .. are calculated based on 1. the time spent by an individual in the age group i in the company of i n Individuals in the age group j and mixing group k in different locations, such as work, school, home, etc. 2. the number of individuals in the age group j and the mixing group k who come into contact with individuals in the age group i potentially causing transmission.
From the above parameters, pj} (pjt Δ *, Ikj} Nk can change dynamically over time as a result of the evolution of the epidemic, the imposition of mitigation policies, or both.
The SO model may include a series of mitigation policies that drive direct medical decision when faced with an outbreak. These policies can be modeled for each particular location, for example, geographic and disease configuration or infectious agent, to take better advantage of available resources. Each policy can be enforced with a realistic compliance / effectiveness that can be estimated from historical data. The io model can predict the results of implementing different mitigation policies, thus providing suitable individuals with the suggested response. Examples of non-limiting mitigation conditions are listed in Table 2: Table 2: mitigation policies represented in the model


In addition to mitigation policies, the SO model can incorporate the results obtained in the field during the execution of surveillance with a variety of different technologies. These include the cartridge systems described here, rapid antigen test, immunofluorescence, immunotests, real-time PCR, 5 viral culture test, physiological measurements, urine and blood count, etc. The model includes the representation of the sensitivity and specificity of each test for samples of both asymptomatic and symptomatic individuals. In addition, the turn around time for the different tests can be included in the model. io Depending on each particular system, the various forms of surveillance strategies can be included in the model. In one embodiment, surveillance comprises testing individuals for testing voluntarily. Surveillance can also be carried out for the population of groups that include, but are not limited to, the following: • Children between 1-4 years of age • Children between 5-14 years of age • Pregnant women • Young adults between 15 -30 years of age • Physicians of first line of response workers • Individuals identified for high risk of mortality • Geriatrics • Individuals of middle age between 30-60 years of age
Each of these population groups can be tested using any of the test methods or their combinations. Different proportions of asymptomatic individuals and symptomatic individuals for the voluntary test can also be accounted for in the model.
In another embodiment, surveillance includes testing based on the implementation of a surveillance policy, as defined by the end user. The catalog of surveillance policies captured by the model includes, without limitation, the following: • Household Surveillance: Test of the whole family based on the confirmed case index • School surveillance: test of schoolchildren based on the confirmed case index • local surveillance work: employee test based on the confirmed index case.
For confirmed cases identified as a result of surveillance tests, appropriate quarantine measures, prophylaxis or hospitalization can be taken
In some embodiments, the SC allows an automated analysis to be made using these methodologies for the selection, parameterization, and / or the exploration of an epidemic model suitable to implement the optimal screening and containment strategy. The model can be modified according to a cost-effective health-saving model. In some embodiments, the model is configured to predict the spread of an infectious pathogen in a heterogeneous human population. The models can take into account regional demographic data and individual risk factors. As described in more detail below, in one embodiment, the model allows for the assessment of health care mitigation policies, including, without limitation: a) strategic surveillance / testing; b) hospitalization, isolation of origin, and quarantine policies; c) prophylactic vaccination and treatment policies, for example, anti-viral therapy, and d) measures of social distance, such as school and work closures.
In addition to dynamics of infectious outbreaks, the model can provide cost assessment as well as quality-of-life-adjusted (QALY) assessment unless comparing alternative mitigation approaches. The model can be configured to take into account non-economic cost measures. The model can be configured to adjust for the cost associated with different errors, based on economic cost, thunderstorm costs, or other factors, in order to minimize the cost of errors made by a model. For example, the model may attribute a high cost to misdiagnosing an infected individual so that mitigation strategies are not put in place. The model could then adjust to favor avoiding such errors. Likewise, a diagnosis of a chronic condition can cost less than the individual can be tested again before the disease has progressed too far. In the case of an epidemic, the predictions may refer not only to the case of an individual, but to populations of people in different regions. Based on large demographic data sets, the SC analytical system can be configured to predict optimized risks and costs for treatment and trial delivery. For example, locations with lower expected risk can be sampled less than locations with higher expected risk.
The OS has built-in actions that are triggered when certain events are detected. For example, alerts can be sent to government officials when an infected individual is detected. Rules can be set to automatically notify a clinician by email, phone or fax, when a case is detected. The detected individual and contacts, for example, family members, co-workers, or anyone who has had contact with the individual in the past few days, weeks, months or years, can also be notified. The rules that trigger the action can be customized before deployment or during a follow-up period, depending on the needs of the situation.
The operating system models also perform sanity and outlier control of the data received from the FS. In some embodiments, actions are taken when variability or noise is identified in the data. In some embodiments, an assay for an individual is repeated when outliers are detected.
In some modalities, minimal models can predict results for individuals and populations. In some modalities, the prediction models correspond - in response to infection, the ideal treatment regimen for an individual or a population, and projected the spread of the virus - to the actual historical data, for example, data from the spring flu season. In some modalities, the models consider the effectiveness of the proposed intervention strategies for individuals and populations, including the use of preventive antiviral therapies, anti-viral reactive therapies, quarantine, hospitalization, targeted closures and establishment of "safe zones" in key hotels , restaurants, schools, factories and other locations. Models can also quantify the socio-economic effect (out-of-pocket expenses, lives saved, lost days of productivity, etc.) that the recommended interventions would have at the time of each case.
In some modalities, Field Systems and Operating Systems are also customized to provide solutions for various configurations in which the systems can improve results and reduce the cost of service. For example, FS and OS can provide health monitoring solutions for pharmaceutical and biotechnology companies and consumers. II. Deployment of the Health Shield
[00112] In some embodiments, Health Protection has a fully integrated Health Record of diagnosis / patient / electronic medical record platform. Field implanted system devices can be configured to be portable and therefore can be deployed at a variety of care points, including, without limitation, a clinic, a community website (eg, school, community center), a hospital, a doctor's office or an individual's home. As described here, the portable FS devices can be configured to connect wirelessly to a network, requiring only an optional cable for power. In some embodiments, the network connection is made to a portal, where the test data is sent in real time. FS systems can be deployed in urban environments close to care centers and the same devices can, for example, be deployed in remote environments, even when patients live long distances from nearby medical clinics.
The performance of FS tests will vary from test to test, but all tests are developed with a high precision objective, for example, through high specificity and sensitivity. In some embodiments, the specificity is greater than about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94% , 95%, 96%, 97%, 98% or greater than about 99%. In some embodiments, specificity approaches 100%. In some embodiments, the sensitivity is greater than about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94% , 95%, 96%, 97%, 98% or greater than about 99%. In some embodiments, the sensitivity approaches 100%. The exact performance of an assay can depend on a number of factors, including but not limited to the performance of the marker to be detected, the user's ability and assay performance inherent in the device. In some embodiments, FS systems are designed to be highly user friendly and require minimal skill to operate effectively. The time required for the performance of the test will also vary according to the use case for implantation. Each system is fully customized to better achieve the deployment goals for which all specifications are defined accordingly. In some embodiments, the tests are performed in a matter of minutes, for example, less than about 30 min, 25 min, 20 min, 15 min, 10 min, 9 min, 8 min, 7 min, 6 min, 5 min , 4 min, 3 min, 2 min, or less than about 1 minute. In some embodiments, HS-out performs standardized centralized laboratory test analyzes across wide test intervals.
The system can also be configured to detect infection with mutant or other strains that are not yet characterized. Before these strains are identified, spikes in inflammatory markers may indicate that an individual is infected with a strain that has not yet been identified, thus allowing for rapid potential containment and identification of the fact that the virus is mutated. Defensive measures (such as investments in vaccines) can then be updated accordingly.
HS technology is configurable to be simple to use and eliminates the various steps of sampling data analysis that would occur in existing situations (for example, collection, transport, remote control analysis, decision making). As a result, HS can provide greater accuracy and speed of decision, providing real-time data from the field to a central monitoring location, for example, that of a government agency. The system therefore offers the opportunity for optimal health support and direction. For example, FS systems can be located in community friendly locations, such as pharmacies, schools, clinics, or recreation centers, so that citizens could easily be tested and / or treated on a desirable basis, for example, to monitor infectious diseases, such as influenza. In addition, because the device can be portable, community workers can visit the elderly and others unable to travel, or make home visits when infection, for example, by the flu, is suspected. In some embodiments, the collected data is analyzed on both an individual and a population-based circumstance. These test data collected by the implanted FS devices can be made available to suppliers, government authorities, hospitals or the like.
When deployed in a region of interest, for example, a school, community center, shopping center, local, regional or national, HS can be used to develop security systems to monitor possible adverse events and health pandemics. The FS device can also be used in elevated screening strategies, where a large number of individuals, eg, all at risk or suspected of being at risk, can be tested on a routine basis, either preventively or in reaction to an outbreak. . The data collected by the FS is accumulated in the operating system, which then aggregates and manages the collective data. In some embodiments, the system requires only a small sample of body fluid, for example, a bite on the blood, saliva or sputum finger, the typical safety issues that arise from blood flame are greatly reduced or eliminated. In some embodiments, real-time data is used to help select the optimal biomarker assays for a given situation. In some embodiments, the analyte set is chosen prospectively as a sub-set from a large assay menu. Thus, the ideal test suitable for defining the initial stage of an epidemic (which may emphasize detection of the antigen) can be changed after the epidemic, for example, to look for antibodies that provide information about the likely stage of community immunity that may be relevant. the management of subsequent epidemics.
By monitoring infectious diseases, the Health Shield deployment strategy can provide screening and sampling for the population at risk from the minimum number of expected initial outbreaks. In some modalities, the system assumes the same range of cases that occurred to provide empirical data from the real world to model the spread of the disease.
An index case can potentially infect any number of secondary individuals. The number of secondary individuals can depend on any number of factors in the index case, including but not limited to age, mobility, life situation, work environment, socialization and geographic location. HS can model these factors and others to estimate the potential spread of a given outbreak. In a non-limiting example, real-world data suggests that a typical index case is likely to infect 50 other individuals. An exemplary infection pattern can include 4 or 5 family members and 45 or 46 co-workers, friends and others with whom the infected person has come into contact. In the HS rapid response model, each index case would require 25 to 50 secondary screens (regardless of age group) to prevent people in contact with the infection index case and the spread of the virus. Depending on the characteristics of the indexing process and infectious agent, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100 secondary screens may be required. In some embodiments, more than 100 secondary screens may be required for an index case.
In some embodiments, the HS is equipped with an initial amount of FS device cartridges, for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50 , 60, 70, 80, 90 or 100 times the expected number of index cases. In some embodiments, the system provides approximately 50 times the cartridges per expected number of index cases. Each cartridge can be used to test a body fluid sample, as described herein. The abundance of cartridges provides on-demand, proactive containment for pandemic mitigation. Once the infrastructure is activated, HS provides additional on-demand shipments as needed. This regime provides for sufficient screening and sampling to cover the population at risk around index cases.
Individuals can be provided with a device when purchasing a drug prescription by any common methods, for example, at a pharmacy. The individual may be given a device at a school, a workplace, or another area of interest. The devices can also be manually distributed by health care workers. When the device is distributed to an individual, individual contact information, including, without limitation cell phone, e-mail address, text message address or other wireless communication means, can at that time be entered into the databases. data from the operating system component and associated with the individual there. The system can include an OS script or other program that can detect when a signal generated from a detection device has not yet been sent to the OS system, for example at a certain time, and the OS system can then send an alert notifying the individual to test a body fluid sample.
Because of the portability and size of the FS components of the Health Shield, HS can become a part of everyday life for the management of disease and potential health risks. In some embodiments, the systems are placed in homes and in easily available locations. Real-time data collection and data analysis provide a proactive rapid health care system to respond to sudden outbreaks.
HS systems can provide the most efficient surveillance measures for the management of the disease. The HS system can identify outbreaks as early as possible to control and contain the spread to allow for appropriate, rapid mitigation strategies to be put in place. The model for a given location can be optimized to take into account different factors to provide better surveillance and mitigation strategy. A factor that includes prioritization tests based on risk factors and symptoms, including prioritization tests for babies, children, pregnant women, medical personnel, high-risk individuals and geriatrics. Another factor includes testing contacts close to index cases, such as test concentration at home, schools and workplaces where confirmed or suspected cases exist. In addition, the system how to assess the impact of alternating diagnostic tests based on various factors, such as sensitivity, specificity, turn around (that is, the time to obtain results from an assay). In some embodiments, the assays performed comprise one or more of the cartridge assays, real-time PCR, rapid antigen tests, viral culture, and immunoassays. In some embodiments, a less expensive test can be used for a large number of secondary tests to minimize the expense. Based on these data, a smaller number of more expensive, but more sensitive and specific assays can be used to test selected individuals.
When individuals suspected of being infected are detected by the SH, whether the individual is symptomatic or asymptomatic, assays can be performed in the field with the FS and the results and the location of the object can be relayed to the OS, for example, to a central server in a central monitoring location. At the monitoring site, results can be viewed and alerts recorded if applicable, so that containment efforts, including the deployment and testing of FS components, can be initiated. In some modalities, the model contained in the software will automatically suggest where the disease is likely to spread and where resources will have to be mobilized to contain the disease and do more in the field of monitoring. The system can contact individuals involved in surveillance, for example, government or health officials, for example, by phone, pager, fax, email, text message or quick form of communication. In some modalities, the data and analysis provided by HS is provided to employees and healthcare professionals, not to individual users. This helps to ensure that medical decision is made properly.
An advantage of the Health Shield as described here is that the results of testing the field systems can be substantially immediately communicated to a third party who can benefit from obtaining the results. For example, once the results of a measurement taken by an FS device are transmitted to the operating system, an analyte concentration can be determined in the operating system component and transmitted to an individual or medical staff who may need to take more measures. This may include the identification of an index case. The step of communicating to a third party can be done wirelessly, as described here, and by transmitting the data by hand from a third party device, the third party can be notified of the test results practically anytime and anywhere. Thus, in a time-sensitive scenario, a patient can be contacted immediately from anywhere if urgent medical action may be required.
The systems of the invention can be designed to interface with any combination of different electronic health records (CSR) and any other relevant databases. In addition, the system can be configured to automatically translate data that currently exists in different formats into a standard format. Once the system imports and translates the data, it can centralize the information in one or more repositories and pass the imported data through predictive models. In this way, the system can compile and take advantage of multiple data sources for a better hatching model and provide adequate containment responses. These models learn from each new data point, making it more and more predictable over time. In some embodiments, pattern recognition models that predict how a particular individual's disease is likely to progress.
A pilot program can be used to help refine the system's parameters. In some embodiments, an initial screening and containment strategy is developed. The HS is then deployed to pilot this model in a region of interest, for example, an area of the district, neighborhood, hospital or commercial. With this pilot project, the robustness of the assumptions underlying the modeling effort can be tested, and the containment strategy can be adjusted. In some modalities, fine-tuning is performed automatically by the operating system's learning algorithms. For example, the modeling software contains pattern recognition technologies that allow algorithms to predict the spread of the disease to be refined continuously with each new data point sent to the software portal. As such, the system becomes more and more predictive over time. In some embodiments, these improvements continue even after the system is in place, after the pilot phases.
After a system is developed using historical data, archived samples and even the pilot phase, the systems can be placed in strategic locations to begin to prevent the spread of an outbreak. Since each instrument can process different cartridges that can be customized for a given disease of interest, for example, with a specific strain of influenza of concern, the same systems can be used to contain and prevent the spread of a virus, even what mutations. In some embodiments, the cartridges contain tests based on proteins that measure inflammation and response to infection agents allowing to recognize serious infection even if the virus mutates, and specific assays for new viral trains can be immediately developed and deployed via the infra existing structure and instruments. In addition, the same instruments put in place to monitor infectious diseases are then available to control other health-related problems such as diabetes, obesity, cardiovascular disease and oncology concerns, for example, cancer therapy. Different cartridges and additional models for the software can be customized across existing HS systems. Validation data for each application can be performed before deployment and adjusted prospectively, learning from the data received.
Non-compliance with the recommended treatment may impair the effectiveness of the containment strategy of the present invention. As such, in some embodiments of the system of the present invention it can be used to monitor patient compliance and notify the patient's staff or other doctors of such non-adherence. For example, a patient having a pharmaceutical agent as part of the medical treatment plan may have a body fluid sample that is assayed as described here, but a metabolite concentration, for example, detected by the system may be at a high level compared to a known profile that will indicate multiple doses of the pharmaceutical agent have been taken. Personnel of the patient or physician may be notified of non-compliance by any method discussed here, including, without limitation, notification through such a portable device. PDA or cell phone, or through third parties, as a health professional who also receives the communication of non-compliance Such a profile of an acquaintance can be located or stored on an external device described here.
In one embodiment, the system can be used to identify sub-populations of patients who are benefited or harmed by therapy. In this way, drugs with potential toxicity can be administered to only those who will benefit.
In terms of pharmaceuticals related to adverse events, shield health systems can be placed in an individual's residence. In some embodiments, HS is used to monitor the safety and effectiveness of treatments for acute, for example, debilitating or life-threatening illnesses, or for chronic conditions. The FS components can also be placed in central locations, such as pharmacies such that individuals can be tested when filling prescriptions.
Case studies have been carried out for diabetes, infection, and oncology, considering the needs of government disease management systems, as well as healthcare companies. Such a study was aimed at a model for the prevention and reversal of diabetes. The modeled data demonstrated a dramatic reduction in costs associated with the elimination of centralized blood infrastructure and analysis of health information data and instead of using the systems of the present invention with FS systems placed at various points of care, including the environment domestic. The system provided savings, in part, by limiting transportation costs, reducing personnel costs associated with implementing reductions, analyzing costs associated with false positives, reducing time associated with waiting for results. In various modeling environments, the HS system would reduce the costs associated with conventional testing by more than about 50%, in addition to the value of the saved time in acquiring the relevant data. 3. Monitoring of Influenza outbreaks
In one aspect, the systems of the invention are deployed to monitor and contain disease outbreaks. HS is particularly beneficial in the flu scenario because the containment strategies that initially depend on mass vaccination programs may not be effective enough to contain an outbreak. Influenza A virus strains are classified according to two proteins found on the surface of the virus: hemagglutinin (H) and neuraminidase (N). All influenza A viruses contain these two surface proteins, but the structures of these proteins differ between strains of virus, due to rapid genetic mutation in the viral genome. There are 16 known H and 9 subtypes of N in birds, but only a subset, for example, H 1, 2 and 3, and N 1 and 2, are generally found in humans. The pathogenicity of a strain varies between subtypes. For example, the H5N1 strain, commonly referred to as "bird flu" or "bird flu", most commonly affects birds, but a recent outbreak of the strain in humans in Asia has killed more than 60% of those infected.
Although flu vaccines can help prevent the spread, the subtypes of influenza variables and mutations make vaccination only a partial solution. For example, the H1N1 flu virus, commonly referred to as the Swine Flu, is responsible for the 2009 pandemic. Like H5N1, H1N1 can be virulent in humans. The United States Center for Disease Control and Prevention (CDC) maintains information on the 2009 H1N1 pandemic at www.cdc.gov/H1N1FLU/. The CDC is concerned that the new H1N1 flu virus could result in a particularly serious flu season in 2009, for example, due to widespread illness, medical appointments, hospital admissions and deaths. The first H1N1 vaccine will not be available until mid-October at the earliest, and vaccine supplies will not be sufficient to treat even the most at-risk populations until later in the fall. As a result, the best way to avoid a widespread epidemic and public panic will be to control the virus, preventing it from spreading, particularly to those who are most at risk.
Some governments have tried methods of containing the flu that have been effective with Severe Acute Respiratory Syndrome (SARS), including screening for fever or respiratory symptoms. However, these methods are not sufficiently oriented to contain H1N1. One problem is that flu victims can be contagious at least the day before a fever or other symptoms present. In some embodiments, the Health Protection of the invention systematically tests not only those who are symptomatic, but also family members and close working associates. Thus, infected individuals can be treated and isolated before they have an opportunity to spread the infection widely, reducing the real and psychological impact of the flu. The spread and death rate of the flu in the fall of 2009 would mitigate how to keep patients from flooding emergency rooms for testing and treatment. Potentially hundreds of millions of dollars can be saved by reducing costly emergency and hospital visits, by using medication correctly, and by reducing viruses spread in hospitals. The SH models of the invention can identify optimal intervention and timing strategies for administering appropriate medication, such as Tamiflu. These steps can cut down on hospital and emergency care and allow people to get back to work more quickly. Eliminating these unnecessary emergency room visits can help prevent the virus from spreading and reduce hospitalization and emergency room expenses.
Influenza, for example, H1N1 and H5N1, can be detected from a body fluid, for example, a blood prick on the sputum, saliva, or a combination thereof, using SC point-of-care instruments. These instruments can be placed in appropriate locations (for example, at home, schools, restaurants, primary care units, livestock facilities, etc.) and can be deployed in many cases without local support infrastructure other than a power supply. The test can be done quickly, for example, in minutes, less than about 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55 or 60 In some embodiments, SC results are reported back to a central OS monitoring location in real time. Blood or saliva assays can detect influenza by a variety of methods, including immunodetection by sensitive antibodies to specific epitopes of the virus itself, for example, hemagglutinin and / or neuraminidase. The assays can distinguish between the various types of influenza strains identified, for example, influenza A, influenza B, H5N1, H1N1, etc. The assays can detect individual particles of a particular virus strain, even on a background of different strains or genetic variants. The assays can detect biomarkers, viral proteins, coat proteins, and the like.
In some embodiments, the assays measure inflammatory markers and immune response markers, for example, cytokines, which allow clinicians to identify the severity of the infection, the extent of the acute phase and / or inflammatory reactions of the subject. This can, for example, help to determine the appropriate treatment regimen for an individual. The ability to measure the response to infection allows characterization of the infection even for strains of virus that have not yet been characterized. Once these strains are characterized, specific tests can be customized and added to the cartridges. Depending on the test required, the new tests can be implemented immediately, within days, weeks, or in a matter of months.
There are currently more than 100 cytokines / chemokines whose coordinates or conflicting regulations are of clinical interest. Examples of cytokines that can be used in systems and methods of the invention include, but are not limited to, BDNF, pS133 CREB, Total CREB, DR-5, EGF, ENA-78, eotaxin, protein binding fatty acid, FGF- base, granulocyte colony stimulating factor (G-CSF), GCP-2, granulocyte-macrophage IFN colony stimulating factor GM-CSF (GM-CSF), growth-related keratinocyte oncogene (GRO-KC), HGF , ICAM-1, -alpha, IFN-gamma, interleukins. IL-10, IL-11, IL-12, IL-12 p40, IL-12 p40 / p70, IL-12 p70, IL-13, IL-15, IL-16, IL-17, IL-18, IL -1alpha, IL-lbeta, IL-1ra, IL-1 ra / IL-1 F3, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL -9, inducible protein interferon (10 IP-10), JE / MCP-1, keratinocytes (KC), KC / GROa, LIF, Lymphotacin, M-CSF, chemo-attracting protein monocyte-1 (MCP-1), MCP- I (MCAF), MCP-3, MCP-5, MDC, MIG, inflammatory macrophage (MIP-1 alpha), MIP-1 beta, MIP-1 gamma, MIP-2, MIP-3 beta, OSM, PDGF-BB , regulated by activation, normally expressed and secreted T cell (RANTES), Rb (pT821), Rb (total), Rb pSpT249 / 252, Tau (pS214), Tau (pS396), Tau (total), Tissue Factor, factor tumor necrosis alpha (TNF-alpha), TNF-beta, TNF-R1, TNF-RII, VCAM-1, and VEGF. In some embodiments, the cytokine is IL-12p70, IL-10, IL-1 alpha, IL-3, IL-12 p40, IL-1 ra, IL-12, IL-6, IL-4, IL-18, IL-10, IL-5, eotaxin, IL-16, MIG, IL-8, IL-17, IL-7, IL-15, IL-13, IL-2R (soluble), IL-2, LIF / HILDA , IL-1 beta, Fas / CD95 / Apo-1, and MCP-1.
Inflammation markers that can be used with the systems and methods of the invention include ICAM-1, RANTES, MIP-2, MIP-1-beta, MIP-1-alpha, and MMP-3. Additional inflammation markers include adhesion molecules, such as α1β1, a2β1, a3β1, a4β1, a5β1, a6β1, a7β1, a8β1, a9β1, aVβ7, a4β7, a6β4, aDβ2, aLβ2, aMβ2, aVβ, aVβ, aVβ, , aXβ2, allβ3, alELbβ7, beta-2 integrin, beta-3 integrin, beta-2 integrin, beta-4 integrin, beta-5 integrin, beta-6 integrin, beta-7 integrin, beta-8 integrin, alpha-1 integrin, alpha-2 integrin, alpha-3 integrin, alpha-4 integrin, alpha-5 integrin, alpha-6 integrin, alpha-7 integrin, alpha-8 integrin, alpha-9 integrin, alpha-D integrin, alpha-L integrin, alpha-M integrin, alpha-V integrin, alpha-X integrin, alpha-llb integrin, alphalELb integrin; Integrin associated molecules such as beta IG-H3, Melusin, CD47, MEPE, CD151, osteopontin, IBSP / sialoprotein II, RAGE, IGSF8; selectins, such as E-selectin, P-selectin, L-selectin, and ligands such as CD34, GlyCAM-1, AdMCAM-1, PSGL-1, receptor, vitronectic vitronectin, fibronectin, vitronectin, collagen, laminin, ICAM - 1, ICAM-3, BL-CAM, LFA-2, VCAM-1, NCAM, and PECAM. Additional inflammation markers include cytokines, such as IFN-α, IFN-β, IFN-E, -K, -T, e-Ç, IFN-w, IFN-y, IL29, IL28A and IL28B, IL-1, IL- 1a and β IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13 , IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL -26, IL-27, IL-28, IL-29, IL-30, eTCCR / WSX-1. Additional inflammation markers include cytokine receptors, such as common beta chain, IL-3 R alpha, IL-3 R beta, GM-CSF R, IL-5 R alpha, common chain / IL-2 R gamma, IL- 2 R alpha, IL-9 R, IL-2 R beta, IL-4 R, IL-21 R, IL-15 R alpha, IL-7 R alpha / CD127, IL-1 ra / IL-1 F3, IL -1 R8, IL-1 RI, IL-1 R9, IL-1 Rll, IL-18 R alpha / IL-1 R5, IL-1 R3 / IL-1 R ACP, IL-18 R beta / IL-1 R7, IL-1 R4 / ST2 SIGIRR, IL-1 R6 / IL-1 R rp2, IL-11 R alpha, IL-31 RA, CNTF R alpha, R leptin, G-CSF R, R LIF alpha, IL- 6 R, OSM R beta, IFN-alpha / beta R1, R2 IFN-alpha / beta, IFN-gamma R1, IFN-gamma R2, IL-10 R alpha, IL-10 R beta, IL-20 R alpha, IL -20 R beta, IL-22 R, IL-17 R, IL-17 RD, IL-17 RC, IL-17B R, IL-13 R alpha 2, IL-23 R, IL-12 R beta 1, IL -12 R beta 2, TCCR / WSX-1, and IL-13 R alpha 1. Additional inflammation markers include chemokines such as CCL-1, CCL-2, CCL-3, CCL-4, CCL-5, CCL-6 , CCL-7, CCL-8, CCL-9, CCL-10, CCL-11, CCL-12, CCL -13, CCL-14, CCL-15, CCL-16, CCL-17, CCL-18, CCL -19, CCL-20, CCL-21, CCL-22, CCL -23, CCL-24, CCL-25, CCL-26, CCL-27, CCL-28, MCK-2, MIP-2, CINC-1, CINC-2, KC, CINC-3, LIX, GRO, Thymus Chemokine-1, CXCL-1, CXCL-2, CXCL-3, CXCL-4, CXCL-5, CXCL-6, CXCL-7, CXCL-8, CXCL-9, CXCL-10, CXCL-11, CXCL- 12, CXCL-13, CXCL-14, CXCL-15, CXCL-16, CXCL-17, XCL1, XCL2, and Chemerin. Other inflammatory markers include chemokine receptors such as CCR-1, CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR- 9, CCR-10, CXCR3, CXCR6, CXCR4, CXCR1, CXCR5, CXCR2, Chem R23. Additional inflammation markers include tumor necrosis factors (TNF), such as TNFa, 4-1 BB Ligand / TNFSF9 and LIGHT / TNFSF14 and APRIL / TNFSF13 and lymphotoxin, BAFF / TNFSF13B and lymphotoxin beta / TNFSF3 and CD27 Ligand / TNFSF7 and ligand / 0X40 TNFSF4, CD30 Ligand / TNFSFδ, TL1A / TNFSF15, CD40 Ligand / TNFSF5, TNF-alpha / TNFSF1A, EDA, TNF-beta / TNFSF1B, EDA-A2, TRAIL / TNFSF10, Fas Ligand / TNFSF6, TRANCE / TNFSF11, GITR Ligand / TNFSF18, and TWEAK / TNFSF12. Additional inflammation markers include TNF receptors, such as Superfamily 4- 1BB / TNFRSF9, NGF R / TNFRSF16, BAFF R / TNFRSF13C, Osteoprotegerin / TNFRSFUB, BCMA / TNFRSF17, OX40 / TNFRSF4, CD27 / TNFRSF7, RANK11, RANK11 / TNFRSF8, RELT / TNFRSF19L, CD40 / TNFRSF5, TACI / TNFRSF13B, DcR3 / TNFRSF6B, TNF RI / TNFRSF1A, DcTRAIL R1 / TNFRSF23, TNF RII / TNFRSF1B, DcTRAIL R2 / TNFRSF1 / TNFRSF10B, DR6 / TNFRSF21, TRAIL R3 / TNFRSF10C, EDAR, TRAIL R4 / TNFRSF10D, Fas / TNFRSF6, TROY / TNFRSF19, GITR / TNFRSF18, TWEAK R / TNFRSF12, HVEM / TNFRSF14 and XEDAR. Additional inflammation markers include TNF superfamily regulators, such as FADD and TRAF-2, RIP1 and TRAF-3, TRADD, TRAF-4, TRAF-1, and TRAF-6. Additional inflammation markers include acute-phase reagents and acute-phase proteins. Additional inflammation markers include TGF-beta superfamily ligands, such as Activivins, Activin A, Activin B, Activin AB, Activin C, BMPs (Bone Morphogenetic Proteins), BMP-2, BMP-7, BMP-3, BMP-8, BMP -3b / GDF-10, BMP-9, BMP-4, BMP-10, BMP-5, BMP-15 / GDF-9B, BMP-6, Decapentaplegic, Growth / Differentiation Factors (GDFS), GDF-1, GDF-8, GDF-3, GDF-9 GDF-5, GDF-11, GDF-6, GDF-15, GDF-7, the ligands of the GDNF family, Artemin, neurturin, GDNF, persephin, TGF- beta, TGF-beta, TGF-beta 3, TGF-beta 1, TGF-beta 5, LAP (TGF-beta 1), BP1 latent TGF-beta, Latent TGF-beta 1, BP2 latent TGF-beta, from TGF- beta 1,2, Latent TGF-beta BP4, TGF-beta 2, Lefty, MIS / AMH, Lefty-1, Nodal, Lefty-A, Activin RIA / ALK-2, GFR alpha-1 / GDNF R alpha-1, Activin RIB / ALK-4, GFR alpha -2 / GDNF R alpha-2, Activin RIIA, GFR alpha-3 / GDNF R alpha-3, Activin RIIB, GFR alpha-4 / GDNF R alpha-4, ALK-1, MIS Rll, ALK-7, Ret, BMPR-IA / ALK-3, TGF-beta RI / ALK-5, BMPR-IB / ALK-6, TGF-beta Rll, BMPR-II, TGF-b eta R11b, Endoglin / CD105, and TGF-beta R111. Other inflammation markers include TGF-beta superfamily modulators such as Amnionless and NCAM-1 / CD56, Bambi / NMA, Noggin, BMP- 1 / PCP, Nomo, Charonte, PRDC, Cerberus 1, SKI, Chordin, Smadi and Chordin-Like 1 , Smad2, Chordin-Like 2, Smad3, COCO, Smad4, CRIM1, Smadõ, Crypto, Smad7, Crossveinless-2, Smad8, Cryptic, SOST, DAN, TGF-beta Latent BP1, decorin, BP2 TGF-beta Latent, FLRG, BP4 TGF-beta Latent, follistatin, TMEFFI / Tomoregulin-1, follistatin-like 1, TMEFF2, GASP-1 / WFIKKNRP, TSG, GASP-2 / WFIKKN, TSK, Gremlin, and Vasorin. Additional inflammation markers include ligands, such as EGF Amphiregulin, LRIG3, Betacellulin, Neuregulin-1 / NRG1, EGF, Neuregulin-3 / NRG3, Epigen, TGF-alpha, Epiregulin, TMEFF1 / Tomoregulin-1, HB-EGF, TMEFF2, and LRIG1. Additional inflammation markers include EGF R / ErbB family of receptors, such as EGF R, ErbB3, ErbB2, and ErbB4. Additional inflammation markers include fibrinogen. Additional inflammation markers include AEA. Additional inflammation markers include glial markers, such as alpha. 1-antitrypsin, C-reactive protein (CRP), a2-macroglobulin, glial fibrillary acidic protein (GFAP), Mac-1, and F4 / 80. Additional inflammation markers include myeloperoxidase. Additional inflammation markers include Complement markers, such as C3d, C1q, C5, C4d, bp C4, and C5a-C9. Additional inflammation markers include major histocompatibility complex (MHC) glycoproteins, such as HLA-DR and HLA-A, D, C. Additional inflammation markers include microglial markers, such as CR3 receptor, MHC I, II MHC, CD 31 , CD11a, CD11b, CD11c, CD68, CD45RO, CD45RD, CD18, CD59, CR4, CD45, CD64, and CD44. Other inflammatory markers include a2 macroglobulin receptor, fibroblast growth factor, Fc IR gamma, Fc gamma Rll, CD8, LCA (CD45), CD18, CD59, Apo J, Clusterin, type 2 plasminogen activator inhibitor, CD44, colony of macrophages stimulating factor receptor, MRP14, 27E10, 4-protein-hydroxinonenal conjugates, IkB, NFkB, CPLA 2, COX-2, matrix metalloproteinases, membrane lipid peroxidation, and ATPase activity. HSPC228, Emp1, Cdc42, TLE3, SPRY2, p40BBP, HSPC060 and NAB2, or a sub-regulation of HSPA1A, HSPA1B, MAPRE2 and OAS1 expression, TACE / ADAM17, alpha-1-acid glycoprotein, angiopoietin-1, MIF, angiopoietin- 2, CD14, beta-defensin 2, MMP-2, ECF-L / CHI3L3, MMP-7, EGF, MMP-9, EMAP-II, MSP, PT-RAGE, nitric oxide, endothelin-1, Osteoactivin / GPNMB, FPR1, PDGF, FPRL1, pentraxin 3 / TSG-14, FPRL2, Gas6, PLUNC, GM-CSF, RAGE, S100A10, S100A8, S100A9, HIF-1 alpha, substance P, TFPI, TGF-beta 1, TIMP- 1, TIMP-2, TlMP-3, TIMP-4, TLR4, LBP, TREM-1, leukotriene A4, TSG-6 hydrolase, lipocalin-1, uPA, M-CSF, eVEGF.
The physiological data of each individual can also be measured and communicated from the SC instruments or points of care to the SO. Such data may include, without limitation, temperature, heart rate / pulse, blood pressure, oximetric signals, weight, water retention, plethysmographic signals, respiratory rate, fat content, water content, blood perfusion, mobility, posture , bioelectrical impedance, electrocardiogram (ECG), or galvanic skin response.
In some embodiments, assays are used to detect antibodies from the host against a specific pathogen or marker. A potential problem when measuring such antibodies is the interference that can occur in individuals who have had a flu vaccination in the past. In such situations, high levels of influenza antibodies in the blood may interfere with the assay. Influenza viruses, mainly repetitions in the lungs, can therefore be detected in, for example, sputum, nasal lavage and saliva. Therefore, a sample of base saliva can also be processed at the point of care for verification. The hemagglutinin antigen (H antigen) on the surface of the influenza particles is believed to be essential for the entry of the virus into target cells. Hemagglutinin can bind red cells and in appropriate conditions causes the cells to clump together. Therefore, red blood cells can act as concentrating agents for the virus. This phenomenon can be exploited in assays for the virus since red cells can be concentrated before a blood sample is analyzed. In addition, the red cells can be collected (and concentrated) on a suitable surface in a test cartridge, and may contain large amounts of virus for analysis and detection.
Two main measures of evaluation of any medical examination or diagnostic test are its sensitivity and specificity, which measures how well the test performed is to accurately detect all affected individuals, without exception, and without false indication, including in individuals who they do not have the target disease (value prediction).
A true positive result (TP) is when the test is positive, the condition is present. A false result (FP) is positive when the test is positive, but the condition is not present. A true result (TN) is negative, where the test is negative and the condition is not present. A false negative (FN) result is that the test is negative, but the condition is not present. In this context: Sensitivity = TP / (TP + FN); Specificity = TN / (TN + FP) and predictive value of a positive = TP / (TP + FP).
Sensitivity is a measure of a test's ability to correctly detect the target disease in an individual being tested. A test that has little sensitivity produces a high rate of false negatives, that is, individuals who have the disease, but are falsely identified as being free from the particular disease. The potential danger of a false negative is that the sick individual will remain undiagnosed and treated for a period of time, during which the disease may progress to treatments where at a later stage, if any, it may be less effective. An example of a test that has a low sensitivity is a blood test based on HIV proteins. This type of test exhibits insufficient sensitivity because it fails to detect the presence of the virus until the disease is well established and the virus has invaded the bloodstream in substantial numbers. In contrast, an example of a test that has a high sensitivity is viral load detection using the polymerase chain reaction (PCR). High sensitivity is achieved because this type of assay can detect very small amounts of the virus. High sensitivity is particularly important when the consequences of missing a diagnosis are high.
Specificity, on the other hand, is a measure of a test's ability to accurately identify patients who are free from disease. A test having poor specificity produces a high rate of false positives, for example, individuals who are falsely identified as having the disease. A disadvantage of false positives is that they force patients to undergo unnecessary medical treatments with their inherent risks, emotional and financial stresses, and which could have adverse effects on the patient's health. Specificity is important when the cost or risk associated with other diagnostic procedures or additional medical intervention is very high.
In some embodiments, HS performs multiple assays to improve assay sensitivity and / or specificity. For example, the sensitivity and specificity of disease monitoring can be improved. In some embodiments, multiple body samples are tested for an individual. For example, tests based on saliva and blood (finger) can be performed simultaneously by people who have previously been vaccinated against the flu. Testing multiple samples can increase the possibility of identifying the infection. In addition, it can be important to control false negatives to maximize containment. In some embodiments, the present invention addresses false negatives by including tests for both inflammation and infection markers in each test cartridge. When the flu test is negative, but these other markers are strongly suggestive of flu, confirmatory tests can be included for that specific subset of patients. A variety of exemplary marker panels, also referred to as the test menus, are revealed here for the definitions of various diseases. One skilled in the art will appreciate that the use of multiple assays and / or physiological parameters to improve sensitivity and / or specificity is not limited to these exemplary embodiments, but can be an effective technique in controlling many diseases and disorders.
In some embodiments, the decentralized detection capability provided by ES units can provide early identification of people with a confirmed case of influenza, that is, an "index case", and then consult all contacts close to those individuals as well. identified. Given this network of contacts, containing the spread of the epidemic ideally requires rapid implantation, identification and preventive action in an exposed and / or asymptomatic infected population. HS provides a system to perform these operations and prevent the spread of the disease.
The Health Protection system can be implemented for the surveillance and containment of an outbreak of influenza. HS can be deployed in a variety of configurations, for example, at the local, regional or national level. The operating system of a given context can be used in silicon modeling to simulate various implantation strategies to better contain the flu or other condition and can be optimized for each configuration. In some embodiments, the model comprises an epidemiological model that includes a variety of parameters appropriate to model the expected and / or contained outbreak. In some embodiments, the system uses Monte Carlo simulations to test a spectrum of tracking and containment strategies that will, in turn, be analyzed for cost / benefit ratio, etc. For example, the system can design where and how to deploy limited resources, for example, medical personnel, therapeutic treatments and vaccines. The OS model can be preloaded with individual and specific population information for the configuration to be monitored. These factors include, but are not limited to the incubation times, the connectivity of the susceptible population, the mode of infection, virulence of the virus, the mortality and hospitalization rates, the disease incidence rates, the mode of transmission, the rate of infection, the results of therapeutic intervention, the effectiveness of the vaccine, and resistance or effectiveness of anti-viral therapies, for example, Tamiflu. Parameters for the individuals being monitored include, without limitation, age, sex, social contacts (living conditions, family, co-workers, etc.), previous history of illness, general health (for example, other pre-conditions) parameters), etc. The Model can be continuously updated once the system is implemented.
FS instruments are deployed to operate in conjunction with the configured operating system. In some embodiments, FS data is provided to an OS through a software portal. The remote operating system can then perform the desired calculations. In general, FS systems are deployed at selected points. In some embodiments, the OS model is used to drive the optimal deployment of FS instruments. Ideal locations and hot spots include, without limitation, areas where people gather, for example, shopping areas, schools and workplaces. Places where sick people gather are also targeted, including, without limitation clinics, pharmacies and hospitals. In some embodiments, SC devices are deployed for homes, as described here
Once deployed, SC systems are used to test themes. In some embodiments, this includes testing for disease antigens, for example, viral coat proteins. The analytes also include host proteins as disease markers, for example, immunological markers, including cytokines, and inflammatory markers that indicate an ongoing infection. In detecting infectious disease agents and assessing the status and prognosis of patients, it may be desirable to be able to measure multiple analytes simultaneously. For example, this increases the possibility of detecting disease as any single analyte cannot be found at abnormal levels. Multiple analyte measurements also reduce noise and can make the system more accurate in monitoring the disease. The following table provides an example menu for detecting the H1N1 virus, also known as swine flu: Table 3
[00.154] In the table, "Ab: Ag" represents the complex formed between an antibody (Ab) and an antigen (Ag). For example, "anti-H1: H1 IgG" represents a complex between the host of anti-H1 IgG-antigens and antibodies to the hemagglutinin H1 flu. As different strains of influenza are monitored, the menu will be adjusted accordingly. For example, a menu to control H1N5 viruses that comprise the detection of N5 antigen and anti-N5 antibodies.
Detection of IgM against IgG or IgA can be used to determine whether an individual has had prior exposure to the flu particles of interest. IgM antibodies are made quickly in the days after infection on the first exposure to an immunogen. When previously exposed individuals encounter a second infectious agent that has a similar or identical antigenic character, IgG and IgA antibodies are produced very quickly. This secondary response is typically much stronger and more specific than the original IgM response. In primary infections and very serious infections, an active virus is more likely to be present in the blood and to be directly detectable. In secondary infections, where the antibody is present, it will generally be in excess of the antigen and the antigen can be masked for immunoassay methods. In some embodiments, the complex formed by antigen and antibody is detected using a sandwich immunoassay in which one reagent is directed to the antigen and the other IgG. Since a subject produces IgG and IgA antibodies, these can be found in the blood well after the infection has resolved. [00.156] As shown in Table 3, the menu can also include one or more cytokines as a marker of immune response and / or inflammation. Cytokines of interest include, without limitation, IL-1 β, IL-6, IL-8, IL-10 and TNFα. Cytokines such as these can be produced in large quantities during the early part of a viral infection. In some cases, the level of these markers will rise and fall quickly. Valuable information regarding the patient's condition and prognosis can be obtained by taking serial measurements of one or more cytokines. For example, fevers of viral and bacterial origin can be distinguished by measuring changes in cytokine levels. A recent study found that "CRP speed" (CRPV), defined as the relationship between C-reactive protein in the blood on admission to an emergency room and the number of hours since the onset of fever, can differentiate between bacterial and non-acute tracts -bacterial febrile diseases. Paran et al., C-reactive speed protein to distinguish febrile bacterial infections from non-bacterial febrile illnesses in the emergency department, Crit Care. 2009; 13 (2): R50. The study also revealed that blood levels of other acute-phase proteins, such as IL-1, IL-6 and TNF-ct, correlated with CRPV.
The levels of detection of influenza markers are shown in Table 4:
The exemplary markers in Tables 3 and 4 correspond to a menu for the detection of H1N1. The threshold levels for detecting a certain marker are shown in Table 4. When measurements are made over a period of time, the increase in folding in a marker, for example, cytokines or C-reactive protein, can be detected. Here, a 10 x change is considered to be indicative of an event. When time courses data for an individual are not available, the bend-change can be determined by comparison with a reference threshold. For example, the detected level 15 of a given marker can be compared with the average level of the marker in the general healthy population. It will be appreciated that different strains of influenza, for example, H5N1, H3N2, etc., can be detected using appropriate analytical methods. A clear action recommended by the SO for influenza when the detection of a given marker is shown in Table 5.

As stated above, the example in Table 5 highlights H1N1 swine flu. It will be appreciated that different strains of influenza, for example, H5N1, H3N2, etc., can be detected using appropriate analytical methods. In addition, the action will depend on a number of factors, including but not limited to expected virulence, cost transmission, treatment, etc. For example, a quarantine may be required for a virulent strain, but not for a lesser outbreak. serious. The course of action for drug resistance may depend on the drug. In the flu setting, resistance to oseltamivir (Tamiflu ®) can be especially important. Oseltamivir is an orally active antiviral drug that acts as a neuraminidase inhibitor. The drug slows down the spread of the influenza virus (flu) between cells in the body, stopping the new virus from chemically cutting ties with its host cell. It can be used for both influenza A and B. Resistance can be determined by a number of methods, for example, a functional assay (culture) or identification of a genetic marker. Zanamivir is also used to treat flu infection.
Specific strains of influenza virus can be detected using a sandwich assay format. A number of test configurations can be used. Figure 3 illustrates assays for H1N1 antigen illustrating sandwich complexes in four different assay types. One skilled in the art will understand that a similar arrangement can be used to detect other strains of viruses, for example, H5N1, H2N3, etc. shown are the final reaction products for four assay configurations for measuring H1N1 viruses (having multiple copies of each in the particle viral). The assays involve: 1) adding a sample, for example, blood, serum, saliva or nasal wash, to a capture surface to have an antibody to one of the viral surface antigens (H1, N1), 2) the addition of enzyme -labeled antibody to one of the surface antigens, and 3) washing the surface to remove unbound viral particles. Different test configurations can detect several particles. A-H1 / a-N1 and a-N1 / a-H1 configurations will measure H1N1 viruses, the a-H1 / a-H1 configuration detects any virus having H1 antigen, and a-N1 / a-N1 configuration detects any virus having the N1 antigen. A cartridge system for detecting assays is described in U.S. Patent Application 11/746, 535, filed May 9, 2007 and titled "real time detection of influenza virus." Sandwich assays can also be used to detect antibodies host for influenza strains, for example, human H1N1 swine flu antibodies. A first embodiment of the assay is shown in Figure 4A. In the Figure, the capture phase assay has antibody to viral antigen bound to a solid phase, the viral particle (antigen) can be captured by the solid phase and a detection reagent, for example, with alkaline phosphatase antibody labeled to the viral antigen, can be used to detect host antibodies.
This assay is set up as an antigen assay. antibody is detected by adding viral antigen to the sample, for example, body fluid such as blood or plasma, and by comparing the assay response, with and without added antigen, anti-viral antibodies can be measured by adding (spiking) a fixed, known amount of virus or viral antigen to the patient sample. After incubation, the spiked sample is used in an assay for the viral antigen. If antibodies are present, the assay will exhibit a reduction in the measured antigen (low ear recovery), the sample dilution or the level of the spiked antigen can be titrated to give a quantitative value for the antibody. When antibody to the viral antigen is present, there is little or no signal generated in the absence of added antigen, there is a reduced (or zero response) when the antigen is added compared to the antigen response to negative control samples that were added with antigen. In other words, the "spike recovery" antigen is low or zero, the amount of antibody can be deduced from the spike recovery if it is more than zero, antibody in the sample can also be titrated using growing antigen peaks until a response from the test is obtained. One skilled in the art will appreciate that the assays can be adapted to detect the host antibody to other strains of virus, for example, H5N1. The method can also be adapted to detect host antibodies to any appropriate antigen, for example, to other microbial insults.
Another configuration for detecting host antibodies to influenza virus particles is shown schematically in Figure 4B. This is a method of direct detection. In this embodiment, the capture phase assay has viral antigen bound to a solid phase and uses a detection reagent-alkaline phosphatase compound antibody labeled with human immunoglobulin. As described herein, the host antibody ideotype can determine whether the host is naive to the antigen (IgM antibodies are found) or has had previous exposure (IgG or IgA antibodies are found). Through the use of antibodies specific to the species of immunoglobulins, for example, IgM, IgG, IgA, etc.), the type of antibody can be determined. The assay involves: 1) incubating the sample with a capture surface to which it is bound with virus and / or viral antigen, 2) washing the Surfact to remove uncoupled IgG, then 3) incubating with an enzyme labeled with immunoglobulin anti-human specific for IgG or IgM, 4) wash to remove uncoupled enzyme labeled and 4), incubate with substrate. Figure 4B shows the test status after the fourth step.
FS systems are used to monitor analytes and other individual parameters (blood pressure, temperature, weight, etc.) over time. In some embodiments, tests are performed on an individual on a defined schedule, for example, one or more tests can be performed at least each h, 2 h, 3 h, 4 h, 5 h, 6 h, 7 h, 8 h, 9 h, 10 h, 11 h, 12 h, 13 h, 14 h, 15 h, 16 h, 17 h, 18 h, 19 h, 20 h, 21 h, 22 h, 23 h, 24 h , 36 h, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 10 days, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 2 months, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or at least every year. The frequency of tests can vary between individuals and between different diseases. For example, those considered at risk, for example, school children, the elderly, health workers and doctors, can be tested more frequently. In some embodiments, the operating system directs the frequency of tests. For example, the operating system can identify people at risk in more frequent hourly tests. The test can also be scheduled in real-time or semi-real time. For example, when an index case is identified, individuals in social contact with the index case could be tested immediately and more frequently afterwards. In some embodiments, the frequency of testing is increased at an access point with increased risk. In some embodiments, the test frequency is reduced as the risk is decreased, which allows to conserve resources.
As noted, a variety of field devices can be used with the systems and methods of the invention. The operating system can drive an optimized deployment of FS devices. In some embodiments, the types of assays are adjusted over time such as threat changes, for example, to monitor different analytes. In some embodiments, the type or types of sample are adjusted over time as the threat changes. In addition, viral nucleic acid was detected in the blood using PCR techniques, for example, real-time PCR. In some embodiments, multi-type sample cartridges as described herein are used. These cartridges allow sample processing and analysis of a limited number of analytes in more than one type of sample, for example, using one or more blood, concentrated red cells, sputum, saliva, nasal wash, or other body fluid . In some embodiments, multi-analyte cartridges as described herein are used. These cartridges can carry out the analysis of many analytes on a single sample type. Both types of cartridges can be used in a configuration considered to be optimal in a given context.
The implanted FS systems are used to test the types of samples selected using the selected assays, and the results are reported back to the OS system, as described here. In assessing individuals for possible flu infection, it is advantageous to take a series of measurements over time. Based on the initial measurements, the ideal analyte set can be changed to optimize the information collected by the test system. The use of such longitudinal measurements allows the calculation of trends at the analyte levels that indicate trends in disease processes. In some embodiments, the longitudinal measurements of the invention take into account the dynamic data of particular individuals, along with information from the population gathered in previous epidemics. In some embodiments, the models also adjust for cut data from individuals exposed to a current epidemic.
The SO monitors the data received for the incidence of infection, and provides recommendations for assessment and containment when an infection is found. When an infection is observed, the appropriate parties are notified, for example, individuals, social contacts, health professionals and government officials. In some embodiments, the course of action recommended by the OS is used to contain the spread of the virus. In some embodiments, the course of action includes providing therapeutic treatment for an infected individual. In some embodiments, prophylactic treatment is administered to anyone who is in contact with the infected individual. This may include vaccination. In some embodiments, depending on the severity of the outbreak, infected individuals can be quarantined. Those who have contact with the infected individual can be quarantined as well.
FS and OS continue to monitor the entire and continuously update the OS database with the information received. In some embodiments, the operating system adjusts the recommended action in response to actual measurements. In this way, the Health shield of the present invention provides a dynamic response to the detected outbreak. Once the outbreak has been contained, the FS system components can be moved to alternative hotspots, etc. 4. Monitoring of infectious disease
It will be appreciated that the systems of the invention as described above can be employed to control the incidence of a number of infectious diseases in addition to influenza. For example, HS can be deployed to monitor and prevent the spread of infectious diseases in areas where resources are limited, for example, rural or in remote areas, or developing countries. In some embodiments, HS is used to monitor Acquired Immunodeficiency Syndrome (AIDS), tuberculosis (TB), and / or malaria. AIDS is a disease of the human immune system caused by the human immunodeficiency virus (HIV). HIV is transmitted through direct contact of a mucous membrane or in the bloodstream with a body fluid containing HIV, such as blood, semen, vaginal fluid, the preseminal fluid, and breast milk. The disease is also spread by sharing infected syringes used to inject illicit drugs. AIDS progressively reduces the effectiveness of the immune system and leaves individuals sensitive to opportunistic infections and tumors. This weakening of the immune system exacerbates the risks of tuberculosis and malaria. Tuberculosis is a common and often deadly infectious disease caused by mycobacteria, tuberculosis, for example, Mycobacterium. Tuberculosis mainly resides in the lungs and spreads through the air when infected individuals cough, sneeze, or saliva. Malaria is an infectious vector-borne disease caused by parasitic protozoa, and is transmitted by the bite of an infectious female Anopheles mosquito. AIDS, tuberculosis and malaria each kill more than one million people a year, mainly in developing countries. Treatments are available for these infectious agents, but the cost of treatment varies widely. Treatments for TB and malaria are relatively inexpensive, but AIDS treatments can be expensive. Drug resistance can be a problem for all of these pathogens.
In some modalities, the HS system is implemented to monitor and limit the spread of infectious diseases such as AIDS, TB and malaria. In some modalities, this configuration of the Health Shield is implemented in developing countries. The general infrastructure may be similar to that described above for the flu. The data entered in the model can include pharmacokinetics and pharmacodynamics (PK / PD) data for the various drugs and the combination of drugs administered for the diseases. Tests for drug resistance can also be included in FS systems. The system can also collect information about individuals' drug therapy compliance. The system can thus estimate the optimal treatment regime for each individual. Given the profile of an individual, a person can be treated with a drug regimen designed to aggressively cure or halt the progression of the disease. Another individual may be assigned a treatment that is less ideal for performing quick healing, but will have a higher rate of adherence (eg, fewer treatments, eg, fewer pills per day) and ultimately achieve better long-term results for that individual.
FS systems can be located in the development of hot spots. Hot spots can include, for example, areas with the highest amount of infectious mosquitoes, or areas where individuals are less able to protect themselves from mosquito bites. In some embodiments, the central test zones can be built within the access points. In some embodiments, individuals without access to food may have blood samples taken and / or analyzed in a central laboratory setting that has the necessary resources. These labs can be located in or near hotspots. In some embodiments, the central laboratories are contained in mobile units that can be moved to the location of individuals.
The HS systems of the invention can be configured to provide strategies and recommendations for controlling the spread of the disease. Individuals and organizations in a hotspot or monitored area can be educated about the disease, for example, causes, treatments and methods to prevent it from spreading. In some embodiments, OS models suggest active protection measures. For example, if the system identifies an emerging tuberculosis hotspot, extra mosquito nets, bug sprays, insecticides, or anti-pesticides can be deployed to that area. Vaccines or prophylactic treatments can also be administered. In some embodiments, the model provides for areas where the infection is most likely to spread, thus allowing the initial vaccination or preferably in the areas to prevent the disease. Infected individuals or groups of individuals can be placed under supervision or quarantined. In some embodiments, they are quarantined individuals within their home, a hospital or other care facilities. In addition, the contacts of an infected person, for example, friends, family and co-workers, can be quarantined or placed under close surveillance or inspection. In some embodiments, the HS system identifies carriers, that is, individuals who carry a disease, but are not symptomatic. For example, about 80% of Africa's test population was positive for tuberculosis. In some embodiments, measures are taken to limit the spread by carriers. For example, carriers can be treated, trained on methods to reduce spreading, for example, avoiding exchange of body fluids or hygiene methods, or quarantined as appropriate. The OS system can provide estimates of the overall benefits and cost-benefit analysis of various actions to be taken.
FS systems assays can be designed to measure specific analytes for the disease or illness to be monitored. Non-limiting examples of analytes measured when monitoring AIDS, TB and malaria include HIV viruses, HIV viral RNA, IgM antibodies to HIV, IgG antibodies to HIV, CD4, CD8, and / or drug treatments. Non-limiting examples of analytes measured when monitoring tuberculosis include TB antigens, TB anti-antibodies, Mycobacterium antibodies and gamma interferon, which can rise to infection. Non-limiting examples of analytes as far as monitoring malaria include malarial antigens and anti-malaria antibodies. Various actions that can be taken when detecting AIDS analytes include, but are not limited to, the actions listed in Table 6. Table 6: AIDS analyte and action matrix

In some embodiments, the systems of the invention are used to monitor chronic, incurable infectious diseases. These diseases are more widespread through contact with infected blood and other body fluids. AIDS is currently incurable, but individuals with HIV can sometimes live for decades through the use of antiviral treatments. Transmission can be reduced by more than 80% through the correct use of condoms, the restriction of sexual partners and abstinence. Hepatitis B and C are chronic liver diseases caused by infection with the hepatitis B virus and virus C, respectively. The Health Shield of the present invention can be used to monitor the health status of people with hepatitis, in a manner similar to other infectious diseases as described herein. For example, methods of restraint in hot spots can be implemented, for example, education, and distribution of condoms can be used to stop the spread of the hepatitis C virus, which can be transmitted through sexual contact. At the individual level, infected individuals can be assigned appropriate education and therapy or interventions in the event of worsening. For example, liver damage in the final stages of hepatitis can be made worse by alcohol abuse. Infected individuals can be educated about such adverse effects of alcohol. Non-limiting examples of analytes measured when monitoring hepatitis include hepatitis B viral antigens, hepatitis C viral antigens, viral DNA hepatitis B, viral DNA hepatitis C, anti-hepatitis B antibodies surface antigen, anti-hepatitis C antibodies surface antigen, anti-hepatitis B core proteins, central anti-hepatitis C antibodies to antibodies against protein antigens. Non-limiting examples of analytes measured when monitoring liver function include aspartate transaminase (AST) or alanine transaminase (ALT). The AST / ALT ratio is sometimes useful in differentiating between the causes of liver damage when liver enzymes are elevated. For example, a proportion greater than 2.0 is more likely to be associated with alcoholic hepatitis while a ratio less than 1.0 is more likely to be associated with viral hepatitis.
Those skilled in the art will appreciate that the health shield system can be configured and adapted for the monitoring and containment of any number of infectious agents, using similar approaches as described here. The present invention includes monitoring the following non-limiting infectious agents and analytes themselves: adenovirus, whooping cough, Chlamydia Bordella pneumoiea, Chlamydia trachomatis, cholera toxin, cholera β Toxin, Campylobacter jejuni, cytomegalovirus, diphtheria toxin, Etein -Barr NA, Epstein-Barr EA, Epstein-Barr VCA virus, Helicobacter pylori, hepatitis B virus (HBV) core, hepatitis B envelope virus (HBV), surface hepatitis B virus (HBV) (Ay), hepatitis C virus (HCV) core, hepatitis C virus (HCV) NS3, hepatitis C virus (HCV) NS4, hepatitis C virus (HCV) NS5, hepatitis A, hepatitis D, hepatitis E virus (HEV) ORF2 3 KD, hepatitis E virus (HEV) ORF2 6 KD, hepatitis E virus (HEV) ORF3 3KD, human immunodeficiency virus (HIV) -1 p24, human immunodeficiency virus (HIV) -1 gp41, the immunodeficiency virus human (HIV) -1 gp120, human papilloma virus (HPV), Herpes simplex virus HSV-1/2, Herpes simplex virus HSV-1 gD, Herpes simpl ex HSV-2 gG virus, human T cell leukemia virus (HTLV) -1 I 2, Influenza A, M3 Influenza A H3N2, Influenza B, Leishmania donovani, Lyme disease, M. pneumoniae, M. tuberculosis, Parainfluenza 1, Parainfluenza 2, Parainfluenza 3, polio, respiratory syncytial virus (RSV), rubella, rubella, streptolysin O, tetanus toxin, T. pallidum 15 kd, T. pallidum p47, T. cruzi, Toxoplasma , and chickenpox Zoster. 7. Chronic Disease Monitoring and Treatment Effectiveness
In addition to monitoring infectious disease, the Health Shield makes it possible to understand an individual's trajectory and his / her disease / response to therapy. Given both the inherent genetic variance embedded in the human species and the variability of an individual's environment, the ability to monitor and control the most informative pathophysiological factors in a disease process allows us to determine whether a treatment is effective. This monitoring can help ensure that health care dollars are spent on treatments and drugs that work. With traditional laboratory systems, up to 50% of individuals do not comply with laboratory test prescriptions and as many as 60% of therapeutic prescriptions do not have the intended effects. HS offers greater adherence through home implantation and greater drug efficacy by real-time monitoring of efficacy. Because HS provides a service point for testing, it helps to facilitate the fulfillment of laboratory test orders.
In some embodiments, the integrated technologies of the invention are used to control chronic diseases, such as congestive heart failure. This monitoring can help to improve the quality of life and avoid costly hospitalizations through preventive action. For diabetic individuals, the systems can provide automated counseling that helps coordinate and manage lifestyle changes and reverses disease progression and prevents (and predicts) complications. By improving results and allowing for previous interventions, significant health savings can be achieved. In some embodiments, the same systems can be used to control interactions between drugs for chronic disease patients, having multiple therapies. This ability not only prevents adverse reactions to medications and reduces the costs of complications associated with influenza, but it also allows potentially life saving drugs to be used more widely in chronic disease populations Diabetes mellitus (diabetes) is a condition in which the body or fail to produce or respond to insulin, a hormone produced in the pancreas that allows cells to absorb glucose in order to turn it into energy. In diabetes, the body either does not respond properly to insulin, does not produce enough insulin, or both. This causes glucose to accumulate in the blood, leading to various complications. Acute complications, including hypoglycemia, diabetic ketoacidosis, or non-ketotic hyperosmolar coma can occur if the disease is not adequately controlled. Serious long-term complications include cardiovascular disease, chronic kidney failure, retinal damage and blindness, various types of nerve damage and microvascular damage, which can cause erectile dysfunction and poor wound healing. Wound healing, particularly of the feet, can lead to gangrene and possibly amputation. In type 1, diabetes, or juvenile diabetes, the body cannot produce insulin. Currently, almost everyone with type 1 diabetes should take insulin injections. Type 2 diabetes, also known as early adulthood or late-onset diabetes, results in insulin resistance, a condition in which cells fail to use insulin properly, sometimes combined with relative insulin deficiency. About 90% of Americans who are diagnosed with diabetes have type 2 diabetes. Many people destined to develop type 2 diabetes spend many years in a state of pre-diabetes, a condition that occurs when a person's blood glucose levels are higher than normal, but not high enough for the diagnosis of type 2 diabetes. As of 2009 there are 57 million Americans who have prediabetes.
Pre-diabetes has been called "America's largest health epidemic." Handelsman, Yehuda, MD. Diagnosis The doctor: pre-diabetes. Power of Prevention, Vol. 1, Issue 2, 2009. Sugar and high-fat diets are causing an earlier onset of obesity and diabetes, especially in rich countries. Young people consume a diet high in fat and sugar and become obese, which in turn can progress to serious illnesses and disorders, including but not limited to pre-diabetes, diabetes, heart disease. In many environments, easy access to carbonated drinks that contain high levels of sugar and fast high-fat foods promotes this process.
The HS system of the invention can be used to assist in the response to the spread of diabetes. In some embodiments, the system is used to identify individuals at high risk. In some modalities, the system can identify places such as, for example, geographic location, communities, school systems or schools, where the risk of disease progression is greatest. In a non-limiting example, consider HS implanted within a school. The FS system would be implemented for the school in a manner similar to that described above for infectious diseases. In some embodiments, school staff, for example, a nurse, can administer trials for all students or for a subset of students, for example, at-risk students. The test can be taken at regular intervals, for example, at least once a school year, at least once a semester, at least once a quarter, at least monthly, at least every three weeks, at least every every two weeks, or at least weekly. In some embodiments, subsets of students could be tested at different intervals. For example, the entire student body can be tested at a first frequency, and a subset of the student body, for example, those identified at risk for various factors, for example, obesity or previous test results, can be monitored at a second frequency. In a non-limiting example, the first frequency can be at least once a school year and the second frequency can be at least once a semester, at least once a quarter, or at least monthly. Any similar scheme, where risk groups are tested more often, can be used.
FS systems deployed in schools can be used to monitor a variety of analytes that are indicative of risk or disease, for example, hormone levels and glucose levels. In some embodiments, such analytes are measured in the blood. Non-limiting examples of suitable analytes that can be measured by the FS systems include glucose, hemoglobin A1c, insulin, glucagon, glucagon-like peptide 1 (GLP-1), the C-peptide insulin precursor, leptin, adiponectin, HDL cholesterol , cholesterol, LDL cholesterol and triglycerides. Other physiological data, for example, body mass, can also be entered into the system for the HS operating system component to calculate individual and group risks. The system can also monitor drug treatment, introducing a regime into an individual's health profile, or directly by detecting drug levels with FS. In some embodiments, the system monitors the progression of any or all of these variables over time.
When HS identifies an individual, for example, a student, or a population, for example, a student body, having or at risk of developing pre-diabetes or diabetes, the system may recommend a course of action. In the case of a population, the system can issue a warning and / or recommend action if the population's incidence or risk increases above a threshold level. In some modalities, the course of action comprises counseling for individuals and caregivers, or other individuals who can influence an individual's lifestyle to mitigate illness or risk. For example, parents or school officials can be notified. The system can also recommend treatments or interventions, including physical exercise, weight loss, altered eating habits, etc. For a population, a recommendation can include population control measures, including, without limitation, the removal of sodas and soft drinks sweetened from school facilities, healthy cafeteria menus, and improved physical education.
Susceptibility to type II diabetes is not only due to poor lifestyle choices, but is affected by other factors, for example, genetic factors. In the United States, such variation, for example in the Native American population and those with significant indigenous ancestry, such as the Hispanic population, are potentially at high risk. Environmental factors are also potential factors. The OS model can be extended to take into account additional factors, including, without limitation, genetic and environmental factors. For example, the model can be configured to include adaptive sampling based on non-test measures of risk. Such risk measures include, without limitation of body weight, medical history, blood pressure, family history, activity level, genetic variability, and alcohol use. The model can also be configured to adaptive sampling based on data from the FS trial in conjunction with geographic, family, demographic employment, health care provider, and other data. Likewise, the system can model the therapeutic adaptation treatment based on results for the individual and for a population that the analysis system determines to be similar for the variables that best indicate risk. The system can also incorporate visualization that helps the doctor to explain and clarify to the user their risk factors, and appropriate measures to mitigate the risk, for example, therapeutic and / or prophylactic treatments and / or interventions, weight loss, changes in diet , exercise and other lifestyle changes. Such a visualization can include, for example, a decision tree or heat map. In some embodiments, the visualization shows cumulative risk from additive factors. An exemplary use of a decision tree for diabetes is shown in Example 4. Each of these approaches can be applied to the model for diabetes and other chronic or infectious diseases.
In another embodiment, the SH's real-time and point-of-care control capability can be used to improve the efficiency of clinical trials. The time-saving impact of Escudo Saúde was quantified alongside conventional testing and data analysis by pharmaceutical companies. Modeling studies show that HS can reduce the clinical trial process for potentially a number of years and save $ 100ms per program. In addition, the data generated can provide better success and results for drugs monitored by defining patient populations and identifying possible adverse effects in a predictable way.
In a separate embodiment, a monitoring method more than a pharmacological parameter useful for assessing the efficacy and / or toxicity of a therapeutic agent is provided. For example, a therapeutic agent can include any substance that has therapeutic and / or potential utility. Such substances include, but are not limited to, biological or chemical compounds, such as simple or complex organic or inorganic molecules, peptides, proteins (for example, antibodies) or a polynucleotide (for example, antisense). A wide range of compounds can be synthesized, for example polymers, such as polypeptides and polynucleotides, and synthetic organic compounds based on different core structures, and these can also be included as therapeutic agents. In addition, several natural sources can provide compounds for therapeutic use, such as extracts from plants or animals, and the like. It should be understood, although it is not always explicitly stated that the agent is used alone or in combination with another agent, having the same different biological or activity as the agents identified by the inventive screen. The agents and methods are also intended to be combined with other therapies. For example, small molecule drugs are often measured by mass spectrometry, which can be inaccurate. ELISA (antibody-based) assays can be much more accurate and precise. Physiological parameters according to the present invention include, without limitation, parameters such as heart rate / pulse rate, blood pressure, and respiratory rate. Pharmacodynamic parameters include concentrations of biomarkers, such as proteins, nucleic acids, cells, and cell markers. Biomarkers can be indicative of disease or it can be a result of the action of a drug. Pharmacokinetic (PK) parameters according to the present invention include, without limitation drug and drug metabolite concentration. Identifying and quantifying the pharmacokinetic parameters I in real time from a sample volume is | 5 extremely desirable for the adequate safety and efficacy of drugs. If, the drug and metabolite concentrations are outside a desired range and / or। unexpected metabolites are generated due to an unexpected reaction to the drug,! immediate action may be necessary to ensure patient safety.
Likewise, if any of the pharmacodynamics (PD) parameters fall outside the desired range during a treatment regimen, immediate action may have to be taken as well.
Be able to control the rate of change of an analyte concentration or PD or PK parameters over a period of time in a single subject, or perform trend analysis of the concentration, PD, or pharmacokinetic parameters, if they are concentrations of drugs or their metabolites, can help to avoid potentially dangerous situations. For example, if glucose were the analyte of interest, the concentration of glucose in a sample at a given time, as well as the rate of change in the concentration of glucose over a given period of time can be very useful in predicting and avoiding, for example, 20 example, hypoglycemic events. Such trend analysis has broad beneficial implications for drug dosing regimens. When multiple drugs and their metabolites are involved, the ability to detect a trend and take proactive measures is often desirable.
A number of other diseases and conditions can be monitored using the HS system and methods described here. For example, the system can be used to monitor and control the spread of a microorganism, virus, or Chlamydiaceae. Exemplary microorganisms include, but are not limited to! bacteria, viruses, fungi and protozoa. Analytes that can be detected by the subject method also include blood born pathogens I 30 selected from a non-limiting group consisting of Staphylococcus epidermidis, Escherichia coli, methicillin-resistant Staphylococcus aureus (MSRA), Staphylococcus aureus, Staphylococcus hominis , Enterococcus faecalis, Pseudomonas aeruginosa, Staphylococcus capitis, Staphylococcus warned, Klebsiella pneumoniae, Haemophilus influenzae, Staphylococcus simulans, Streptococcus pneumoniae and Candida albicans.
Other microorganisms that can be detected by the subject method also encompass a variety of sexually transmitted diseases selected from the following: gonorrhea (Neisseria gorrhoeae), syphilis (Treponena pallidum) and chlamydia (Clamyda tracomitis), non-gonococcal urethritis (Ureaplasm urealyticum), yeast Infection (Candida albicans), chancroid (Haemophilus ducreyi), trichomoniasis (Trichomonas vaginalis), genital herpes (HSV type I and II), HIV I, II and HIV, hepatitis A, B, C, G, as well as hepatitis caused by TTV. Additional microorganisms that can be detected by the subject methods encompass a variety of respiratory pathogens including, but not limited to, Pseudomonas aeruginosa, Staphylococcus aureus resistant to Staphlococccus (MSRA), Klebsiella pneumoniae, Haemophilis influenzae, Staphylococcus Staphlocische, coli, Enterococcus faecalis, Serratia marcescens, Vibrio parahaemolyticus Haemophilus, Enterococcus cloacae, Candida albicans, Moraxiella catarrhalis, Streptococcus pneumoniae, Citrobacter freundii, Enterococcus faecium, Klebsella oxytoca, Pseudomonas fluorscens, meningitidis Neiseria, Streptococcus pyogenes, Pneumocystis carinii, Klebsella pneumoniae Legionella pneumophila, Mycoplasma pneumoniae and Mycobacterium tuberculosis.
Any number of biomarkers can be detected on an implanted Health Shield. Listed below are additional exemplary markers according to the present invention: Theophylline, PCR, CKMB, PSA, myoglobin, CA125, Progesterone, TxB2, 6-keto-PGF-1-alpha, and theophylline, hormone, Lutenizing estradiol, triglycerides , tryptase, Low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, IGFR. Exemplary liver markers include, without limitation, LDH, (LD5), (ALT), Arginase 1 (liver type), alpha-fetoprotein (AFP), alkaline phosphatase, alanine dehydrogenase, lactate aminotransferase and bilirubin.
Exemplary kidney markers include, without limitation TNFα receptor, Cystatin C, urinary prostaglandin D lipocalin, synthatase (LPGDS), hepatocyte growth factor receptor, polycystin 2, polycystin 1, Fibrocystin, Uromodulin, alanine, aminopeptidase, N- acetyl-protein BD-glucosaminidase, albumin and retinol (RBP).
Exemplary cardiac markers include, without limitation, troponin I (Tnl), troponin T (TnT), CK, CKMB, myoglobin, fatty acid binding protein (FABP), CRP, D-dimer, S-100 protein, BNP, NT- proBNP, PAPP -A myeloperoxidase, (MPO), glycogen phosphorylase isoenzyme BB (GPBB), activable thrombin inhibitor fibrinolysis (TAFI), fibrinogen, Modified albumin ischemia (IMA), cardiotrophin-1, and MLC-I (Light chain myosin) I) .Exemplary pancreatic markers include without limitation Amylase, Pancreatitis-Associated protein (PAP-1), and Regeneratein proteins (REG).
Exemplary muscle tissue markers include, without limitation, myostatin. Exemplary blood markers include, but are not limited to, Erythopoeitin (EPO). Exemplary bone markers include, but are not limited to, collagen-like bone N-telopeptide (NTx) cross-linking of bone collagen telopeptide, lysyl-pyridinoline (deoxypyridinoline), pyridinoline, tartrate-resistant acid phosphatase, type procollagen IC type propeptide of procollagen in propeptide, osteocalcin (bone Gla-protein), alkaline phosphatase, K cathepsin, COMP (Cartilage Oligimeric Protein Matrix), Osteocrin, osteoprotegerin (OPG), RANKL, sRANK, TRAP 5 (TRACP 5), Specific osteoblast factor 1 (OSF-1, Pleiotrophin), soluble cell adhesion molecules, sTfR, sCD4, sCD8, sCD44, and specific osteoblast factor 2 (OSF-2, Periostin).
In some embodiments, markers according to the present invention are disease specific. Exemplary cancer markers include, but are not limited to, PSA (total prostate specific antigen), creatinine, prostatic acid phosphatase, PSA complexes, Prostrate specific gene-1, CA 12-5, carcinoembryonic antigen (CEA), Alpha fetus protein (AFP ), hCG (human chorionic gonadotropin), Inhibin, ovary CAA C1824, CA 27.29, CA 15-3, CEA sinuses C1924, Her-2, pancreas, CA 19-9, pancreas CAA, neuron-specific enolase, angiostatin DcR3 (soluble decoy receptor 3), Endostatin, Ep-CAM (MK-1), Free Immunoglobulin Light Kappa Chain, Free Immunoglobulin Light Chains Lambda, Herstatin, Chromogranin A, Adrenomedulin, Integrin, Epidermal Growth Factor Receptor, Growth Factor epidermal receptor tyrosine kinase, peptide N-terminal Pro-adrenomedullin 20, vascular endothelial growth factor, vascular endothelial growth factor receptor, cell factor receptor stem, c-kit / KDR, KDR, and Midkine .
Examples of infectious disease conditions include, but are not limited to: viremia, sepsis, bacteremia, and markers: PMN elastase, PMN elastase / α1-PI complex, Surfactant protein D (SP-D), HBVc antigen, HBVs antigen, Anti-HBVc , Anti-HIV, T-suppressor cell antigen, T-cell antigen ratio, T-cell helper antigen, anti-HCV, pyrogens, p24 antigen, muramyl dipeptide.
Exemplary diabetes markers include, without limitation of C-peptide, hemoglobin A1c, glycated albumin, advanced glycosylation end products (AGE), 1,5-anhydroglucitol, gastric inhibitory polypeptide, glucose, hemoglobin, ANGPTL3 and 4.
Exemplary inflammation markers include, without limitation, TNF-D, IL-6, 1L1 □, rheumatoid factor (FR), antinuclear antibody (ANA), acute phase markers, including C-reactive protein (CRP), Clear Cell Protein (Uteroglobin).
Exemplary allergy markers include, without limitation, total IgE and specific IgE.
Exemplary autism markers include, without limitation Ceruloplasmin, Metalothioneine, Zinc, Copper, B6, B12, glutathione, alkaline phosphatase, and activation of alkaline apo-phosphatase.
Exemplary coagulation marker disorders include, without limitation b- Thromboglobulin, platelet factor 4, Von Willebrand factor.
In some embodiments, a marker may be a specific therapy. COX inhibitors include, without limitation TxB2 (Cox-1), 6-keto-PGF-1-alpha (COX 2), 11-dehydro-TxB-1a (Cox-1).
Other markers of this include, without limitation of the receptor, leptin A, leptin and procalcitonin, Brain protein S100, substance P, 8-lso-PGF-2a.
Examples of geriatric markers include, without limitation, Neuron specific enolase, GFAP, and S100B. exemplary nutritional status markers include, without limitation pre-albumin, albumin, retinol protein (RBP), transferrin, Acylation-Stimulating Protein (ASP), adiponectin, Agouti Protein Related (AgRP), angiopoietin-like protein 4 (ANGPTL4, FIAF), C-peptide, AFABP (Fatty Binding adipocyte acid protein, FABP4), Protein Stimulating Acylation (ASP), EFABP (Fatty Binding Epidermal acid protein, FABP5), Glicentin, glucagon, glucagon-like peptide-1, glucagon -Like Peptide-2, ghrelin, insulin, receptor, leptin A leptin, PYY, RELMs, resistin, amd sTfR (soluble transferrin receptor).
Examples of markers of lipid metabolism include, without limitation, Apo-lipoproteins (various), Apo-A1, Apo-B, Apo-C-CIl, Apo-D, Apo-E.
Exemplary coagulation status markers include, without limitation, Factor I: fibrinogen, Factor II: Prothrombin, Factor III: Tissue factor, factor IV: Calcium, Factor V: Proaccelerin, Factor VI, Factor VII: proconvertin, Factor VIII :, Anti -hemolytic factor, factor IX: Natal factor, X factor: Stuart-Prower factor, factor XI: Plasma antecedent thromboplastin, factor XII: Hageman factor, factor XIII: fibrin stabilizing factor, pre-kallikrein, high molecular kininogen , Protein C, Protein S, D-dimer, tissue plasminogen activator, plasminogen, a2-antiplasmin, inhibitor of plasminogen activator 1 (PAI1).
Examples of monoclonal antibodies include those for EGFR, ErbB2, and IGF1R.
Examples of tyrosine kinase inhibitors include, without limitation, Ab1, Kit, PDGFR, Src, ErbB2, ErbB 4, EGFR, EphB, VEGFR1-4, PDGFRB, flt3, FGFR, PKC, Met, TIE2, RAF, and TrkA.
Exemplary Serine / Threoline Kinas Inhibitors include, without limitation AKT, Aurora A / B / B, CDK, CDK (pan), CDK1-2, VEGFR2, PDGFRB, CDK4 / 6, MEK1-2, mTOR, and PKC-beta.
GPCR targets include, but are not limited to, Histamine Receptors, Se Rotonin Receptors, Angiotensin Receptors and Muscarinic Receptor Receptors and GnRH Receptors, Dopamine Receptors, Prostaglandin Receptors, and ADP Receptors.
Because HS comprises a number of integrated technologies that can be quickly adapted to perform additional tests, the system offers a customizable technology package that is distinct from other systems currently available. For example, systems that focus on a specific technology / application will find it difficult to be widely applied to improve outcomes and reduce health care costs across all diseases. 7. Field Systems Cartridge System (a) Field device system
Custom cartridge devices for use with the FS of the invention are described in U.S. Patent Application No. 11/389, 409, filed March 24, 2006 and entitled "point-of-care systems and their uses," USA Patent Application No. 11/746535, filed May 9, 2007 and titled "real-time detection of the influenza virus", and the US Patent Application No. 0 12/244, 723, filed October 2, 2008 and entitled "modular point attendance, devices, systems and uses THE SAME ". More details are provided here. [00.214] In one embodiment, an FS device for use with the invention comprises an automatic analyte detection device in a body fluid sample comprising an array of addressable dosing units Configured to perform a chemical reaction that produces a signal detectable indicative of the presence or absence of the analyte. In some embodiments, the device further comprises a set of addressable reagent units, each of which is directed to correspond to one or more addressable dosing units in said device, such that the individual reagent units can be calibrated with reference to the corresponding test (s) before the dies are mounted on the device. In some embodiments, at least one of the test units and at least one of the reagent units is mobile relative to each other inside the device such that the reagents for carrying out the chemical reaction are automatically brought into contact with the sample of body fluid in the dosing unit. The set of test units or reagent units can be approached according to the chemical reaction to be performed by the Configured test unit.
In one embodiment, the device is self-contained and comprises all reagents, liquid and solid phase reagents, necessary to perform a plurality of tests in parallel. When desired, the device is configured to perform at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 100, 200, 500, 1000 or more tests. One or more control tests can also be incorporated in the device to be carried out in parallel, if desired. [] 00219 The assays can be quantitative immunoassays and can be conducted in a short period of time. Another type of assay can be performed with a device of the invention, including, but not limited to, measurements of nucleic acid sequences and measurements of metabolites, such as cholesterol and enzymes such as alanine aminotransferase. In some embodiments, the test is completed in no more than an hour, preferably less than 30, 15, 10 or 5 minutes. In other embodiments, the test is performed in less than 5 minutes. The duration of the detection test can be adjusted according to the type of test that is to be performed with a device of the invention. For example, if necessary for greater sensitivity, an assay can be incubated for more than an hour or even more than a day. In some instances, trials that require a long duration may be more practical in other applications, such as POC household use, than in a clinical POC configuration.
Any body fluids suspected to contain an analyte of interest can be used in conjunction with the system or devices of the invention. Commonly used body fluids include but are not limited to blood, serum, saliva, gastric urine, and digestive fluid, tears, feces, semen, vaginal fluid, interstitial fluids derived from tumor tissue, and cerebrospinal fluid.
A body fluid can be extracted from a patient and delivered to a device in a variety of ways, including but not limited to, lancing, injecting, or pipetting. As used herein, the term subject and patient are used herein interchangeably and refer to a vertebrate, preferably a mammal, more preferably a human being. Mammals include, but are not limited to, murines and apes, humans, farm animals, sport animals, and pets. In one embodiment, a lancet punctures the skin and a sample is collected using, for example, gravity, capillary action, aspiration, or vacuum force. The lancet can be part of the device, or part of a system, or a stand alone component. Whenever necessary, the lancet can be activated by a variety of mechanical, electrical, electromechanical activation mechanisms, or any other known or any combination of such methods. In another embodiment where no active mechanism is required, a patient can simply supply a body fluid to the device, as for example, it could occur with a saliva sample. The collected fluid can be placed in the sample collection unit inside the device. In yet another embodiment, the device comprises at least one microneedle that pierces the skin.
The volume of body fluid to be used with a device is generally less than about 500 microliters, typically between about 1 to 100 microliters. When desired, a sample of 1 to 50 microliters, 1 to 40 microliters, 1 to 30 microliters, 1 to 10 microliters or even 1-3 microliters can be used for the detection of an analyte using the device.
In one embodiment, the volume of body fluid used for the detection of an analyte using the subject devices or systems is one drop of fluid. For example, a drop of blood from a pricked finger can provide the sample of body fluid to be analyzed with a system device, or the method described here.
A sample of body fluid can be collected from a subject and delivered to a device of the invention, as described below.
In one embodiment, the test matrices and reagent units are configured to be a set of mixing and matching-components. The dosage units may comprise at least one capture surface capable of reacting with an analyte from the body fluid sample. The test unit can be a tubular tip with a capture surface inside the tip. Examples of tips of the invention are described herein.
A reagent unit usually stores the liquid or solid reagents needed to perform an assay that detects an analyte. Each individual assay and reagent unit can be independently configured for the assay function. To assemble a device, the units can be assembled in a way only at the moment for use in integrated cartridges. Separate components, both in the liquid and solid phase, can be made and then tested for performance and stored. In one embodiment, the assembly of the device is carried out in on-demand fashion at a manufacturing site. The device can be modular and include components, such as a housing, which is generic for all tests, test units, such as tips, and reagent units, such as a variety of rupture or operable instrument containers that encapsulate liquid reagents. In some cases, an assembled device is then tested to define and / or verify the calibration (the relationship of the system's response to the known analyte levels). Dosing devices can be assembled from a library of prefabricated elements and calibrated on demand. In some embodiments, fluid pathways within a device can be simple and avoid any possibility of trapping bubbles and provide an efficient way to flush out excess labeled reagents in excess reagent assays, such as ELISAs. a housing for a FS device of the invention may be made of polystyrene or other moldable or machinable plastic and may have defined locations for placing test units and reagent units. In one embodiment, the housing has means for blotting tips or dosing units to remove excess liquid. The means for blotting may be a porous membrane, such as cellulose acetate, or an absorbent material such as a piece of filter paper.
In some embodiments, at least one of the components of the device can be constructed of polymeric materials. Non-limiting examples of polymeric materials include polystyrene, polycarbonate, polypropylene, polydimethysiloxanes (PDMS), polyurethane, polyvinyl chloride (PVC), polysulfone, polymethyl Imethacrylate (PM MA), acrylonitrile-butadiene-styrene (ABS), and glass.
The device or subcomponents of the device can be manufactured by a variety of methods including, without limitation, stamping, injection molding, stamping, casting, blow molding, machining, welding, ultrasonic welding, and thermal bonding. In one embodiment, a device manufactured by injection molding, thermal bonding, and ultrasonic welding. The subcomponents of the device can be affixed to each other by a thermal connection, ultrasonic welding, friction fitting (mounting press), adhesives, or, in the case of certain substrates, for example, glass, or semi-rigid and non-rigid polymeric substrates, a natural adhesion between the two components.
An exemplary device as described herein is illustrated in Figure 5. Device 100 is also sometimes referred to here as a cartridge 100. Device 100 comprises a housing 130 with locations to accommodate 121 test units and reagent units 103, 122, 124, 125. In the exemplary embodiment of Figure 5, the dosing units 121 occupy a center line of the housing 130 of the device 100. The 121 unit test can optionally include at least one calibration unit 126 In one example, dosing units 121 are similar to pipette tips and are referred to as test tips 121 and calibration units 126 are referred to as here calibration 126 tips, however, dosing units 121 can be of any form and size as they are accommodated widely by a device 100 as described herein. Dosing units 121 and 126 calibration units are exemplary test units 121 and are described in more detail. The 121-unit assay in Figure 5 can comprise a capture surface and are capable, for example, of carrying out a chemical reaction, such as nucleic acid assays and immunoassays. The 121 unit test can be mounted inside the housing according to the instructions or tests that a user wants to perform, on a sample.
As shown in Figure 5, the housing of device 100 may comprise a collection sample unit 110 Configured to contain a sample. A sample, such as a blood sample, can be placed within the sample unit collection 110. A sample tip 111 (for example, a pipette tip that pairs for a fluid transfer device, as described in more detail here) may occupy another portion of housing 130. When a test is to be performed the sample tip 111 may distribute the sample to reagent pretreatment units or pretreatment units 103, 104, 105, 106, 107, or dosing units 121. Exemplary pretreatment units 103, 104, 105, 106, 107 include, but are not limited to: mixing units 107, diluent or dilution units 103, 104, and, if the sample is a blood, plasma sample or removal of recovery units 105, 106. Pretreatment units 103, 104, 105, 106, 107 can be of the same type of unit or different types of units. Other pretreatment units 103, 104, 105, 106, 107 as needed to perform a chemical reaction can be incorporated into device 100, as would be apparent to a person skilled in the art with knowledge of the present disclosure. Units 103, 104, 105, 106, 107 can contain various amounts of reagents or diluents, flexible to whatever is necessary to perform the test on the current cartridge 100.
Often, the 121 unit test can be manufactured separately from the housing 130 and then inserted into the housing 130 with pick-and-place methods. The 121-unit assay can fit comfortably inside box 130 or it can fit freely inside box 130. In some embodiments, housing 130 is manufactured such that it holds reagent units 103, 122, 124, 125 and / or units of reagent dosing 121 firmly in place, for example when transporting or handling a cartridge. Reagents units 103, 122, 124, 125 are shown in Figure 5, which contain a conjugate reagent 122 (for example, for use with an immunoassay), a washing reagent 125 (for example, for washing said to conjugate from surfaces of capture), and a substrate 124 (e.g., an enzyme substrate). Other embodiments of the device 100 and the components in the example of Figure 5 are described herein. Reagent units 103, 122, 124, 125 can be manufactured and filled separately from housing 130 and then placed into housing 130. In this way, a cartridge of 100 can be constructed in a modular way, therefore, increase the flexibility of the cartridge 100 to be used for a variety of assays. Reagents from a reagent unit 103, 122, 124, 125 can be chosen according to the assay to be performed. Examples of reagents and assays are described here. a device, such as the example shown in Figure 5, can also comprise other characteristics that may be needed to perform a chemical reaction. For example, if the dosing units 121 are the test tips 121 as described herein, the device may comprise touch-off tip pads 112 to remove excess sample or reagent from a test tip 121 or a sample tip 111 after transferring fluids, for example, by a system as described herein. The housing 130 may also comprise units or areas 101, 102 within the device 100 for placing a used tip or unit, for example, in order to avoid cross-contamination of a tip 111 of the sample or dosing unit 121. In Figure 5, device 100 comprises a sample tip 111 for transferring a sample between units of device 100. Device 100, as illustrated in Figure 5 also comprises a pretreatment of tip 113 for transferring a sample that has been pre - treated in one unit of device 100 to other units of device 100 to perform a chemical reaction. For example, the tip 111 of the sample can be used to remove a blood sample from the sample unit collection 110 and transfer the blood sample to the pretreatment units 103, 104, 105, 106, 107, as described. Red blood cells can be removed from the blood sample in pretreatment units 103, 104, 105, 106, 107 and the pretreatment of tip 113 can then be used to collect blood plasma from the pretreatment units - treatment 103, 104, 105, 106, 107 and transferring the blood plasma to another unit (for example, a diluent unit) 103 pre-treatment, 104, 105, 106, 107 and / or to at least one test unit 121. In one embodiment, a sample tip 111 is the sampling unit 110. In another embodiment, the sampling unit 110 is similar to a well and is configured to contain a sample received by a user. assay units 121 and reagent units 103, 122, 124, 125, as shown in Figure 5 can be addressable to indicate the location of the units in cartridge 100. For example, a column of cartridge 100, as shown in Figure 5 can contain a dosing unit 121 to perform an assay to detect Configured C-reactive protein, and the column can contain corresponding reagent units 103, 122, 124, 125 for which assay in the same column, where the units are directed to match one to the other other. For example, addresses can be entered and stored on a computer system, and cartridge 100 can be given a label, such as a bar code. When the barcode of the cartridge 100 is scanned for use, the computer system can send the addresses of the units to a system, such as those described herein, to transfer the fluids and perform a reaction according to the addresses entered in the computer. The addresses can be part of a protocol sent to operate the system. The addresses can be in any configuration and can be changed if you need to change the protocol for running an assay, which in turn can offer a change in the assay protocol or steps for a user of the cartridge that has not typically been available on prior art POC devices. In some embodiments, the housing units 130 and are configured in an array of units 6 by 8, as shown in Figure 5. The layout of the units can be of any shape, for example, rectangular arrangements or random layouts. A cartridge 100 can comprise any number of units, for example, between 1 and about 500. In some embodiments, a cartridge 100 has between 5-100 units. As an example, as shown in Figure 5, cartridge 100 has 48 units.
Two-sided cross-sectional views of the exemplary device 200 of Figure 5 are shown in Figures 6A and 6B. A cavity can be molded in a housing 220 of a device to accommodate the test units (for example, test tips) in a vertical orientation (horizontal housing) with their projections towards the top of the device 200. As shown in Figure 6, a cavity can also be configured to accommodate a reagent unit 210, 212 or a sample collection unit or tip 202. There may be resources in housing 220 to accurately capture the units and hold them. Such features can also be designed to operate with a mechanism for moving the tips, such as pick-up and drop-off tips. In another embodiment, the sample collection unit comprises a flexible or breakable element that serves to protect a small collection tube during shipment and to hold a plunger device in place within a capillary. Also shown in Figure 6A are two exemplary embodiments of reagent units 210, 212 which are described herein. The bottom of the housing 220 can be configured to collect residual liquids, for example, washing the reagents after use which are transferred back through a hole in the housing 220 to the bottom. The housing 220 may comprise an absorbent pad for collecting residual fluids. The dosing units 201 and sample units 202 can be positioned to fit through a cavity in the housing 220 of the device 200 and extend beyond an internal support structure. Reagent units 210, 212 fit into the housing as shown in Figure 6 and do not extend beyond the inner support structure. Housing 220 and the areas where dosing units 201 and reagent units 210, 212 can be made and positioned can adapt to a variety of standards. .
In some embodiments, each tip provides for a single assay and can be paired with or matched to an appropriate reagent, such as the reagents needed to perform the designated assay. Some tips provide for the control test units and have known amounts of analyte attached to their capture surfaces, either in the manufacturing process or during the performance of a test. In the case of a control test unit, the unit is configured to perform a control test for comparison. The control test unit can comprise, for example, a capture and analyte surface that are in a solid or liquid state. in many embodiments, the device holds all reagents and liquids required by the assay. For example, for a luminogenic ELISA assay the reagents within the device may include a sample diluent, capture surfaces (eg, three capture antibodies), a detector conjugate (for example, three enzyme-labeled antibodies), a wash solution, and an enzyme substrate. Additional reagents can be provided as needed.
In some embodiments, the reagents can be incorporated into a device to provide for sample pre-treatment. Examples of pretreatment reagents include, without limitation, white cell lysis reagents, red blood cell lysis reagents, red blood cell removal reagents, reagents for binding factor releasing analytes, enzymes, and detergents. Pretreatment reagents can also be added to a diluent contained within the device.
An individual reagent unit can be configured to receive a mobile dosing unit. In some embodiments, the individual test unit comprises an open type hollow cylindrical element comprising a capture surface and a reaction cuvette. A cylindrical test unit can be referred to as a test tip here. In some embodiments, the individual test unit is configured to perform an immunoassay. A dosing unit 301 comprising a small tip or tubular formation is shown in Figure 7A. In some cases, the tip 301 is configured to provide a cylindrical inner catch surface 311 and a projection 321 capable of effecting with the housing of the device. In some cases, the projection 321 and the tip 301 are configured to cooperate with a mechanism for moving the tip 301, such as a system, as described herein, or for example, a fluid transfer device. A test tip 301, as shown in Figure 7A, may comprise an opening 331 at the bottom of the tip. Aperture 331 can be used for transferring fluids or reagents into and out of a dosing unit 301. In one embodiment, a dosing unit 301 as described, is or is similar to an improved pipette tip that the dosing unit 301 comprises a capture surface of 311 configured to detect an analyte in a sample.
The tip 301 can be manufactured by an injection molded process. In one embodiment, tip 301 is made of a clear polystyrene for use with chemiluminescence assays. As shown in Figure 7A, an exemplary tip 301 comprises a protrusion (shown as most of the tip 301), which can interact with a housing and can involve, for example, conical elements of a fluid transfer device and / or pipetting devices to form a tight pressure seal. Also shown in Figure 7A, the exemplary tip 301 comprises a smaller cylindrical part. In many embodiments, a test capture surface is contained within the smallest cylindrical part. The test capture surface can be anywhere inside the tip 301 or outside the tip 301. The surface of the tip 301 can be of various geometries including, but not limited to, tubular, cubic, or pyramidal. In base chemiluminescence and fluorescence tests, the 301 tip can serve as a convenient means of presenting the test product to the test optics.
Figure 7B demonstrates an exemplary sample 302 collection unit comprising a tip of 302 samples. The sample tip 302, as shown in Figure 7B can also be separated from a sample collection unit 302 and used to transfer the sample from the sample collection units to other units of a device as described herein. The sample tip, as shown in Figure 7B, comprises a protrusion 322 as described herein for tip 302 paired with a device housing and a fluid transfer device. The sample tip 302 also comprises an opening 332 to allow the transfer of fluids or samples in and out of the sample tip. In some embodiments, the 302 of the sample tip is the same shape as a 301 test tip. In other embodiments (such as those shown in
Figures 7A and 7B), the sample tip 302 is a different shape than the test tip 301.
In one embodiment, a function of a tip is to allow samples and liquid reagents to be brought into contact with the capture surface of the test unit. Movement can occur by a variety of means including, but not limited to, capillary action, aspiration, and controlled pumping. The small size of the tips allows for quick control of the temperature required for a chemical reaction. Heat transfer and / or maintenance can be accomplished by simply placing the end into a temperature-controlled block or chamber.
In some embodiments, the tip is capable of containing about 1 to 40 microliters of fluid. In a further embodiment, the tip is capable of containing about 5 to 25 microliters of fluid. In one embodiment, the tip contains 20 microliters of fluid. In some cases, a tip may contain 1 microliter of fluid or less. In other cases, a tip can contain up to 100 microliters.
When desired, the tip tip can be disposed of into an absorbent material (for example incorporated into a disposable cartridge) before introducing the next test component to avoid contamination with a small amount of sample and / or reagent. Due to physical forces, any liquid dragged to a subject tip can be held in any desired location with minimal risk of the liquid draining out, even when held in a vertical orientation.
The test unit (for example, a test tip) can be coated with reagents for the capture test before use, using similar fluids, as in the test (for example, controlled capillary or mechanical aspiration).
A capture surface (also referred to here as a reaction site) can be formed by a binding antibody or other capture reagents covalently linked or by adsorption into the assay unit. The surface can then be dried and kept in a dry condition until used in a test. In one embodiment, there is a reaction site for each analyte to be measured.
In one embodiment, the dosing unit can be moved to fluid communication with the reagent unit and / or a sample collection unit, such that a reagent or sample can interact with a reaction site, where the connected probes can detect an analyte of interest in the body fluid sample. A reaction site can then provide a signal indicative of the presence or concentration of the analyte of interest, which can then be detected by a detection device described herein.
In some embodiments, the location and configuration of a reaction site is an important element in a dosing device. Most, if not all, disposable immunoassay devices have been configured with their capture surface as an integral part of the device.
In one embodiment, a molded plastic testing unit is either commercially available or can be made by injection molding with precise shapes and sizes. For example, the characteristic dimension can be a diameter of 0.05-3 mm or it can be a length of 3 to 30 mm. The units can be coated with capture reagents and using the method similar to those used for coating microtiter plates, but with the advantage that they can be processed in large quantities by placing them in a large container, adding coating reagents and processing, using sieves, supports, and the like to recover the pieces and wash them as needed.
The dosing unit can offer a rigid support on which a reagent can be immobilized. The test unit is also chosen to provide adequate characteristics with interactions with light. For example, the test unit can be made of a material, such as functionalized glass, Si, Ge, GaAs, GAP, SiO2, SiN4, modified from silicon, or from any of a wide variety of gels or polymers, such as such as (poly) tetrafluoroethylene, (poly) vinylidenedifluoride, polystyrene, polycarbonate, polypropylene, PMMA, ABS, or combinations thereof. In one embodiment, a dosing unit comprises polystyrene. Other suitable materials can be used in accordance with the present invention. A transparent reaction site can be advantageous. In addition, in the case where there is an optically transmissive window allowing light to reach an optical detector, the surface can advantageously be opaque and / or preferably spread light.
A reagent immobilized on the capture surface can be anything useful for detecting an analyte of interest in a sample of body fluid. For example, such reagents include, without limitation, nucleic acid probes, antibodies, cell membrane receptors, monoclonal antibodies and antisera reactive with a specific analyte. Various commercially available reagents, such as a series of polyclonal and monoclonal antibodies specifically developed for specific analytes, can be used.
One skilled in the art will appreciate that there are many ways of immobilizing various reagents on a support where reaction can take place. The immobilization can be covalent or non-covalent, through a binder portion, or tie them to an immobilized portion. Exemplary non-limiting portions of binding to bind either nucleic acids or protein molecules such as antibodies to a solid support include streptavidin or avidin / biotin bonds, carbamate bonds, ester bonds, amide, thiolester, (N) -thiourea, maleimide, amino functionalized functionalized, disulfide, amide, hydrazone bonds, and among others. In addition, a silyl moiety can be attached to a nucleic acid directly to a substrate such as glass using methods known in the art. Surface immobilization can also be achieved through a poly-L-lysine tie rod, which provides a load-load coupling for the surface.
The dosing units can be dried after the last step of incorporating a capture surface. For example, drying can be carried out by passive exposure to a dry atmosphere or by using a vacuum manifold and / or applying dry, clean air through a manifold.
In many embodiments, a test unit has been designed to allow the unit to be manufactured in a high volume, fast manufacturing process. For example, tips can be assembled on a large scale to coat batch matrices from the catch surface to the inside or to the tip. In another example, tips can be placed on a table with the rotation movement band or for series processing. In yet another example, a wide variety of tips can be connected to vacuum and / or pressure varieties for simple processing.
In one embodiment, a test unit can be operatively coupled with a fluid transfer device. The fluid transfer device can be operated under automatic control, without human interaction. In test units comprising tips, the control of the installed height of a disposable liquid tip is based on the conical interference connection of the tip to the liquid dispenser. A fluid transfer device may involve the tip. In some cases, the length of a liquid immersion tip to be transferred must be known to minimize the contact of the liquid with the outside of the tip that can be controlled. In order to two or adhere a tip to the fluid transfer device a rigid stop can be molded into the bottom of the tapered connector that engages the dispenser nozzle. An airtight seal can be made by an O-ring that is half the way above the cone or at the flat bottom of the nozzle. By separating the tip sealing function from the controlled tip height, both can be adjusted separately. The modular device and fluid transfer device can allow many tests to be carried out in parallel.
The reagent units of a device can store reagents that are needed to perform a chemical reaction for the detection to give a given analyte of interest. Liquid reagents can be dispensed in small capsules that can be manufactured from a variety of materials, including, without limitation, plastic, such as polystyrene, polyethylene, or polypropylene. In some embodiments, the reagent units are cylindrical cups. Two examples of a reagent unit 401, 402 comprising a cup are shown in Figures 8A and 8B. When desired, units 401, 402 fit into cavities in a device housing. The 401 units, 402 can be sealed to the open surface to prevent spillage of reagents, 411 412 on board. In some embodiments, the seal is an aluminized plastic and can be sealed with the cup by thermal bonding. A unit can be in any form as it is necessary to contain a reagent. For example, a cylindrical-shaped reagent unit 401 is shown in Figure 8A, and the reagent unit contains a liquid reagent 411. A different reagent unit in shape 402 is illustrated in FIGURE 8B also containing a liquid reagent 412. Both units of Exemplary reagents 401, 402 comprise optional slight modifications close to the top surface that allow units 401, 402 to fit comfortably in a housing of a device as described herein.
In many embodiments of the invention the reagent units are modular. The reagent unit can be designed to allow the unit to be manufactured in a large volume, rapid manufacturing process. For example, the reagent units can be filled and sealed in a large-scale process simultaneously. The reagent units can be filled according to the type of assay or assays to be performed by the device. For example, if a user wants different assays than another user, the reagent units can be manufactured according to the preference of each user, without the need to manufacture an entire device. In another example, the reagent units can be placed on a table with the rotation movement band or for series processing.
In another embodiment, the reagent units are accommodated directly in cavities in the housing of a device. In this embodiment, a seal can be made in housing areas around the units.
Reagents according to the present invention include, without limitation, washing buffers, enzyme substrates, dilution buffers, conjugates, enzyme labeled conjugates, DNA amplifiers, sample diluents, washing solutions, pretreatment of reagent samples including additives such as detergents, polymers, chelating agents, reagent binding albumin, enzyme inhibitors, enzymes, anticoagulants, red cell binding agents, antibodies, or other materials necessary to perform an assay on a device. An enzyme-labeled conjugate can be either a polyclonal antibody or an enzyme-labeled monoclonal antibody that can produce a detectable signal after reaction with a suitable substrate. Non-limiting examples of such enzymes are alkaline phosphatase and horseradish peroxidase. In some embodiments, the reagents comprise immunoassay reagents. In general, reagents, especially those that are relatively unstable when mixed with a liquid, are confined separately to a defined region (for example, a reagent unit) within the device.
In some embodiments, a reagent unit contains about 5 microliters to about 1 milliliter of liquid. In some embodiments, the unit can contain about 20-200 microliters of liquid. In a further embodiment, the reagent unit contains 100 microliters of fluid. In one embodiment, a reagent unit contains about 40 microliters of fluid. The volume of liquid in a reagent unit may vary depending on the type of test run to be performed or the sample of body fluid provided. In one embodiment, the volumes of the reagents are not required predetermined, but must be more than a known minimum. In some embodiments, the reagents are initially stored dry and dissolved after the start of the test to be performed on the device.
In one embodiment, the reagent units can be filled using a siphon, funnel, pipette, syringe, needle, or a combination of these. The reagent units can be filled with liquid, using a filling channel and a vacuum draw channel. The reagent units can be filled individually or as part of a mass manufacturing process.
In one embodiment, an individual reagent unit comprises a different reagent as a means of isolating the reagents from each other. The reagent units can also be used to contain a washing solution or a substrate. In addition, reagent units can be used to contain a luminogenic substrate. In another embodiment, a plurality of reagents are contained within a reagent unit.
In some cases, the device configuration allows for the pre-assay calibration capability of units and reagent units prior to assembly of the subject device's disposables.
In one aspect, a FS system of the invention comprises a device comprising test units and reagent units comprising reagents (both liquid and solid phase reagents). In some embodiments, at least one of the entire device, a test unit, a reagent unit, or a combination thereof is disposable. In a system of the invention, the detection of an analyte with a device is operated by an instrument. In most embodiments, the instrument, device and method offer an automated detection system. The automatic detection system can be automated based on a defined protocol or a protocol provided to the system by a user.
In one aspect, an automated system for detecting an analyte in a body fluid sample comprises a device or cartridge, and a detection or detector assembly for detecting the detectable signal indicates the presence or absence of the analyte.
In one embodiment, the user applies a sample (for example, a measurement or an unmeasured blood sample) to the device and inserts the device into the instrument. All subsequent steps are automatic, programmed either by the instrument (hard-wired), the user, a remote or system user, or changing the operation of the instrument according to an identifier (for example, a bar code or RFID in the device).
Examples of different functions that can be performed using a system of the invention include, but are not limited to, diluting a sample, removing parts of a sample (for example, red blood cells (RBC)), the reaction of a sample in a test unit, the addition of liquid reagents to the sample and the test unit, the washing of the reagents from the sample and the dosing unit, and containing liquids during and after using the device. Reagents can be onboard the device in a reagent unit or in a reagent unit to be mounted on the device.
An automated system can detect a particular analyte in a biological sample (for example, blood) by an enzyme immunoassay (ELISA). The system is capable of multiplexing and is particularly suitable for the detection of an analyte of interest present in a small volume of a whole blood sample (for example, 20 microliters or less). The system can also detect analytes at different dilutions from a single sample, allowing different sensitivities to be tested on the same device, when desired. All reagents, materials, and waste can be contained in the system device.
In use, a sample from a subject is applied to the assembled device and an instrument is inserted from the device. In one embodiment, an instrument can begin processing the sample by some combination of removing red cells (blood sample), diluting the sample, and moving the sample to the test unit. In an embodiment with multiplexing assays, a plurality of assay units are used and a portion of the sample is moved to individual assay units in sequence or in parallel. Assays can then be performed by a controlled sequence of incubations and reagent applications to the capture surfaces.
An exemplary fluid transfer device is formed by any component capable of performing precise and precise fluid movements. Examples of components include, but are not limited to, pumps for aspirating and ejecting exactly known volumes of fluid from wells or units of the device, at least one translation phase to improve the precision and accuracy of movement within the system. The system also comprises a detector to detect a signal generated by a signal generator (such as an enzyme in contact with its substrate), in a dosing unit. Detectors include PMTs, diodes, CCD and the like. In the case of absorbance or fluorescence based assays, a light source is used. For luminescence based assays, no light source is needed in the instrument system and a PMT or an Avalanche photodiode detector can be employed. When desired, the instrument has temperature regulation to provide a temperature-regulated environment for the incubation of tests. In an embodiment of the invention, the instrument controls the temperature of the device. In a further embodiment, the temperature is in the range of about 30-40 degrees Celsius. In some embodiments, the temperature control by the system may comprise active cooling. In some cases, the temperature range is around 0-100 degrees Celsius. For example, for nucleic acid assays, temperatures up to 100 degrees Celsius can be achieved. In one embodiment, the temperature range is about 15-50 degrees Celsius. A temperature control unit of the system may comprise a thermoelectric device, such as a Peltier device.
Cartridges, devices and systems, as described here, can offer many features that are not available in existing POC systems or integrated systems analysis. For example, many POC cartridges rely on a closed fluidic system or loop to handle small volumes of liquid efficiently. The cartridges and fluid devices described herein may have open fluid movement between the cartridge units. For example, a reagent can be stored in a unit, a sample from a sample collection unit, a diluent in a diluent unit, and the capture surface can be in a test unit, where in a cartridge state, none of the units are in fluid communication with any of the other units. Using a fluid transfer device or system, as described herein, the test units do not have to be in fluid communication with one another. This can be advantageous in some configurations, because each chemical test does not interact physically or chemically with others to avoid interference due to crosstalk testing. The units can be mobile relative to each other in order to bring some units in fluid communication. For example, a fluid transfer device may comprise a head that engages a dosing unit and moves the test unit in fluid communication with a reagent unit.
The devices and systems here can provide an effective means for high throughput and real-time detection of analytes present in a body fluid from a topic. Detection methods can be used in a wide variety of circumstances, including the identification and quantification of analytes that are associated with specific biological processes and physiological conditions, disorders or stages of disorders. As such, the systems have a wide spectrum of utility in, for example, drug screening, disease diagnosis, phylogenetic classification, parental and forensic identification, disease onset and recurrence, individual response to treatment bases versus population, and therapy monitoring. The present devices and systems are also particularly useful for advancing the preclinical and clinical stage of development of therapies, improving patient compliance, monitoring ADRs associated with a prescribed medication, the development of individualized medicine, the outsourcing of drug testing. blood from the home's central laboratory or on a prescription basis, monitoring and therapeutic agents after regulatory approval or during clinical trials. The devices and systems can provide a flexible system for personalized medicine. Using the same system, a device can be modified or exchanged, along with a protocol or instructions for a programmable processor of the systems to perform a wide variety of tests as described. The systems and devices here offer many features of a lab environment on a desk top or smaller automated instrument. Because of these characteristics, the devices are particularly well suited for implantation as FS devices for the HS systems of the invention.
In some embodiments, an individual being controlled by HS is provided with a plurality of devices to be used for the detection of a variety of analytes. An individual can, for example, use different fluidic devices on different days of the week. In some embodiments of the software on the external device associating the identifier with a protocol may include a process for comparing the current day with the day on which the fluidic device is to be used based on a clinical trial, for example. In another embodiment, the individual is provided units of different reagents and test units that can be fitted into a device housing interchangeably. In yet another embodiment, as described, the individual does not need a new device for each test day, but instead, the system can be programmed or reprogrammed by downloading new instructions from, for example, an external device, such as a server. If, for example, the two days of the week are not identical, the external device can send a wireless notification to the individual using any of the methods described herein or known in the art to notify them of the appropriate device and / or appropriate instructions. to the system. This example is illustrative only and can easily be extended to, for example, notifying a subject that a fluidic device is not being used at the correct time of day. Using these methods, the FS devices can be quickly adjusted as the disease to be monitored. For example, the operating system can target the FS to test subjects immediately in contact with an index case.
In one embodiment, a cartridge as illustrated in Figure 5 comprises a variety of 20 assay units and reagent units. The assay units can comprise a capture surface according to an analyte to be detected. The test units can then be fitted with the rest of the device in a way only at the moment. In many prior art POC devices, the capture surface is an integral part of the device and, if the capture surface is incorrect or not properly formed, the entire device may malfunction.
Using a device as described herein, the capture surface and / or the test unit can be individually controlled and customized in quality regardless of the reagent units and the device box. 30 reagent units can be filled with a variety of reagents in a just-the-moment way. This provides the flexibility for the device to be customizable. In addition, the reagent units can be filled with different volumes of reagents without affecting the stability of a device or the chemical reactions being performed inside the device. Coupled with a system such as that described with a fluid transfer device, the devices and units described herein provide flexibility in the assay methods and protocols to be performed. For example, a batch of similar devices containing the same reagents can be given to a community to be monitored by the SH. After a period of monitoring, the SO identifies that the assay can be optimized by changing the dilution of the sample and the amount of reagent supplied to the assay unit. As provided herein, the assay can be altered or optimized by just changing the instructions for a programmable processor of the fluid transfer device. For example, the batch of cartridges in the patient pool had excess thinner loaded in the cartridge. The new protocol requires four times as much diluent as the previous protocol. Due to the methods and systems provided here, the protocol can be changed on the central OS server and sent to all systems to execute the methods with the devices without having to provide new devices for the patient pool. In other words, a POC device and system, as described herein, can offer much of the flexibility of a standard laboratory practice where excess reagents and often excess sample are often available. Such flexibility can be achieved without compromising the advantages of the POC test scenario or the capacity of small sample test volumes.
In some cases, in which the cartridge units are separated, the devices and systems provide flexibility in the construction of the systems described herein. For example, a cartridge can be configured to run 8 runs using an array of test units and a set of reagent units. Due to the characteristics of the cartridge, as described here, from the same box, or a casing of the same design can be used to manufacture a cartridge with up to 8 different tests than the previous cartridge. This flexibility is difficult to achieve on many other device models
POC because of the closed systems and fluid channels and therefore the devices may not be modular or as easy to assemble, as described, Currently, there is a need for the detection of more than one analyte in which the analytes are present over a wide range of concentration ranges, for example, one analyte is in the pg / ml concentration range and another is in the ug / ml concentration range. In a non-limiting example, a viral antigen can be detected in pg / ml gamma while a host antibody to that antigen is detected in the ug / ml range. See Table 4. The system, as described here, has the ability to simultaneously assay analytes that are present in the same sample in a wide range of concentrations. Another advantage of being able to detect concentrations of different analytes present in a wide range of concentrations is the ability to relate the concentration ratios of these analytes to the safety and efficacy of multiple drugs administered to a patient. For example, unexpected drug interactions can be a common cause of adverse drug reactions. A real-time, simultaneous measurement technique to measure different analytes would help to avoid the potentially disastrous consequences of adverse effects of drug interactions. This can be useful when quickly deploying drugs to control an outbreak.
Be able to control the rate of change of an analyte concentration and / or concentration of pharmacodynamics (PD) or pharmacokinetics (PK) markers over a period of time in a single subject, or perform the trend analysis of the concentration, or PD markers, or PK, if they are concentrations of drugs or their metabolites, can help you avoid potentially dangerous situations. For example, if HS is being used to monitor diabetes and glucose was the analyte of interest, the concentration of glucose in a sample at a given time, as well as the rate of change in the concentration of glucose over a given period of time can be extremely useful to predict and prevent, for example, hypoglycemic events. Such trend analysis has broad beneficial implications for drug dosing regimens. When multiple drugs and their metabolites are involved, the ability to detect a trend and take proactive measures is often desirable.
Therefore, the data generated with the use of subject fluidic devices and systems can be used to perform a trend analysis of the concentration of an analyte in a subject.
Often, multiple tests on the same cartridge may require different dilutions or pretreatments. The dilution range can be substantial between assays. Many current POC devices offer a limited range of dilution and, therefore, a limited number of tests that can potentially be performed on the POC device. However, a system and / or cartridge as described herein can offer a wide range of dilutions, for example, 1: 2-1: 10,000 due to the system's ability to serially dilute a sample. Therefore, a large number of potential tests can be performed on a single cartridge or a plurality of cartridges without modifying the detector or reading the instrument for the tests.
In one example, a system, as provided herein, is configured to run multiple (e.g., five or more) different target analyte detection assays. In order to bring the expected analyte concentration within the detection range of an immunoassay, as described here and commonly used in the POC field, a sample must be diluted for example, 3: 1, 8: 1, 10: 1, 100: 1, and 2200: 1, to perform each of the five tests. Because the fluid transfer device is capable of holding and moving the liquid within the device, serial dilutions can be performed with a system as described herein to achieve these five different dilutions and to detect all five different target analytes. As described above, the protocol for carrying out the tests is also capable of being adjusted, without modifying the device or the system.
In a traditional pipetting laboratory environment, typically larger sample volumes are used than in a POC configuration. For example, a laboratory can analyze a blood sample taken from a patient's arm at a volume in the milliliter range. In a POC configuration, many devices and users require that the process is quick, easy and / or minimally invasive, therefore small samples (in the order of a volume in the microliter range), such as one obtained by a finger prick ) are typically analyzed by a POC device. Because of the difference in the sample, current POC devices may lose flexibility in performing an assay that is provided in the laboratory environment. For example, to run multiple assays from a sample, a minimum determined volume may be required for each assay to allow accurate detection of an analyte, therefore, placing some limits on a device in a POC configuration.
In another example, a fluid transfer system and / or device as described herein provides great flexibility. For example, the fluid transfer device can be automated to move a test unit, test tip, or empty pipette from a device unit to a separate device unit, and not in fluid communication with the device. other. In some cases, this can prevent cross-contamination of the units of a device as described. In other cases, it allows the flexibility of the movement of fluids within a multi-device, as described in contact with each other according to a protocol or instructions. For example, a cartridge comprising 8 different reagent sets in 8 different reagent units can be treated and enveloped by a fluid transfer device in any order or combination as instructed by a protocol. Therefore, many different sequences can be run for any chemical reaction to run on the device. Without changing the volume of reagents in the cartridge or the type of reagents inside the cartridge, the test protocol can be modified or different, without the need for a second cartridge or a second system.
For example, a work order FS a cartridge with a specific type of capture surface and specific reagents to perform an assay to detect an analyte (for example, C-reactive protein (CRP)) in a sample. The FS worker protocol originally planned for may require 2 washing steps and 3 dilution steps. After the FS worker has received the device and the system, those at the OS site responsible for the FS implanted devices determines that the protocol must have 5 washing steps and only one dilution step. The devices and systems here can allow flexibility for this change in the protocol without having to reconfigure the device or the system. In this example, only a new protocol or instruction set is required to be sent from the OS component to the FS system's programmable processor or fluid transfer device.
In another example, a system, as provided herein, is configured to run five different target analyte detection assays, where each assay needs to be incubated at a different temperature. In many prior art POC devices, incubating multiple assays at different temperatures is a difficult task, because multiple assays are not modular and the capture surfaces cannot be moved relative to the heating device. In a system, as described herein, in which an individual test unit is configured to perform a chemical reaction, an individual test unit can be performed in an individual heating device. In some embodiments, a system comprises a plurality of heating units. In some cases, a system comprises at least many heating units as test units. Therefore, a plurality of tests can be performed as a plurality of temperatures. systems and devices as described herein can also provide a variety of quality control measures not previously available with many prior art POC devices. For example, due to the modularity of a device, the test units and reagent units can be quality controlled separately from each other and / or separately from the housing and / or separately from a transfer system or device. fluid. Examples of quality control systems and methods offered by the systems and devices are described here.
A FS system as described for use with the invention can be performed from a variety of assays, regardless of the analyte to be detected from a sample of body fluid. A protocol depending on the identity of the device can be transferred from the external OS component where it can be stored for a reader set, to allow the reader assembly to carry out the specific protocol on the device. In some embodiments, the device has an identifier (ID) that is detected or read by an identifier detector described herein. The identifier detector can communicate with a communication set via a controller, which transmits the identifier to an external device. When desired, the external device sends a protocol stored on the external device to the communication assembly based on the identifier. The protocol to be executed on the system may include instructions for the system controller to execute the protocol, including but not limited to a particular test to be performed and a detection method to be performed. Once the assay is performed by the system, a signal indicating an analyte in the body fluid sample is generated and detected by a system detection set. The detected signal can then be communicated to the communications set, where it can be transmitted to the external device for processing, including, without limitation, the analyte concentration in the sample.
In some embodiments, the identifier can be a barcode identifier with a series of black and white or reflective lines or blocks, which can be read by an identifier detector, such as a barcode reader, which are well known or a Radio Frequency Identification Tag (RFID) with an appropriate detector. Other identifiers can be a series of alphanumeric values, colors, protrusions, or any other identifier, which can be located on a device and be detected or read by an identifier detector. The identifier detector can also be a light emitting diode, which can interact with an identifier that reflects light and is measured by the identifier detector to determine the identity of a device. In some embodiments the identifier may comprise a storage or memory device and may transmit information to an identification detector. In some embodiments, a combination of techniques may be used. In some embodiments, the detector is calibrated using an optical source, such as an LED.
In one example, a sample of body fluid can be supplied to a device, and the device can be inserted into a system. In some embodiments, the device is partially inserted manually, and then a mechanical switch on the reader assembly automatically properly positions the device within the system. Any other mechanism known in the art for inserting a disc cartridge or into a system can be used. In some embodiments, manual insertion may be necessary.
In some embodiments of a method of automatically selecting a protocol to be performed on a system it comprises providing a device comprising an identifier detector and an identifier; detect the identifier; transfer said identifier for the external OS component of the systems of the present invention; and selecting a protocol to be executed in the system from a plurality of protocols on the external OS component associated with said identifier.
In one embodiment, a FS system of the invention for the automatic detection of a plurality of analytes in a body fluid sample consists of: a fluidic device (such as those described herein) comprising: a sample collection unit configured for contain the body fluid sample; a matrix of test units, in which an individual test unit of said set of dosing units is configured to perform a chemical reaction that produces a signal indicating that an individual analyte of said plurality of analytes is detected, and a set of units reagent unit, wherein an individual reagent unit of said set of reagent units contains a reagent. The system further comprises a fluid transfer device comprising a plurality of heads, wherein an individual head of the plurality of heads is configured to engage the individual test unit, and wherein said fluid transfer device comprises a programmable processor configured to direct fluid transfer from the fluid sample body from the sample collection unit and the reagent from the individual reagent unit to the individual assay unit. For example, an individual test unit comprises a reagent and is configured to perform a chemical reaction with that reagent.
In some cases, the configuration of the processor for direct effects of fluid transfer a degree of dilution of the body fluid sample in the array of dosing units to bring signals indicative of the plurality of analytes being detected within a detectable range, such that said plurality analytes are detectable with said system. In one example, the body fluid sample comprises at least two analytes that are present in concentrations that differ by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 50, or 100 orders of magnitude. In one example, the body fluid sample is a single drop of blood. In one embodiment, the concentrations of at least two analytes in a sample differ by up to 10 orders of magnitude (for example, an analyte is present in the first 0.1 pg / ml and a second analyte is present in 500 ug / ml. In another example, some protein analytes are found in concentrations greater than 100 mg / mL, which can extend the range of interest to about 12 orders of magnitude.
A degree of dilution of the body fluid sample can bring the indicative signals of at least two analytes within the detectable range. In many cases, a system further comprises a detector, such as a photomultiplier (PMT). With a photo multiplier, for example, a detectable range of the detector can be from about 10 to about 10 million counts per second. Each count corresponds to a single photon. In some cases, PMTs are not 100% efficient and the observed count rate may be slightly lower, but close to, the actual number of photons that reach the detector per unit of time. In some cases, counts are measured at about 10 intervals of about a second and the results are averaged. In some embodiments, the test ranges are 1000 - 1,000,000 counts per second when using a PMT as a detector. In some cases, counting rates as low as 100 per second and counting rates as high as 10 million are measurable. The linear response range of PMTs (for example, the range in which the count rate is directly proportional to the number of photons per unit of time) can be around 1000-3,000,000 counts per second. In one example, an assay has a detectable signal at the lower end of about 200-1000 counts per second and at the high end of about 10,000-2,000,000 counts per second. In some cases for protein biomarkers, the count rate is directly proportional to the alkaline phosphatase bound to the capture surface and also directly proportional to the analyte concentration. Other exemplary detectors include avalanche photodiodes, avalanche photodiodes arrays, arrays, CCD super-cooled CCD arrays. Many other detectors have an output that is digital, and generally proportional to the photons that reach the detector. The range detectable by sample detectors may be suitable for the detector to be used.
An individual head of a fluid transfer device can be configured to adhere to the individual test unit. The fluid transfer device can be a pipette, such as an air-displacement pipette. The fluid transfer device can be automated. For example, a fluid transfer device can further comprise a motor in communication with a programmable processor and the motor can move the plurality of heads based on a protocol from the programmable processor. As described, an individual test unit can be a pipette tip, for example, a pipette tip with a capture surface or reaction site.
Often, in a POC device, such as the systems and devices described herein, the dilution factor must be estimated and reasonably accurate. For example, in environments where inexperienced users operate the system, there must be ways to ensure correct dilution of a sample.
As described herein, a fluid transfer device can affect a degree of dilution of a sample to provide accurate test results. For example, a programmable fluid transfer device can be multi-directed to serially dilute or dilute samples, as well as providing a sample and diluent mixture. A fluid transfer device can also provide fluid movement in POC devices.
As described, the systems and devices here can activate many features and the flexibility of the laboratory environment in a POC environment. For example, samples can be collected and handled automatically at a table top size or smaller than the device or system. A common problem with POC devices is achieving different dilution scales when performing a plurality of assays, where assays can have significantly different sensitivity or specificity. For example, there may be two analytes in a sample, but one analyte has a high concentration in the sample and the other analyte has a very low concentration. As predicted, the systems and devices of this invention can dilute the sample to significantly different levels in order to detect both analytes. For example, if the analyte is in a high concentration, a sample can be serially diluted to the appropriate detection range and supplied to a capture surface for detection. In the same system or device, a sample with an analyte in a low concentration may not need to be diluted. In this way, the dosing range of the POC devices and systems provided herein can be expanded from many of the current POC devices.
A fluid transfer device can be part of a system that is a bench instrument. The fluid transfer device may comprise a plurality of heads. Any number of heads as is necessary to detect a plurality of analytes in which a sample is provided for a fluid transfer device of the invention. In one example, a fluid transfer device has about eight heads mounted on a line and separated by a distance. In one embodiment, the heads have a tapered nozzle that engages by pressure-mounted accessory with a variety of tips, such as the test unit or sample collection units as described herein. The tips may have a feature that allows them to be removed automatically by the instrument and arranged in a box on a device as described after use. In one embodiment, the test leads are clear and transparent and may look like a cuvette from within which a test is performed that can be detected by an optical detector such as a photomultiplier tube.
In one example, the programmable processor of an FS system can comprise instructions or commands and can operate a fluid transfer device according to the instructions for transferring liquid samples, either by withdrawing (to draw liquid in) or extending (to expel the liquid). liquid) a piston into a closed air space. Both the volume of air displaced and the speed of movement can be precisely controlled, for example, by the programmable processor. Mixing samples (or reagents) with diluents (or other reagents) can be achieved by aspirating components to be mixed into a common tube and then repeatedly aspirating a significant fraction of the combined liquid volume up and down at one end. Dissolving the dry reagents in a tube can be done in a similar way. The incubation of samples and liquid reagents with a capture surface to which a capture reagent (for example an antibody) is attached can be accomplished by pulling the appropriate liquid to the tip and holding it for a predetermined time. Removal of samples and reagents can be accomplished by expelling the liquid into a reservoir or an absorbent pad in a device as described. Another reagent can then be dragged to the tip according to the instructions or protocol of the programmable processor.
In one example, as illustrated in Figure 9, a liquid 1111 previously on a tip 1101 can leave a thin film 1113 inside the tip 1101 when expelled. Therefore, a system can use the action of the left (for example upper) part of the liquid of 1112 next to combing the liquid previously present 1111 from the tip 1101. The portion of the subsequent contaminated liquid with the liquid previously present 1113 can be performed inside the upper part of the tip 1101 where it does not continue to interact with the capture surface of 1102. The capture surface of 1102 can be in a defined area of the tip 1101 such that the anterior liquid 1111 does not react with the capture surface 1102, for example, as shown in Figure 9, the catch surface 1102 occupies a defined portion of the cylindrical part of the tip 1101 not extending all the way to the head of the tip. In many cases, the incubation time is short (for example 10 minutes) and the separation of the contaminated liquid area is relatively large (> 1 mm), so the diffusion or active components of the contaminated liquid portion 1113 does not occur quickly enough to react with the catch surface of 1102 during incubation. For many high-sensitivity assays, there is a requirement to remove a reagent or wash the capture surface (for example, an antibody detector that is tagged with the assay signal generator). In one example, a fluid transfer device of a system described herein can provide washing by adding additional removal and fluid transfer aspiration cycles, for example, using a washing reagent. In one example, four washing steps demonstrated that the detector unbound antibody in contact with the capture surface is reduced by a factor of more than 106 times. Any detector antibody not specifically bound to the capture surface (highly undesirable) can also be removed during this washing process.
Extending the range of an assay can be performed by diluting the sample. In POC test systems using disposable cartridges containing the diluent, there is often a practical limit to the extent of dilution. For example, if a small blood sample is obtained by pricking the finger (for example, about 20 microliters) it is to be diluted and the maximum volume of diluent that can be placed in a tube is 250 microliters, the practical dilution limit of the whole sample is about 10 times. In an example here, a system can aspirate a smaller sample volume (for example about 2 microliters) causing the maximum dilution factor to be about 100 times. For many assays, such dilution factors are acceptable, but for a PCR-like assay (as described in the examples here), there is a need to dilute the sample much more. Separation assays based on ELISA may have a limitation of the intrinsic ability of the capture surface to bind to the analyte (for example equivalent to about a few hundred ng / ml in the diluted sample for a typical protein analyte). Some analytes are present in the blood at hundreds of micrograms / ml. Even when diluted 100 times, the concentration of the analyte may be outside the calibration range. In an exemplary embodiment of a system, the device, and here fluid transfer device, multiple dilutions can be achieved by performing multiple fluid transfers from the diluent to an individual test unit or sample collection unit. For example, if the concentration of an analyte is very high, in a sample, as described above, the sample can be diluted several times until the concentration of the analyte is within an acceptable detection range. The systems and methods described here can provide accurate measurements, or estimates of dilutions, in order to calculate the original concentration of the analyte.
In one embodiment, a FS system, as described herein, can move a liquid sample and move a dosing unit. A system can comprise a heating block and a detector. In order to move a liquid sample, a system can provide aspiration, syringe, or pipette-type action. In an exemplary embodiment, a fluid transfer device for moving a liquid sample is a pipette and pipette head system. The number of pipette devices required by the system can be adjusted according to the type of substance to be analyzed and the number of tests to be performed. The actions performed by the pipette system can be automated or manually operated by a user.
Figure 10 demonstrates an example of a fluid transfer device 520 and a system of 500 as described herein. The transfer fluid device system can move eight equal or different volumes of liquid at the same time using the eight different heads 522. For example, the cartridge (or device, as described herein) 510 comprises eight dosing units 501. Units of Individual assays 501 are configured according to the type of assay to be performed inside unit 501. Individual assay units 501 may require a certain volume of sample. An individual head 522 can be used to deliver an appropriate amount of sample to an individual test unit 501. In this example, each head 522 corresponds to an individual directed dosing unit 501.
The fluid transfer device mechanism 520 can also be used to deliver the reagents from the reagent units. Different types of reagents include a conjugate solution, a washing solution, and a substrate solution. In an automated system, the 530 step on which the device 510 sits can be moved to move the device 510 in relation to the positioning of the test units 501 and 522 and the heads according to the steps necessary to complete a test as shown in Figure 10. Alternatively, heads 522 and 501 tips or fluid transfer device 520 can be moved relative to the position of device 510.
In some embodiments, a reagent is supplied in a dry form and rehydrated and / or dissolved during the test. Dry forms include lyophilized materials and films coated on and adhered to surfaces.
A FS system can comprise a support or participatory to move the dosing units or tips. A participant can include an empty set or a set designed to fit comfortably in a head of a trial unit. For example, a means for moving the tips can be moved in a similar way to the fluid transfer device heads. The device can also be moved over a phase according to the position of a participant or holder.
In one embodiment, an instrument for moving the tips is the same as an instrument for moving a sample volume, such as a fluid transfer device as described herein. For example, a sample collection tip can be fitted in a pipette head according to the projection on the collection tip. The collection tip can then be used to distribute the liquid throughout the device and the system. After the liquid has been dispensed, the collection immersion can be eliminated, and the pipette head can be fitted over a dosing unit according to the projection on the test unit. The test unit tip can then be moved from the reagent unit to the reagent unit, and the reagents can be distributed to the dosing unit according to the aspiration action or type pipette provided by the pipette head. The pipette head also
can perform mixing within a collection tip, the test unit, or reagent unit by aspiration or syringe-type action.
A FS system may comprise a heating block for heating the test or test unit and / or for controlling the test temperature. Heat can be used in the incubation step of an assay reaction to promote the reaction and shorten the time needed for the incubation step. A system may comprise a heating block configured to receive a dosing unit. The heating block can be configured to receive a plurality of dosing units from a device as described herein. For example, if 8 tests are desired to be performed on a device, the heating block can be configured to receive 8 test units. In some embodiments, the dosing units can be moved in thermal contact with a heating block with the means for moving the test units. Heating can be carried out by a heating medium known in the art.
An exemplary FS system 600, as described here, is demonstrated in Figure 11.0 System 600 comprises a translation phase 630 over which a device 610 (or the cartridge in this example) is placed manually or automatically, or a combination of both. The system 600 also comprises a heating block 640 that can be aligned with the dosing units 611 of the device 610. As shown in Figure 11, the device 610 comprises a series of 8 dosing units 611 and multiple units of corresponding reagent 612, and the heating block 640 also comprises an area of 641 for at least 8 units to be heated simultaneously. Each of the heating zones 641 can provide the same or different temperatures for each individual test unit 611 according to the type of test run to be performed or the type of analyte to be detected. System 600 also comprises a detector (such as a photomultiplier tube) 650 for detecting a signal from a representative assay unit 611 for detecting an analyte in a sample
In one embodiment, a sensor is provided to locate a test unit relative to a detector when a test is detected.
In one embodiment, the detector is a reader of a detection mounting kit box to detect a signal produced by at least one test on the device. The detection assembly can be above the device or in a different orientation with respect to the device based on, for example, the type of test to be performed and the detection mechanism to be employed. The detection assembly can be moved in communication with the test unit or the dosing unit can be moved for communication with the detection assembly.
In many cases, an optical detector is provided and used as the detection device. Non-limiting examples include a photodiode, photomultiplier tube (PMT), photon count detector, avalanche photo diode, or charge-coupled device (CCD). In some embodiments, a pin diode can be used. In some embodiments a pin diode can be coupled to an amplifier to create a detection device with sensitivity comparable to that of a PMT. Some tests can generate luminescence as described here. In some embodiments chemiluminescence is detected. In some embodiments, a detection assembly may include a plurality of fiber optic cables connected as a bundle to a CCD detector or to a PMT array. The bundle of optical fibers may consist of discrete fibers or of many small fibers fused together to form a solid bundle. Such packets of solids are commercially available and easily interfaced with CCD detectors.
A detector can also comprise a light source, such as a lamp or light-emitting diode (LED). The light source can illuminate a test in order to detect the results. For example, the assay can be a fluorescence assay or an absorbance assay, as they are normally used with nucleic acid assays. The detector may also comprise optics, to provide the light source for the test, such as a lens or optical fiber.
In some embodiments, the detection system may comprise non-optical detectors or sensors for detecting a subject specific parameter. Such sensors can include conductivity, temperature, potentiometric signals, and amperometric signals, for compounds that are oxidized or reduced, for example, 02, H2O2, and I2, or oxidizable / reducible organic compounds.
A device and system can, after manufacture, be sent to the end user, together or individually. The device or system of the invention can be packaged with an instruction manual or instructions for use. In one embodiment, the system of the invention is generic for the type of tests performed on different devices. Since the device components can be modular, a user may need only one system and a variety of devices or dosing units or reagent units to perform a multitude of assays in a point-of-care environment. In this context, a system can be used repeatedly with several devices, and it may be necessary to have sensors on the device and the system to detect such changes during transport, for example. During changes in transport pressure, or temperature can affect the performance of a number of system components present, and as such, a sensor located on the device or system can relay these changes, for example, the external device so that adjustments can be made during calibration or during data processing on the external device. For example, if the temperature of a fluidic device is changed to a certain level during transport, a sensor located on the device can detect this change and transmit this information to the system when the device is inserted into the system by the user. There may be an additional detection device in the system to perform these tasks, or such a device may be incorporated into another component of the system. In some embodiments, information can be transmitted to the system or the external device, such as the OS component of the invention, or a personal computer at an installation site. The transmission may include wireless and / or wireless connections. Likewise, a sensor in the system can detect similar changes. In some embodiments, it may be desirable to have a sensor in the transport package as well, either instead of or in addition to the system components. For example, adverse conditions that would render a test cartridge or system invalid that can be detected may include exposure to a temperature exceeding the maximum tolerable violation or the integrity of the cartridge, such as moisture penetration.
In one embodiment, the system comprises a communication set capable of transmitting and receiving information wirelessly from an external device, for example, the OS component of the present invention. Wireless communication, can use, without limitation, Wifi, Bluetooth, Zigbee, satellite, cellular or RTM technology. Various communication methods can be used, such as a wired dial-up connection to a modem, a direct link, such as a T1, ISDN or cable line. In some embodiments, a wireless connection is established using exemplary wireless networks, such as cellular, satellite, networks or pager, GPRS, or a local data transport system, such as Ethernet or Token Ring on a local area network. In some embodiments, information is encrypted before being transmitted. In some embodiments, the communication set may contain a wireless infrared communication component for sending and receiving information. The system can include integrated graphics cards to facilitate display of information.
In some embodiments, the communication assembly may have a memory or storage device, for example located RAM, in which the collected information can be stored. A storage device may be necessary if the information cannot be transmitted at a given time due, for example, to a temporary inability to connect to that of a network. The information can be associated with the device identifier on the storage device. In some embodiments, the communication setup may repeat the sending of the stored information after a certain period of time.
In some embodiments of an external device, for example, the portal component OS of the invention, communicates with the communication set within the reader assembly. An external device can wirelessly or physically communicate with the FS system, but it can also communicate with a third party, including, without limitation, an individual, medical personnel, doctors, laboratory personnel, or others in the healthcare industry. health.
An exemplary method and system is demonstrated in Figure 12. In the example in Figure 12, a patient provides a blood sample to a device as described here, and then the device is inserted into a reader, where the reader can be a table system capable of reading an analyte in the blood sample. The reader can be a system as described herein. The reader may be a bench or table top system and may be able to read a plurality of different devices as described herein. The reader or system is capable of carrying out a chemical reaction and detecting or reading the results of the chemical reaction. In the example in Figure 12, a reader is automated according to a protocol sent from an external device (for example, a server that comprises a user interface). A reader can also send the results of the chemical reaction detection to the server and user interface. In an exemplary system, the user (for example, medical personnel, such as a doctor or researcher) can view and analyze the results, as well as decide or develop the protocol used to automate the system. Results can also be stored locally (on the reader) or on the server system. The server can also host patient records, a patient diary and patient population databases.
Figure 13 illustrates the process flow of building a system for assessing an individual's medical condition according to an embodiment of the HS system disclosed herein. The patient entries of personal data and or measurements from a reader device, and / or system as described here in a database as may be present on a server, for example, the OS component. The FS system can be configured to display personal data on a patient station monitor. In some embodiments, the FS station display is interactive, and the individual can modify the data entered. The OS database contains data from other individuals who are being monitored for health protection. The HS database can also include data on individuals collected historically from public or private institutions. In some embodiments, data from others is internal data from a clinical study.
Figure 13 also illustrates the data flow of reader collection data, which includes the data from the subject to a server that is connected via a public network. The server can manipulate the data or it can simply provide the data from a user station. Patient data can also be entered into the server separately from data relating to a medical condition that is stored in a database. Figure 13 also demonstrates a user station monitor and the flow of information to medical personnel or a user. For example, using the exemplary process flow of Figure 13, a patient at home can introduce a body fluid sample into a cartridge of the invention as described herein and place it in a system or reader as described herein. The patient can read the data from the system to a patient display station and / or modify or input new data for the process flow. Patient data can then travel over a public network, such as the Internet, for example, in an encrypted format, to a server comprising a network interface and a processor, where the server is located on a central computing hub or on a clinical trial center. The server can use medical condition data to manipulate and understand user data and then send the results over a public network, as described to a user station. The user station can be in a doctor's office or laboratory and have a user station display to display test results and manipulate patient data to medical personnel. In this example, medical personnel can receive the results and analysis of a sample from a patient from a test that the patient administered at an alternative location, such as the patient's home. Other embodiments and examples of systems and system components are described herein.
The operating system component of the HS system can store protocols to run on an FS system. The protocol can be transmitted for the assembly of an FS communication system after the OS has received an identifier that indicates which device has been inserted into the FS system. In some embodiments, a protocol may be dependent on a device identifier. In some embodiments, the OS component stores more than one protocol for each field device. In other embodiments, patient information on the external device includes more than one protocol. In some cases, the OS component stores algorithms to process a photon count sent from a communication set and in some embodiments to calculate the concentration of the analyte in a sample of body fluid.
Having the FS and OS components of the systems integrated via a network connection provides a number of advantages. For example, information can be transmitted from the operating system back to not only the FS reader assembly, but to other parts or other external devices, for example, without limitation, a PDA or cell phone. Such communication can be carried out over a wireless network, as disclosed here. In some embodiments, a calculated analyte concentration or other patient information can be sent to, for example, but not limited to, medical or patient personnel. In a non-limiting example, a quarantine notice can be sent both to the infected individual and to medical personnel who can quarantine the location.
In some embodiments, data generated using subject devices and systems can be used to perform a trend analysis of the concentration of an analyte in a subject.
Another advantage, as described here, is that the test results can be substantially immediately communicated to third parties who can benefit from obtaining the results. For example, since the analyte concentration is determined in the operating system component, it can be passed on to a patient or medical personnel who may need to take further action. This may include the identification of an index case. The step of communicating to a third party can be done wirelessly, as described here, and by transmitting the data by hand from a third party device, the third party can be notified of the test results practically anytime and anywhere.
Thus, in a time-sensitive scenario, a patient can be contacted immediately from anywhere if urgent medical action may be required. Upon detecting a device based on an identifier associated with a fluidic device after being inserted into the FS system, the system allows fluidic device-specific protocols to be downloaded from an external device, for example, the OS component, and executed. In some embodiments of the OS component it can store a plurality of protocols associated with the system of or associated with a particular individual or group of individuals. For example, when the identifier is transmitted to the OS component, the software in the OS component, such as a database, can use the identifier to identify the protocols stored in the database associated with the identifier. If only one protocol is associated with the identifier, for example, the database can select the protocol and software on the external device can then transmit the protocol for the assembly of the communication system. The ability to use protocols specifically associated with a device allows any component of a device of the invention to be used with a single system, and thus, virtually any analyte of interest can be detected with a single system.
In some embodiments, several protocols can be associated with a unique identifier. For example, if it is beneficial to detect one analyte from the same individual once a week and another analyte twice a week, protocols on the external device associated with the identifier can also be associated with each different day of the week. , so that when the identifier is detected, the software on the external device can select a specific protocol that is associated with the day of the week. Such an optimized test can reduce the cost of the HS system by just performing tests according to an optimized schedule.
In some embodiments, an individual is provided with a plurality of devices to use to detect a variety of analytes. The individual may, for example, use different devices on different days of the week. In some embodiments of the operating system software associating the identifier with a protocol may include a process for comparing the current day with the day on which the device is to be used based on a clinical trial, for example. If, for example, the two days of the week are not identical, the operating system can wirelessly send a notification to the subject using any of the methods described herein or known in the art to notify that an incorrect device is in the system and also that correct device to be used that day. This example is illustrative only and can easily be extended to, for example, notifying a subject that a device is not being used at the correct time of day.
The system can also use a networking method to assess a subject's medical condition. An information transmission system may or may not include a reader for reading subject data. For example, if biomarker data is acquired by a microfluidic point-of-care device, the values assigned to the different individual biomarkers can be read by the device itself or a separate device. Another example of a reader would be a barcode system for scanning subject data that has been entered into an electronic medical record or a medical letter. Another example of a reader would consist of an electronic database recording patient data from which subjects could be obtained directly through the communications network. In this way, the effectiveness of particular drugs can be determined in real time, thereby helping to determine whether a different mitigation strategy should be put in place. (a) Field system methods
The devices described herein FS provide an effective means for real-time detection of analytes present in a body fluid from a subject. Accordingly, in one embodiment, the present invention makes use of a method of detecting an analyte in a fluid body sample comprising providing a blood sample to an FS device, allowing the sample to react within at least one dosing unit of the device, and detecting the detectable signal generated from the analyte in the blood sample. Figure 5 demonstrates an exemplary embodiment of an FS device comprising at least one dosing unit and at least one reagent unit. The test units (for example, designated as sample tips and calibrator tips in Figure 5) can contain a capture surface and the reagent units can contain items such as conjugates, washes, and substrates. The device exemplified in Figure 5 also comprises a set of blood from the sample collection tip, a plasma sample collection tip, a blood entry well, some spheres of good or plasma separation so, an off-pad touch tip or blotter, a dilution well, a diluted plasma sample well or plasma diluent well, collection of tip disposal areas.
In one embodiment, a method comprises performing an immunoenzymatic assay (ELISA). In one example, a sample is provided to a device sampling unit as described herein. The device is then inserted into a reader system, in which reader system detects the type of cartridge or device that is inserted. The reader system can then communicate with an external device, for example, the operating system component of the HS system, to receive a set of instructions or protocol that allows the reader system to perform the desired test or cartridge tests. The protocol can be sent to the programmable processor of a fluid transfer device in the reader system. In one example, the fluid transfer device engages a sample tip of the cartridge and a certain volume of the sample is chosen from the sample collection unit and moves it to a pre-treatment unit in which the red blood cells are removed. The sample plasma can then be aspirated into a plasma tip or any test tip by the fluid transfer device according to the protocol. The tip containing the plasma can then be chosen as a diluent to dilute the sample, as is necessary for the tests to be performed. Many different dilutions can be performed using serial dilutions of the sample. For example, each tip unit assay or assay can contain a sample of a different dilution. After the sample is aspirated to a test unit by the fluid transfer device, the test unit can then be incubated with the sample to allow any target analyte present to attach to the capture surface. The incubations as described in this example can be in the system or at room temperature for any period of time, for example 10 minutes, or it can be in a system heating device as described herein. The assay unit may involve a reagent unit treated with a reagent corresponding to the assay to be performed on each individual dosing unit that has a capture surface for this assay. In this example, the first reagent is a detector solution of an ELISA, for example, comprising a detector antibody such as an anti-protein labeled antibody other than that of the capture surface. The detector solution is then aspirated out of the test unit and then a washing solution can be aspirated into the dosing unit to remove any excess detector solution. Various washing steps can be used. The final reagent to be added is an enzymatic substrate that makes the detector solution bound to chemiluminesce. In some embodiments, the test results are read by a system detector, while the tip still contains the test product. In other embodiments, the enzyme substrate is expelled from the assay unit and the results of the assay are read by a system detector. At each step, as described, incubations can take place, as needed, as described herein. In this example, the entire process after placing the cartridge in the system is automated and performed by a protocol or set of instructions for the programmable system.
An exemplary method proceeds with the delivery of a blood sample for blood entry as well. The sample can then be picked up by a collection tip and inserted into the plasma separation as well. Alternatively, blood can be deposited directly into a well containing a separator blood. For example, plasma separation can be accomplished by a variety of methods, as described herein. In this example, plasma separation proceeds using magnetizable beads and antibodies to remove non-plasma blood components. The plasma can then be held by a plasma collection tip to not contaminate the sample with the entire blood collection tip. In this example, the plasma collection tip can choose a predetermined amount of diluent and dilute the plasma sample. The diluted plasma sample is then distributed to the test units (sample tips) to attach to a capture surface. The dosing units can be incubated to allow a capture reaction to be carried out. The test unit can then be used to collect a conjugate to bind with the reaction in the dosing unit. The conjugate may comprise an entity that allows the detection of an analyte of interest by a detector, such as an optical detector. Once the conjugate has been added to the test unit, the reaction can be incubated. In an exemplary method using an exemplary device of Figure 5, a reagent unit containing a wash for the conjugate is then accessed by the dosing unit (sample tip) to remove any excess conjugate that may interfere with the detection of any analyte. After washing to remove excess conjugate, a substrate can be added to the test unit for detection. In addition, in the example in Figure 5 and this method, a tip test unit calibrator can be used to perform all of the methods described in this paragraph, except sample collection and distribution. Detection and measurements using the tip calibrator dosing unit can be used to calibrate the detection and measurement of the analyte from the sample. Other processes and methods similar to those used in this example are described below.
Any body fluids suspected to contain an analyte of interest can be used in conjunction with the system or devices of the invention. For example, the well or sample collection unit in the example in Figure 5 can collect to contain any type of body fluids that commonly used include, but are not limited to blood, serum, saliva, gastric urine, and digestive fluid, tears , feces, semen, vaginal fluid, interstitial fluids derived from tumor tissue fluids extracted from tissue samples and cerebrospinal fluid. In one embodiment, the body fluid is blood and can be obtained by a finger prick. In one embodiment, the body fluid sample is a blood plasma sample. In another embodiment, the body fluid sample is an unmodified blood sample.
A body fluid can be extracted from a patient and distributed to the device in a variety of ways including, but not limited to, lancing, injecting, or pipetting. In one embodiment, a lancet pierces the skin and delivers the sample to the device, using, for example, gravity, capillary action, aspiration, or vacuum force. The lancet can be on board the device, or part of a reader assembly, or a stand alone component. Whenever necessary, the lancet can be activated by a variety of mechanical, electrical, electromechanical activation mechanisms, or any other known or any combination of such methods. In another embodiment where no active mechanism is required, an individual can simply supply a body fluid to the device, just as it could, for example, with a saliva sample. The collected fluid can be placed in a collection as well or unit of the device. In some embodiments, there is a user activated lancet and sample capillary collection inside the device.
The volume of body fluid to be used with a method or device described herein is generally less than about 500 microliters, yet it can be between about 1 to 100 microliters. When desired, a sample of 1 to 50 microliters, 1 to 40 microliters, 1 to 30 microliters, 1 to 10 microliters or even 1-3 microliters can be used for the detection of an analyte using the fluidic object device. In one embodiment, the sample is 20 microliters. A slight excess of sample can be collected during the time needed for the test, for example, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 12% , 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 95%, or 100% extra. In some embodiments, more than 100% by volume of extra sample is collected. For example, when the sample volume needed for the tests is, for example, 15 μL, the system can use a volume in the range of 16-50 μL
In one embodiment, the volume of body fluid used to detect an analyte in the field is one drop of fluid. For example, a drop of blood from a pricked finger can provide the sample of body fluid to be analyzed according to the invention.
In some embodiments, body fluids are used directly to detect the analytes present in the body fluid, without further processing. When desired, however, body fluids can be pre-treated before performing the analysis with a device. The choice of pre-treatments will depend on the type of body fluid used and / or the nature of the analyte under investigation. For example, where the analyte is present at the low level in a sample of body fluid, the sample can be concentrated using any conventional means to enrich the analyte. Methods of concentrating an analyte include, but are not limited to, drying, evaporation, centrifugation, sedimentation, precipitation amplification, and. When the analyte is a nucleic acid, it can be extracted using various lytic enzymes or chemical solutions or using nucleic acid binding resins, following the instructions provided by the accompanying manufacturers. For blood or plasma samples, the sample can be mixed with an anticoagulant, such as EDTA or heparin. These agents can be conveniently added dry. When the analyte is a molecule present in or within a cell, extraction can be carried out using lysis agents including but not limited to anticoagulants such as EDTA or heparin, a denaturing detergent such as SDS or non-denaturing detergent such as Thesit ®, sodium deoxycholate, Triton X-100, and Tween-20.
In one embodiment, a user takes a sample of body fluid with a syringe. The sample can introduce the syringe through a capillary tube. In one embodiment measuring an analyte in a blood sample, the subject performs a finger prick and touches the outer end of the glass capillary to the blood so that the blood is extracted by capillary action and fills the capillary tube with a volume. In some cases, the sample volume is known. In some embodiments, the sample volume is in the range of about 5 - 20 microliters or other volume ranges as described herein.
In another embodiment, a method and system is provided to obtain a plasma sample substantially free of red blood cells from a blood sample. When performing an assay, analytes are often contained in blood plasma, and red blood cells can interfere with the reaction.
Often, when measuring a blood sample, the analytes of interest are in serum or plasma. For clinical purposes, the reported final concentration of various blood tests often needs to relate to the concentration of blood serum or blood plasma in a diluted sample. In many cases, blood serum or blood plasma is the test medium of choice in the laboratory. Two operations may be necessary before performing an assay, dilution and removal of red blood cells. Blood samples vary significantly in proportion to the volume of the sample occupied by the red cells (the hematocrit which varies from about 20 - 60%). In addition, in a point-of-care environment when testing systems are operated by non-expert personnel, for example, a device implanted in an individual's home to be monitored by the Health Shield, the sample volume obtained may not be to which it is destined. If a change in volume is not recognized, it can lead to errors in the reported analyte concentrations.
In the related but separate embodiment, the present invention uses a method of recovering plasma from a blood sample which comprises mixing a blood sample in the presence of magnetizable particles in a sample collection unit, where the magnetizable particles comprise an antibody capture surface for binding to non-plasma portions of the blood sample, and applying a magnetic field above a plasma collection area to the mixed blood sample to suspend the non-plasma portions of the sample of blood at the top of the plasma collection zone, thus recovering the plasma from a blood sample.
In order to process blood samples, the device or system of the invention may include a magnetic reagent or object that binds to red cells and allows magnetic removal of red cells from plasma. The reagent can be supplied in lyophilized form, but it can also be present as a liquid dispersion. A reagent made up of magnetizable particles (for example, about 1 micrometer in size) can be coated with an antibody to a red cell antigen or some adapter molecule. In some embodiments, the reagent also contains antibodies not bound to red surface cell antigens, which can be unlabeled or labeled with an adapter portion (such as biotin, digoxigenin, or fluorescein). In one embodiment of the analysis of a blood sample, the red blood cells of a diluted sample co-agglutinate with the magnetizable particles aided by an antibody in the solution phase. Alternatively, a lectin that recognizes a red cell surface carbohydrate can be used as a coagglutination agent. Sometimes, combinations of red blood cell binding agents are used. Alternatively, a device of the invention may comprise a blood filter, such as a fiberglass pad, to assist in the separation of red blood cells from a sample.
When blood is mixed with a magnetic reagent, co-agglutination can occur in which many, if not all, red cells form a binder mixed with the magnetizable particles. The dissolution of the reagent and the mixing process is triggered by repeated aspiration using a tip to tip or the collection of the invention or a pipette-like tip. After the magnetizable mass is formed, the mass can be separated from the blood plasma by using a magnet to hold the mass in place as plasma is allowed to come out of the tip. In one embodiment, the plasma exits the tip by gravity in a vertical orientation, while the magnet holds the mass in place. In another embodiment, the plasma exits the tip by means of vacuum or pressure, while the mass is kept inside the tip. Plasma can be deposited within a unit well with another collection point, or assay, as described herein.
An example of a plasma separation method of the invention is demonstrated in Figures 14A through 14E. In Figure 14, a whole blood sample 901 was aspirated into a sample tip 910 as described herein, for example, in the amount of about 20 microliters. Whole blood from sample 901 is then deposited in a 920 well separation (for example, some well beads containing magnets or particles) from an example device. Figure 14B illustrates a method of suspending and mixing a magnetic reagent in the 902 whole blood sample in a fine separation (eg, magnetic granule particles and free binding molecules). Figure 14C demonstrates a 10 microliter slug of air 930 that can be used to prevent loss of the 910 tip. The whole blood sample and 902 mixed magnetic reagents are incubated for several seconds (for example, 60 to 180 seconds) to allow an agglutination reaction occurs. Undo edits
Figure 14D shows the application of a magnetic field 940 to the whole blood cell and magnetic reagent mixture 902. The magnetic field 940 can be applied by a magnetic collar 942, which is incorporated with a system or with any magnetic means known in the art. The 940 magnetic field attracts any particles that have adhered to the magnetic reagent. In this way, the 903 plasma, which does not adhere with the magnetic reagent, can be separated from the non-plasma portions of a whole blood sample.
Figure 14E shows a method of dispensing a 903 blood plasma sample, such as the magnetic separated reagent described herein, into a well or 950 of a device unit as described herein. The blood plasma sample 903 can also be distributed to a collection tip or the test unit, as well as any other type of dosing device as is obvious to a person skilled in the art. In Figure 14E, the magnetic field 940 is shown to move with the tip 910 to distribute the blood plasma sample 903. In this example, 5 to 8 microliters of plasma were removed from a whole 20 microliter blood sample. 1 to 99% of a whole blood sample can be separated plasma using a method described herein. In one embodiment, 25 to 60% of the volume of the whole blood sample is plasma that can be separated.
Other exemplary steps in a method as described can be completed. In order to move the blood plasma sample to another well or unit, a plasma capillary collection tip (which can be operated by a robotic system or any other system of the invention) collects the blood plasma sample by means of capillarity and aspiration. Another step may comprise the distribution of the plasma sample in a diluent, and the sample can then be diluted by the diluent. The diluted blood plasma sample can then be collected by the collection tip in a predetermined volume. The diluted blood plasma sample can then be mixed and distributed in a well or unit of a device to be distributed to one or a plurality of dosing units of a device of the invention. The sample can also be distributed in any other type of device, such as a microtiter plate, as would be obvious to those skilled in the art.
The example process demonstrated in Figures 14E 14A through can be used with other devices and systems, such as any of the FS devices described herein. For example, a fluid transfer tip can contain the agglutinated mass and the plasma can be deposited on a microtiter plate. Other devices and systems as would be obvious to those skilled in the art could be used to perform the separation of the blood plasma example as disclosed herein.
The body fluid sample can also be diluted in a variety of other ways, such as using a dilution capable sample collection device. The housing of the sample collection device may comprise a tube. In the tube, two movable seals can contain a volume of a diluent. In a preferred embodiment, the volume of the diluent is predetermined, for example, in the range of approximately 50 microliters to 1 milliliter, preferably in the range of about 100 microliters to 500 microliters.
In one embodiment, the FS devices of the invention are used in a method for automatically detecting a plurality of analytes in a body fluid sample comprising: providing the body fluid sample to a fluidic device, wherein the fluidic device comprises : a sample collection unit configured to contain the body fluid sample; a set of test units, wherein an individual test unit of said set of dosing units is configured to perform a chemical reaction that produces a signal indicating that an individual analyte of said plurality of analytes is detected, and a set of units of reagents, in which an individual reagent unit said matrix of reagent units contains a reagent. The method may also comprise involving the individual test unit using a fluid transfer device. Continuing the method, the body fluid sample can be transferred from the sample collection unit to the individual assay unit using the fluid transfer device and the reagent from the individual reagent unit can be transferred to the unit individual assay, thus, the reaction of the reagent with the fluid sample body to produce the signal indicative of the individual analyte of the plurality of analytes being detected. In some embodiments, the fluid transfer device comprises a plurality of heads, wherein an individual head of the plurality of heads is configured to engage the individual test unit, and wherein said fluid transfer device comprises a programmable processor configured to direct fluid transfer from the fluid sample body from the sample collection unit and the reagent from the individual reagent unit to the individual test unit,
In some cases, instructions are provided to the programmable processor, for example, by a user, an individual, or by the manufacturer. Instructions can be provided from an external device, such as a personal electronic device or, preferably, from the operating system component of the Health shield system. The instructions can direct the step of transferring the body fluid sample to the individual test unit. For example, the step of transferring the body fluid sample can affect a degree of dilution of the body fluid sample in the individual test unit to bring the analyte signal to the individual indicative of the plurality of analytes being detected within a detectable range. In some instances, the degree of dilution of the body fluid sample carries signals indicative of at least two individual analytes within a detectable range as described herein, pattern recognition techniques can be used to determine whether the detection of an analyte or an plurality of analytes by a method as described herein is within or outside a certain range. For example, detectable signals outside the reportable range can be rejected. A certain range can be established during the calibration of a fluidic device, the reagent and the test units. For example, the range is established when a device is assembled in a way just in the moment.
In some cases, if the detectable signal from an analyte, as detected with a lower dilution factor or higher degree of dilution than, for a higher dilution factor, the lower dilution result can be identified as insufficient to calculate a quantitative result. In most cases, the concentrations of an analyte in a sample, such as derivatives of signals from samples with different degrees of dilution get lower, as the degree of dilution becomes higher. If this happens, a test result can be verified. The devices described here FS provide the flexibility of quality control rules, such as those described that many POC devices cannot offer. The described FS devices offer many of the quality control features, as you would expect in a laboratory environment.
In one embodiment, a sample is diluted in a proportion that is satisfactory for both high sensitivity and low sensitivity assays. For example, a dilution ratio of the sample to the diluent may be in the range of about 1: 10,000 - 1: 1. The device can allow a sample to be diluted in separate locations or extensions. The device can also allow the sample to be serially diluted. Combining the use of serial dilution with the wide dynamic range of luminescence detection with a PMT provides for the quantification of analytes in a range of about 1000,000,000 fold. For example, for protein biomarkers the range can be from about 1 pg / ml_ to 1000 µg / ml_.
In embodiments, a sample containing an analyte for detection can be moved from a first location to a second location by aspiration, syringe action, or standard pipette. The sample can be dragged to the reaction tip by capillary action or at reduced atmospheric pressure. In some embodiments, the sample is moved to many locations, including a set of dosing units for a device of the invention and the different wells in the housing of a device of the invention. The process of moving the sample can be automated by a system of the invention, as described herein.
The dosing units and / or collection tips that contain the sample can also be moved from a first location to a second location. The process of transferring a test unit or a collection tip can be automated and carried out using a user-defined protocol.
In one embodiment, the dosing units are moved to collect reagent from a reagent unit of the invention. In many embodiments, the movement of a test unit is automated. - Aspiration, syringe, or action type pipette can be used to collect reagent from a reagent unit in a dosing unit. Once a sample has been added to a test unit that comprises a capture surface, the entire unit can be incubated for a period of time to allow a reaction between the sample and the capture surface of the test unit. The amount of time required to incubate the reaction is often dependent on the type of test run being performed. The process can be automated by a system of the invention. In one embodiment, the incubation time is between 30 seconds and 60 minutes. In another embodiment, the incubation time is 10 minutes.
A test unit can also be incubated at an elevated temperature. In one embodiment, the test unit is incubated at a temperature in the range of about 20 to 70 degrees Celsius. The test unit can be inserted into a heating block to raise the temperature of the test unit and / or the contents of the test unit.
In one embodiment of a FS method of the invention, a conjugate is added to the test unit after a sample has been added to the unit. The conjugate can contain a molecule to label an analyte captured by a capture surface in the dosing unit. Examples of conjugates and capture surface are described below. The conjugate can be a reagent contained within a reagent unit. The conjugate can be delivered to the dosing unit by aspiration, syringe action, or standard pipette. Once a conjugate has been delivered to a dosing unit, the dosing unit can be incubated to allow the conjugate to react with an analyte within the assay unit. The incubation time can be determined by the type of assay or the analyte to be detected. The incubation temperature can be any temperature appropriate for the reaction.
In another embodiment, a method of calibrating an automatic analyte detection device in a sample of body fluid is used with the FS device of the invention. A device may comprise an array of addressable assay units configured to perform a chemical reaction that produces a detectable signal indicating the presence or absence of the analyte, and an array of addressable reagent units, each of which is directed to match one or more addressable dosing units in said device, such that the individual reagent units are calibrated with reference to the corresponding test unit (s) incorporated in a complete test device. The final multiplexed device can then be assembled using the calibrated components, making the device, and a method and system using the device, the modular components. In some embodiments, calibration for the multiplexing tests is performed as above using all tests, simultaneously, in a multiplexed dosing device.
Calibration can be pre-established by measuring the performance of test reagents, such as conjugates, before the test units and the reagent unit are mounted on a device of the invention. Calibration information and algorithms can be stored on a wirelessly linked server for the test system. Calibration can be carried out in advance or a posteriori in tests performed on replicate systems in a separate location or using the information obtained when the test system is used.
In one aspect, a control material can be used in a device or system to measure or verify the extent of dilution of a sample of body fluid. For example, another issue of solid-phase assays based on such as ELISA is that an assay uses a solid-phase reagent that is difficult to quality control without destroying its function. The systems and methods here provide methods for determining the dilution achieved in a POC system using a disposable device with automated mixing and / or dilution.
In one embodiment, a method provides a retrospective analysis, for example, by using the OS component to analyze the data in real time, before reporting the results. For example, a test can be performed and a control test can be performed in parallel with the test. The control assay provides a measurement of an expected dilution of the sample. In some examples, the control assay can check the sample dilution and, therefore, the dilution of a sample for the assay or plurality of assays performed within the system can be considered accurate.
A method for measuring a volume of a liquid sample may comprise: reacting a known amount of a control analyte in a liquid sample with a reagent to produce a detectable signal indicative of the control analyte; and comparing an intensity of said detectable signal with an expected intensity of said detectable signal, where the expected intensity of said signal is indicative of an expected volume of the liquid sample, and where said comparison provides a measurement of said said volume liquid sample to be measured. In many cases, the control analyte is not present in said liquid sample, in a detectable amount.
In one embodiment, a method may further include checking the volume of said liquid sample, when measuring the sample volume is within about 50% of the expected volume of the liquid sample.
For example, a method using a FS device described herein may further comprise: reacting a sample of body fluid containing a target analyte with a reagent to produce a detectable signal indicative of the target analyte; and measuring the amount of the target analyte in the sample body fluid using an intensity of said detectable signal indicative of the target analyte and measuring said volume of said liquid sample. The liquid sample and the body fluid sample can be the same sample. In some embodiments, the control analyte does not react with the target analyte in the body fluid sample, which provides that it does not interact with the detection of the target analyte.
In some cases, the liquid sample (to be used as a control) and the body fluid sample are different liquid samples containing the analyte of interest. For example, a control liquid, such as a control solution containing a known level of control analyte. This type of control verifies that the chemistry test is working correctly.
A control analyte used to check the correct dilution of a sample can be, without limitation, labeled with fluorescein albumin, fluorescein IgG labeled, anti-fluorescein, anti-digoxigenin, labeled with digoxigenin albumin, labeled with digoxigenin IgG, biotinylated proteins, not -lgG human. Other exemplary control analytes may be obvious to a person skilled in the art. In one embodiment, the control analyte does not occur in a sample of human body fluid. In some embodiments, the control analyte is added as a liquid or in dry form to the sample.
In a POC system as described herein configured to detect a plurality of analytes within a sample, the system may be able to dilute and mix liquids. In many cases, an automated or user system can use a control test to measure the actually achieved dilution and dilution factor for which the system calibration. For example, a control analyte may never be found in the sample of interest and is dried in a reagent unit. The amount of substance to be analyzed for dry control can be known and mixed with a sample in the reagent unit. The analyte concentration can be measured to indicate the sample volume and any dilution performed on the sample.
Examples of control analytes for an immunoassay include, but are not limited to: fluorescein-labeled protein, biotinylated protein, fluorescein-labeled, AxIexaTM-labeled, rhodamine-labeled, Texas Red-labeled immunoglobulin ,. For example, labeling can be achieved by having at least two haptens attached per protein molecule. In some embodiments, 1-20 haptens are linked per protein molecule. In a further embodiment, 4-10 haptens are linked per protein molecule.
Many proteins have a large number of free amino groups to which haptens can be attached. In many cases, protein-modifying hapten are stable and soluble. In addition, haptens such as fluorescein and Texas red are large and rigid enough that antibodies with high affinity can be made (for example, a hapten is large enough to fill the antibody's binding site). In some embodiments, haptens can be linked to proteins using reagents, such as fluorescein isothocyanate, and fluorescein carboxylic acid NHS to create control analytes in which the part recognized by the assay system is hapten.
In some embodiments, one method uses dry-control analyte. In some examples, a dry control analyte prevents sample dilution and can make the control analyte more stable. Dry control analyte can be formulated so that it dissolves quickly and / or completely on exposure to a liquid sample. In some embodiments, a control analyte can be an analyte for which antibodies have high affinity. In some cases, a control analyte may be an analyte that has no cross-reaction with any component of the endogenous sample. In addition, for example, the analyte can be inexpensive and / or easy to make. In some embodiments, the control analyte is stable for the life of the device or system described herein. Examples of transporters used to create analytes with covalently linked haptens include proteins such as, but are not limited to: albumin, IgG, and casein. Exemplary polymer carriers used to create new analytes with covalently linked haptens include, but are not limited to: Dextran, Poly-vinylpyrolidone. Examples of excipients used to formulate and stabilize control analytes include, but are not limited to: sucrose, salts and buffers (such as sodium phosphate and tris chloride).
A control analyte and method as described herein can be used in a variety of ways, including the examples described herein. For example, a method can measure a volume of a sample. In some embodiments, a method measures dilution or a dilution factor or a degree of dilution of a sample. In some cases, a method provides a concentration of the analyte in a control sample. In a system or device described herein to detect a plurality of analytes, measurements of a method here using a control analyte can be used to verify or describe the measurements of target analytes. For example, a fluid transfer device with multiple heads can be used to deliver liquid to a plurality of test units, including a control unit. In some cases, it can be assumed that the amount of liquid distributed in the plurality of units is the same or similar between the individual units. In some embodiments, a method described here with a control analyte can be used to verify that the correct sample volume has been taken or used within a device or system. In another embodiment, a method verifies the correct volume of the diluent has been provided for the sample. In addition, the dilution factor or degree of dilution can also be checked. In yet another embodiment, a method with a control analyte verifies the correct volume of the diluted sample has been distributed to the plurality of units.
Figure 15 demonstrates an exemplary control assay method as described herein that comprises a known amount of control analyte. A 1010 unit prior to assembly for a cartridge can be filled with a 1.001 solution comprising a known mass of control analyte 1002. The liquid in the solution can be dried to leave control analyte 1002 in the 1010 unit. The 1010 unit can be then be inserted into a device and transported for use. When unit 1010 is used and receives a sample or diluent 1003, sample 1003 can be delivered in an expected volume and mixed with the dry control analyte 1002 inside unit 1010 to create a control solution 1004 with an expected concentration. Control solution 1004 can be optionally diluted. In one embodiment, control analyte 1002 can be detected in the same manner as a target analyte in the device. The concentration of the control analyte in the control solution 1004 is measured. The concentration measurement can be used to calculate the volume of the sample 1003 added to create the control solution 1004. In this way, a user can compare the measured volume of the sample 1003 with the expected volume of the sample 1003.
In one example, red blood cells can be removed from a blood sample. However, if some red blood cells remain, or red blood cells are not removed from a blood sample, a method with a control analyte can be used to correct the effects of red blood cells in the blood sample. . Because hematocrit can vary significantly (for example, from 20 - 60% of the total volume of a sample), the amount of an analyte in a fixed or expected volume (v) of blood can be a function of the hematocrit (H here expressed as a decimal fraction). For example, the amount of analyte with a C concentration in the plasma is C * v * (1-H). Thus, the amount of a sample with a hematocrit 0.3 is 1.4 times greater than for a sample with a hematocrit 0.5. In an exemplary embodiment, undiluted blood can be dispensed in a device as described and red cells can be removed. A concentration of the control analyte in the plasma fraction can then be measured to estimate the plasma volume of the sample and determine the hematocrit.
In some embodiments, the unbound conjugate may need to be washed from a reaction site to prevent unbound conjugates from producing inaccurate detection. The limiting step of many immunoassays is a washing step. The minimum transition leak and high sensitivity is dependent on the washout of unbound conjugate. The washing step can be severely limited in a microtiter plate format, due to the difficulty of removing the washing liquid from a well (for example, by automatic means). A test unit device can have a number of advantages in the way that liquids are handled. An advantage can be an improvement in the signal-to-noise ratio of a test.
The removal of the conjugate can be difficult if conjugates are adhered to the edges of the test units of a device if, for example, there is an excess of a washing solution. A washout of the conjugate can occur by either pushing the wash solution from above or drawing the wash solution upwards and expelling the liquid similar to loading the sample. The washing can be repeated as many times as necessary.
When using a wash buffer in an assay, the device can store the wash buffer in reagent units and the dosing unit can be put in fluid communication with the wash solution. In one embodiment, the washing reagent is capable of removing unbound reagent from the test units at about 99, 99.9, or 99.999% per wash. In general, a high washing efficiency, resulting in a high degree of reduction of undesirable background signals is preferred. Washing efficiency is typically defined by the signal ratio of a test given to the total amount of signal generated by a test with no washing steps and can be easily determined by routine experimentation. It can be generally preferred to increase the volume of the washing solution and the incubation time, but without sacrificing the signals from a given assay. In some embodiments, washing is performed with about 50 µl to about 5000 µl of wash buffer, preferably between about 50 to about 500 µl of wash buffer, for about 10 to about 300 seconds.
In addition, it may be advantageous to use several cycles of small volumes of washing solution that are separated by periods of time when no washing solution is used. This sequence allows for diffusive washing, where labeled antibodies diffuse over time into the washing solution large quantities from protected parts of the assay unit, such as the edges or surfaces where it is loosely attached and can then be removed when the solution wash is moved from the reaction site.
In many embodiments, the last step is to distribute an enzyme substrate to detect the conjugate by optical or electrical means. Examples of substrates are described below.
For example, the reagent in the individual unit of a reagent here device can be an enzyme substrate for an immunoassay. In another embodiment, the step of transferring the substrate reagent from the individual reagent unit can be repeated after a reaction at the capture site.
For example, the enzyme substrate is transferred to a reaction site and incubated. After measuring the test signal produced, the used substrate can be removed and replaced with fresh substrate and the re-measured test signal. A signal indicative of the analyte being individual can be detected using a system as described herein, from both the first and the second substrate application. The second substrate is generally the same as the original substrate. In one embodiment, the second substrate is transferred to a reaction site from a unit of a second reagent here. In another embodiment, the second substrate is transferred to a reaction site from the same reagent unit as the original substrate. The transfer of a second substrate thus creates a second reaction to produce a second signal indicative of the individual analyte. The strength of the original signal and a second strength of the second signal can be compared to calculate the final strength of the signal indicative of the individual analyte and whether the assay was conducted properly.
In one embodiment, the intensities of the multiple signals can be used for quality control of an assay. For example, if the signals differ by 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or more, the test results can be ignored.
In one embodiment, a method as described herein comprises reloading the sample and conjugate or a detector- (antibody labeled with enzyme) and either the substrate of the enzyme and the sample to rectify or confirm a test signal or to use as an internal control. For example, the reuse of a probe or unit as described can be provided to verify function and / or to add additional sample or control materials to obtain a second signal.
In some cases, a method of reloading substrate one by one unit of enzyme is activated by the ability of a system as described herein to automatically transfer samples and liquid reagents into the test units. Some tests do not require the system to provide a result immediately or on a schedule, so a control method as described offers an opportunity to possibly increase the reliability of the results. An observed response following the iterations of adding an enzyme substrate can be used to verify the initial response or to calculate spike recovery.
Experiments have shown that by adding a second aliquot of enzyme substrate to a dosing unit, the reproducibility of the results can be maintained. In some embodiments, a control method provides replicated analyzes using a test unit gave a response significantly less than expected.
With the control methods described here, there are several possible errors that can be accounted for or postulated from the execution of a control method. Exemplary test errors include, but are not limited to, inadequate production of a test apparatus or device, improper aspiration of a sample and / or one or more reagents, a test unit is not positioned correctly in relation to the photomultiplier during detection, and a test unit missing from the device or system.
In some embodiments, a method of automatically monitoring compliance with an individual's medical treatment using the subject devices or systems is provided using the FS devices. The method comprises the steps of allowing a sample of body fluid to react with the assay reagents in a device to produce a detectable signal indicating the presence of an analyte in said sample; detecting said signal with said device, comparing said signal with a known profile associated with said medical treatment to determine whether the individual is compatible or incompatible with said medical treatment, and notifying individual or associated individuals, for example, agents local health officials, compliance or non-compliance said. This can be important for the HS systems of the invention, because mitigation policies will not be as effective if the recommended treatments are not followed. In some embodiments, non-adherence events are reported to the OS systems. The model can be updated to meet non-compliance. Employees monitoring the results of modeling the operating system can also contact local authorities to take action.
In another embodiment, the system and methods of the invention can identify trends in the levels of biomarkers and daily patient information over time that can be used to adjust a drug dose to an optimal level for particular patients (eg, adaptive dose - varying). In some embodiments, they may include abandonment having an incorrect dose of a pharmaceutical agent, including, without limitation, multiple doses or no doses, or may improperly include the mixture of pharmaceutical agents. In preferred embodiments a patient is notified substantially immediately after the signal is compared to a known profile.
An individual monitored by Health Protection may forget to take a sample of body fluid for analysis, as described here. In some embodiments of a method of alerting an individual to test a sample of body fluid with a device as described herein comprises providing a protocol to be performed on said device, said protocol communicated from the OS component, associated with said device individual, and comprising a time and date for testing said body fluid sample; and notifying individual to test said body fluid at said date and time, if said sample has not been tested. In some embodiments an individual can be notified as described here, for example, via a wireless connection. Compliance with therapeutic regimens can be improved through the use of instructions on a display and responses obtained from patients (for example, via a touch screen).
In one embodiment, the system includes a convenient way to package the elements required for multiple FS complex tests in a safe manner for transportation. For example, the dosing elements click to fit a housing.
Field system tests A variety of tests can be performed on a fluidic device described herein to detect an analyte of interest in the sample. A wide variety of labels are available in the art that can be used for conducting subject essays. In some embodiments, labels are detectable by spectroscopic, photochemical, biochemical, electrochemical, immunochemical, or other means. For example, useful nucleic acid tags include radioisotopes 32P, 35S, C14, H3, 1125, and 1131, fluorescent dyes, electron-dense reagents, and enzymes. A wide variety of labels suitable for labeling biological components are known and are reported extensively in both scientific and patent literature, and are generally applicable to the present invention for labeling biological components. Suitable labels include enzyme radionucleotides, substrates, cofactors, inhibitors, fluorescent halves, chemiluminescent halves, labels, colorimetric bioluminescent labels or redox labels. Reagents define specificity of the assay, optionally including, for example, monoclonal antibodies, polyclonal antibodies, proteins, nucleic acid probes or other polymers, such as affinity matrices, carbohydrates or lipids. Detection can proceed by any of a variety of known methods, including spectrophotometric or optical monitoring of radioactive, fluorescent, or luminescent markers, or other methods that discriminate a molecule based on charge, size or affinity. A detectable portion can be of any material having a detectable physical or chemical property. Such detectable markers have been well developed in the field of gel electrophoresis, column chromatography, solid substrates, spectroscopic techniques, and the like, and in general, labels useful in such methods can be applied with the present invention. Thus, a label includes, without limitation, any composition detectable by spectroscopic, photochemical, biochemical, nucleic acid, immuno-chemical, electrical, optical, thermal, or by chemical means.
In some embodiments of the label, it is coupled directly or indirectly to a molecule to be detected such as a product, substrate, or enzyme, according to methods well known in the art. As indicated above, a wide variety of labels are used, with the choice of label depending on the required sensitivity, ease of conjugation of the compound, the requirements for stability and available instrumentation, and disposal arrangements. Non-radioactive labels are often associated by indirect means. Generally, a specific receptor for the analyte is attached to a signal generation portion. Sometimes the analyte receptor is attached to an adapter molecule (such as biotin or avidin) and the assay reagent set includes a binding portion (such as a biotinylated reagent or avidin) that binds to the adapter and stops the analyte. The analyte binds to a specific receptor at the reaction site. A labeled reagent can form a sandwich complex in which the analyte is at the center. The reagent can also compete with the analyte for receptors at the reaction site or bind to unoccupied receptors at the reaction site not occupied by the analyte. The tag is inherently detectable or linked to a signal system, such as a detectable enzyme, a fluorescent compound, a chemiluminescent compound, or a chemiluminogenic entity such as an enzyme with a luminogenic substrate. A number of ligands and anti-ligands can be used. Whenever a ligand has a natural anti-ligand, it can be used in conjunction with labeled, anti-ligands. Exemplary ligand - anti-even ligands include, without limitation, biotin - avidin thyroxine, - anti-T4, digoxigenin - anti-digoxin, cortisol and - anti-cortisol, alternatively, any haptenic or antigenic compound can be used in combination with an antibody .
In some embodiments the label can also be conjugated directly to signal generating compounds, for example, by conjugation with an enzyme or fluorophore. Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases. Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl groups, and umbelliferone. Chemiluminescent compounds include dioxetanes, acridinium esters, luciferin, and 2,3-dihydrophthalazinediones, such as luminol. label detection methods are well known to those skilled in the art. Thus, for example, where the label is radioactive, means for detection include scintillation counting or photographic films as in autoradiography. When the label is fluorescent, the fluorochrome can be detected by excitation with light of an appropriate wavelength and detect the resulting fluorescence by, for example, visual inspection microscopy, using photographic film, using electronic detectors, such as digital cameras, charging coupled devices (CCD) or photomultipliers and phototubes, or other detection device. Likewise, enzyme tags are detected by providing appropriate substrates for the enzyme and detecting the resulting reaction product. Finally, simple colorimetric labels are often detected simply by observing the color, that is, the absorbance, associated with the label. For example, conjugated gold often appears pink, while several conjugated beads appear the color of the bead.
In some embodiments, the detectable signal can be provided by luminescence sources. Luminescence is the term commonly used to refer to the emission of light from a substance for any reason other than an increase in its temperature. In general, atoms or molecules emit photons of electromagnetic energy (for example, light), when they then move from an animated state to a lower energy state (usually the ground state). If a photon is exciting, the luminescence process is referred to as photoluminescence. If the exciting cause is an electron, the luminescence process can be referred to as electroluminescence. More specifically, the electroluminescence results from direct injection and removal of electrons to form an electron-hole pair, and subsequent recombination of the electron-hole pair to emit a photon. Luminescence that results from a chemical reaction is commonly referred to as chemiluminescence. Luminescence produced by a living organism is commonly referred to as bioluminescence. If photoluminescence is the result of a spin transition allowed (for example, a single singlet transition, the triplet © triplet- transition), the photoluminescence process is usually referred to as fluorescence. Typically, fluorescence emissions do not persist after the exciting cause is removed as a result of short-lived excited states that can quickly relax through such permissible spin transitions. If photoluminescence is the result of a prohibited rotation transition (for example, a singlet-triplet transition), the photoluminescence process is usually referred to as phosphorescence. Typically, phosphorescence emissions persist for a long time after the exciting cause is removed as a result of long-lived excited states that can relax only through such forbidden rotation-transitions. A luminescent label can have any of the properties described above. Suitable chemiluminescent sources include a compound that becomes electronically excited through a chemical reaction and can then emit light that serves as the detectable signal or energy donates to a fluorescent acceptor. A diverse number of families of compounds have been found to provide chemiluminescence under a variety or conditions. A family of compounds is 2,3-dihydro-1,4-phthalazinedione. A frequently used compound is luminol, which is a 5-amino compound. Other members of the family include 5-amino-6, 7,8-trimethoxy-dimethylamino and analog [ca] benz. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium and base hypochlorite. Another family of compounds is 2,4,5-triphenylimidazoles, with lophine as the common name for the main product. Chemiluminescent analogs include para-dimethylamino-methoxy and substituents. Chemiluminescence can also be obtained with oxalates, usually oxalyl esters, for example, p-nitrophenyl and a peroxide such as hydrogen peroxide, under basic conditions. Other useful chemiluminescent compounds that are also known include acridinum-N-alkyl esters and dioxetanes. Alternatively, luciferins can be used in conjunction with luciferase or lucigenins to provide bioluminescence.
The term analytes as used herein include, without limitation, prodrug drugs, pharmaceutical agents, drug metabolites, such as biomarkers, expressed proteins and cell markers, antibodies, serum proteins, cholesterol and other metabolites, polysaccharides, nucleic acids, biological analytes, biomarkers , genes, proteins, or hormones, or any combination of these. Analytes can be combinations of polypeptides, glycoproteins, polysaccharides, lipids and nucleic acids.
Of particular interest are biomarkers that are associated with a particular disease or with a specific disease stage. Such analytes include, but are not limited to, those associated with infectious diseases, autoimmune diseases, obesity, hypertension, diabetes, neuronal and / or muscle degenerative diseases, heart disease, endocrine disease, metabolic disorders, inflammation, cardiovascular disease, septicemia, angiogenesis , cancers, Alzheimer's disease, complications from athletics, and any combinations of them.
Of interest are also biomarkers that are present in different abundance in one or more of the body's tissues, including heart, liver, prostate, lung, kidney, bone marrow, blood, skin, bladder, brain, muscles, nerves and selected tissues that are affected by various diseases, such as different types of cancer (malignant or non-metastatic), autoimmune diseases, inflammatory or degenerative diseases
References Brandeau, M.L., G.S. Zaric, and A. Richter. 2003. Resource Allocation for Control of Infectious Disease in Multiple Independent Populations: Beyond Cost-Effectiveness Analysis. J. Health Econ 22: 575-598 Chiang, C.L. 1978. An Introduction to Stochastic Processes and Their Applications. Kreiger. 517 pages. Choi, BCK and AWP Pak. 2003. A simple approximate mathematical model to predict the number of severe acute respiratory syndrome cases and deaths. J Epidemiol Community Health. 57 (10): 831-5 D’Onofrio, A. 2002. Stability Properties of Pulse Vaccination Strategy in SEIR Epidemic Model. Math. Biosci. 179: 52-72. Dwyer, G. J.S.Elkinton, and J.P. Buonaccorsi. 1997. Host heterogeneity in Susceptibility and Disease Dynamics: Tests of a mathematical Model. Am Naturalist. 150: 685-707 FitzGibbon, W.E., M.E.Parrot, and G.F. Webb. 1995. Diffusion Epidemic Models with Incubation and Crisscross Dynamics. Math. Biosci. 128: 131-155. Gibson, G.J. 1997. Investigating mechanisms of Spatiotemporal Epidemic Spread Using Stochastic Models. Am Phytopathological Society. 87: 139-146 Inaba, H. 1990. Threshold and Stability Results for an Age-structured Epidemic Model. J Math Biol. 28: 411-434. 5 Longini, I.M., S.K. Seaholm, E Ackerman, J.S. Koopman, and A.S. Monto. 1984. Simulation Studies of Influenza Epidemics: Assessment of Parameter Estimation and Sensitivity. Int J Epidemiology. 13: 496-501 McKendrick, A. 1926. Applications of Mathematics to Medical Problems. Proc Edin. Math. Soc. 44: 98-130. io
权利要求:
Claims (17)
[0001]
1. System to model a disease progression within a population characterized by comprising: a plurality of point of care devices, POC, each comprising at least one programmable processor to perform at least one test according to at least one protocol test, in which the test protocol is configured to be executed by the POC devices; a central computing device operatively in data communication with said plurality of POC devices, wherein the POC devices are positioned in distributed locations physically separate from the central computing device; a static database component that comprises static data related to the disease and / or the population; a dynamic database component comprising data about the population and individual subjects, where the data in the dynamic database component comprises an indication of the disease status of individuals in the population; and a computer modeling component that is configured to generate a model of disease progression in the population based on data in (1) the static database component and (2) the dynamic database component, modeling results ; wherein said at least one test protocol when changing the central computing device based on at least the said results model, is sent from the central computing device to said plurality of POC devices for executing a test protocol changed by the devices without having to provide new devices to the distributed locations of the population,
[0002]
2. System for modeling a disease progression within a population according to claim 1, characterized by the fact that the disease is a serious or chronic disease infection.
[0003]
3. System for modeling a disease progression within a population according to claim 2, characterized by the fact that the disease is a serious disease infection caused by a microorganism, microbe, virus, a bacterium, an arche, a protozoan, a protista, a fungus or microscopic plant.
[0004]
4. System for modeling a disease progression within a population according to claim 2, characterized by the fact that the disease is a chronic disease or condition selected from the group consisting of diabetes, pre-diabetes, insulin resistance, metabolic disorder , obesity and cardiovascular diseases.
[0005]
5. System for modeling a disease progression within a population according to claim 1, characterized by the fact that the static database with the client understands information about individuals in the population composed of one or more age, race , sex, location, genetic factors, single nucleotide polymorphisms (SNPs), family history, history of disease or therapeutic history.
[0006]
6. System for modeling a disease progression within a population according to claim 1, characterized by the fact that the static database with component comprises information about the disease, in which the information about the disease comprises one or more viruses persistence, contagiousness, mode of transmission, available treatment, vaccine availability, mortality rate, recovery time, treatment cost, infectiousness, dissemination rate, mutation rate and past outbreak.
[0007]
7. System to model a disease progression within a population according to claim 1, characterized by the fact that the data in the dynamics of the database component is updated in real time.
[0008]
8. System for modeling a disease progression within a population according to claim 1, characterized by the fact that the biomarker is measured in a sample of the individual's body fluid.
[0009]
9. System for modeling a disease progression within a population according to claim 8, characterized by the fact that the body fluid with blood pressure, plasma, serum, sputum, urine, feces, semen, mucous lymphatic saliva or nasal wash .
[0010]
10. System for modeling a disease progression within a population according to claim 1, characterized by the fact that the point of care device performs one or more cartridge tests, real-time PCR, rapid antigen tests, culture viral and immunoassays.
[0011]
11. System for modeling a disease progression within a population according to claim 1, characterized by the fact that the point of care device is positioned in one or more schools, workplaces, shopping centers, community centers, institutions church, hospital, health clinic, mobile unit or home.
[0012]
12. System for modeling a disease progression within a population according to claim 1, characterized by the fact that computer modeling the component is configured to provide one or more courses of action based on the results of the modeling, in which a or more courses of action are classified according to a classification parameter.
[0013]
13. System to model a disease progression within a population according to claim 12, characterized by the fact that the classification parameter includes financial considerations, number of individuals affected, quality-adjusted life year (QALY) and / or quality adjusted life year (QALY) per unit of economic cost.
[0014]
14. System for modeling a disease progression within a population according to claim 12, characterized by the fact that one or more actions that include a strategy to control the spread of the disease.
[0015]
15. System for modeling a disease progression within a population according to claim 14, characterized by the fact that the strategy to control the spread of the disease comprises one or more quarantines of households, individual quarantine, geographical quarantine, detachment social, hospitalization, school closure, closure of the workplace, travel restrictions, closure of public transportation, prophylactic treatment or vaccination intervention, provision of protective clothing, provision of masks and additional testing at the point of care.
[0016]
16. System for modeling a disease progression within a population according to claim 1, characterized by the fact that computer modeling the component is configured to estimate a surveillance strategy based on the modeling results and on which the strategy Surveillance comprises determining the disease status of an individual or group of individuals using a point-of-care device.
[0017]
17. System for modeling a disease progression within a population according to claim 1, characterized by the fact that the data model comprises a plurality of states, in which the plurality of states one or more susceptible individuals, early exposures.
类似技术:
公开号 | 公开日 | 专利标题
US11158429B2|2021-10-26|Integrated health data capture and analysis system
CN101535499B|2017-04-19|Point-of-care fluidic systems and uses thereof
JP7023913B2|2022-02-22|Integrated health data acquisition and analysis system
US20210151156A1|2021-05-20|Microfluidic testing system for mobile veterinary applications
Class et al.2015|Patent application title: INTEGRATED HEALTH DATA CAPTURE AND ANALYSIS SYSTEM Inventors: Elizabeth A. Holmes | Elizabeth A. Holmes | Ian Gibbons Ian Gibbons Daniel L. Young | Seth G. Michelson |
AU2013201509A1|2013-04-04|Point-of-care fluidic systems and uses thereof
Moise et al.2015|In Silico Vaccine Design: Accelerating the Response to BioThreats and Emerging Infectious Disease
RU2391897C2|2010-06-20|Method and system of personalised treatment of infectious and multifactorial diseases
AU2015234373A1|2015-10-29|Point-of-care fluidic systems and uses thereof
同族专利:
公开号 | 公开日
AU2010308329A1|2012-05-31|
AU2010308329B2|2016-10-13|
US9460263B2|2016-10-04|
JP5743288B2|2015-07-01|
EP3859746A1|2021-08-04|
EP2491499A1|2012-08-29|
HK1176440A1|2013-07-26|
CA2778270C|2021-01-05|
IL219324D0|2012-06-28|
US11158429B2|2021-10-26|
JP2018121652A|2018-08-09|
CN105808956A|2016-07-27|
CN105740641A|2016-07-06|
CN102713914B|2016-03-30|
KR20120093972A|2012-08-23|
CA2778270A1|2011-04-28|
AU2016247105A1|2016-11-03|
KR20180078345A|2018-07-09|
CA3081708A1|2011-04-28|
RU2012121204A|2013-11-27|
EP2491499A4|2016-05-18|
CN105825049A|2016-08-03|
WO2011049886A1|2011-04-28|
JP2020030841A|2020-02-27|
JP2013508859A|2013-03-07|
KR101875858B1|2018-07-06|
US11195624B2|2021-12-07|
JP2021073599A|2021-05-13|
MY165876A|2018-05-18|
JP2016197118A|2016-11-24|
IL219324A|2017-02-28|
US20180308585A1|2018-10-25|
MX2012004620A|2012-06-25|
US20150100345A1|2015-04-09|
US20170053091A1|2017-02-23|
NZ624935A|2016-01-29|
AU2016247105B2|2018-05-10|
US20110093249A1|2011-04-21|
JP2015045658A|2015-03-12|
RU2628051C2|2017-08-14|
US8862448B2|2014-10-14|
MX344735B|2017-01-05|
BR112012009196A2|2017-03-01|
US11139084B2|2021-10-05|
US20180374582A1|2018-12-27|
NZ599873A|2014-09-26|
CN102713914A|2012-10-03|
SG10201502531SA|2015-05-28|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

US4146029A|1974-04-23|1979-03-27|Ellinwood Jr Everett H|Self-powered implanted programmable medication system and method|
US4003379A|1974-04-23|1977-01-18|Ellinwood Jr Everett H|Apparatus and method for implanted self-powered medication dispensing|
US4347176A|1980-04-14|1982-08-31|Burroughs Wellcome Co.|Compounds and methods of making same|
GB8422876D0|1984-09-11|1984-10-17|Secr Defence|Silicon implant devices|
US4920213A|1985-06-20|1990-04-24|Biotechnology Research Partners, Ltd.|Method and compositions useful in preventing equine influenza|
JPS6247555A|1985-08-23|1987-03-02|Hoffmann La Roche|Scintillation proximity determination method|
US4731726A|1986-05-19|1988-03-15|Healthware Corporation|Patient-operated glucose monitor and diabetes management system|
US4935346A|1986-08-13|1990-06-19|Lifescan, Inc.|Minimum procedure system for the determination of analytes|
US4946795A|1987-08-27|1990-08-07|Biotrack, Inc.|Apparatus and method for dilution and mixing of liquid samples|
US4910131A|1987-12-23|1990-03-20|Mellman Ira S|Idiotype and anti-idiotype antibodies useful in virus detection|
US5130238A|1988-06-24|1992-07-14|Cangene Corporation|Enhanced nucleic acid amplification process|
US5162237A|1988-04-11|1992-11-10|Miles Inc.|Reaction cassette for preforming sequential analytical assays by noncentrifugal and noncapillary manipulations|
US5104813A|1989-04-13|1992-04-14|Biotrack, Inc.|Dilution and mixing cartridge|
US5089229A|1989-11-22|1992-02-18|Vettest S.A.|Chemical analyzer|
US6176962B1|1990-02-28|2001-01-23|Aclara Biosciences, Inc.|Methods for fabricating enclosed microchannel structures|
US5242606A|1990-06-04|1993-09-07|Abaxis, Incorporated|Sample metering port for analytical rotor having overflow chamber|
US5061381A|1990-06-04|1991-10-29|Abaxis, Inc.|Apparatus and method for separating cells from biological fluids|
US5122284A|1990-06-04|1992-06-16|Abaxis, Inc.|Apparatus and method for optically analyzing biological fluids|
US5527670A|1990-09-12|1996-06-18|Scientific Generics Limited|Electrochemical denaturation of double-stranded nucleic acid|
DE69125441T2|1990-09-28|1997-11-06|Toshiba Kawasaki Kk|Gene detection method|
TW279133B|1990-12-13|1996-06-21|Elan Med Tech|
DE4041905A1|1990-12-27|1992-07-02|Boehringer Mannheim Gmbh|TEST CARRIER ANALYSIS SYSTEM|
US5455166A|1991-01-31|1995-10-03|Becton, Dickinson And Company|Strand displacement amplification|
US5273905A|1991-02-22|1993-12-28|Amoco Corporation|Processing of slide mounted material|
US5264184A|1991-03-19|1993-11-23|Minnesota Mining And Manufacturing Company|Device and a method for separating liquid samples|
US5173193A|1991-04-01|1992-12-22|Schembri Carol T|Centrifugal rotor having flow partition|
US5279607A|1991-05-30|1994-01-18|The State University Of New York|Telemetry capsule and process|
FR2679661B1|1991-07-26|1994-10-14|Sfri|APPARATUS FOR AUTOMATIC SAMPLES ANALYSIS.|
EP0541340B1|1991-11-05|1997-07-16|The Perkin-Elmer Corporation|Biopolymer synthesis apparatus and method|
US5270184A|1991-11-19|1993-12-14|Becton, Dickinson And Company|Nucleic acid target generation|
US5288390A|1992-03-30|1994-02-22|Sun Company, Inc. |Polycyclic aromatic ring cleavage process|
WO1993019827A1|1992-04-02|1993-10-14|Abaxis, Inc.|Analytical rotor with dye mixing chamber|
EP0639223B1|1992-05-01|1996-07-03|The Trustees Of The University Of Pennsylvania|Microfabricated sperm handling devices|
US5744366A|1992-05-01|1998-04-28|Trustees Of The University Of Pennsylvania|Mesoscale devices and methods for analysis of motile cells|
US5380487A|1992-05-05|1995-01-10|Pasteur Sanofi Diagnostics|Device for automatic chemical analysis|
US5357953A|1992-05-21|1994-10-25|Puritan-Bennett Corporation|Measurement device and method of calibration|
US5318557A|1992-07-13|1994-06-07|Elan Medical Technologies Limited|Medication administering device|
EP0723146B1|1992-09-14|2004-05-06|Sri International|Up-converting reporters for biological and other assays using laser excitation techniques|
US5674698A|1992-09-14|1997-10-07|Sri International|Up-converting reporters for biological and other assays using laser excitation techniques|
US5899855A|1992-11-17|1999-05-04|Health Hero Network, Inc.|Modular microprocessor-based health monitoring system|
US7584108B2|1996-12-23|2009-09-01|Health Hero Network, Inc.|Network media access control system for encouraging patient compliance with a treatment plan|
US5478750A|1993-03-31|1995-12-26|Abaxis, Inc.|Methods for photometric analysis|
US5578269A|1993-06-11|1996-11-26|Ortho Diagnostic Systems Inc.|Automated blood analysis system with an integral centrifuge|
EP0631137B1|1993-06-25|2002-03-20|Edward W. Stark|Glucose related measurement method and apparatus|
JP3343156B2|1993-07-14|2002-11-11|アークレイ株式会社|Optical component concentration measuring apparatus and method|
GB9315671D0|1993-07-29|1993-09-15|Dow Corning Sa|Foam control agents and their use|
AU7563294A|1993-08-24|1995-03-21|Metrika Laboratories, Inc.|Novel disposable electronic assay device|
CA2129787A1|1993-08-27|1995-02-28|Russell G. Higuchi|Monitoring multiple amplification reactions simultaneously and analyzing same|
US5397709A|1993-08-27|1995-03-14|Becton Dickinson And Company|System for detecting bacterial growth in a plurality of culture vials|
US5591643A|1993-09-01|1997-01-07|Abaxis, Inc.|Simplified inlet channels|
US6235531B1|1993-09-01|2001-05-22|Abaxis, Inc.|Modified siphons for improved metering precision|
JP3391862B2|1993-10-05|2003-03-31|株式会社日立製作所|Chromatogram analysis method|
JPH07120393A|1993-10-13|1995-05-12|Nippon Tectron Co Ltd|Fluorescence detection method|
EP0892446B1|1993-11-02|2006-05-24|Matsushita Electric Industrial Co., Ltd.|Method of manufacturing an aggregate of semiconductor micro-needles and method of manufacturing a semiconductor device comprising an aggregate of semiconductor micro-needles|
US5403415A|1993-11-17|1995-04-04|Abaxis, Inc.|Method and device for ultrasonic welding|
JPH07151101A|1993-11-29|1995-06-13|Kazuo Sugimura|Vessel having spiral diaphragm contact surface|
JP3504750B2|1993-12-22|2004-03-08|オルソ−クリニカルダイアグノスティクス,インコーポレイティド|Recalibration method of calibration relational expression and quantitative test kit|
JPH07196314A|1993-12-28|1995-08-01|Maruo Calcium Co Ltd|Tubular synthetic inorganic fine particle|
US5590052A|1994-04-14|1996-12-31|Abaxis, Inc.|Error checking in blood analyzer|
US5648211A|1994-04-18|1997-07-15|Becton, Dickinson And Company|Strand displacement amplification using thermophilic enzymes|
JP3584990B2|1994-05-09|2004-11-04|タカラバイオ株式会社|Anti-human influenza virus antibody|
US5624850A|1994-06-06|1997-04-29|Idetek, Inc.|Immunoassays in capillaries|
US5976896A|1994-06-06|1999-11-02|Idexx Laboratories, Inc.|Immunoassays in capillary tubes|
US5639428A|1994-07-19|1997-06-17|Becton Dickinson And Company|Method and apparatus for fully automated nucleic acid amplification, nucleic acid assay and immunoassay|
US5891734A|1994-08-01|1999-04-06|Abbott Laboratories|Method for performing automated analysis|
JP3839524B2|1995-06-07|2006-11-01|アジレント・テクノロジーズ・インク|Miniaturized total analysis system|
JP3652424B2|1994-10-27|2005-05-25|日本政策投資銀行|Automatic analyzer and method|
IE72524B1|1994-11-04|1997-04-23|Elan Med Tech|Analyte-controlled liquid delivery device and analyte monitor|
JP3563140B2|1995-01-19|2004-09-08|株式会社日立製作所|Capillary array electrophoresis device|
FI101829B|1995-03-07|1998-08-31|Erkki Juhani Soini|Biospecific assay method|
US5557596A|1995-03-20|1996-09-17|Gibson; Gary|Ultra-high density storage device|
US6340588B1|1995-04-25|2002-01-22|Discovery Partners International, Inc.|Matrices with memories|
US5961923A|1995-04-25|1999-10-05|Irori|Matrices with memories and uses thereof|
US6352854B1|1995-04-25|2002-03-05|Discovery Partners International, Inc.|Remotely programmable matrices with memories|
US6319668B1|1995-04-25|2001-11-20|Discovery Partners International|Method for tagging and screening molecules|
US5874214A|1995-04-25|1999-02-23|Irori|Remotely programmable matrices with memories|
US5832296A|1995-04-26|1998-11-03|Interval Research Corp.|Wearable context sensitive user interface for interacting with plurality of electronic devices of interest to the user|
US6168948B1|1995-06-29|2001-01-02|Affymetrix, Inc.|Miniaturized genetic analysis systems and methods|
JP3927570B2|1995-07-31|2007-06-13|プレシジョン・システム・サイエンス株式会社|container|
US6299839B1|1995-08-31|2001-10-09|First Medical, Inc.|System and methods for performing rotor assays|
JPH0968533A|1995-08-31|1997-03-11|Brother Ind Ltd|Biochemical substrate measuring device displaying chemical dose|
JP3515646B2|1995-09-18|2004-04-05|大塚電子株式会社|Multi-capillary electrophoresis device|
JPH09113511A|1995-10-18|1997-05-02|Kdk Corp|Dry test piece for measuring glyco-albumin|
US5687716A|1995-11-15|1997-11-18|Kaufmann; Peter|Selective differentiating diagnostic process based on broad data bases|
US5854033A|1995-11-21|1998-12-29|Yale University|Rolling circle replication reporter systems|
JPH09192218A|1996-01-16|1997-07-29|Hitachi Ltd|Blood-sugar level control system|
DE19601487C2|1996-01-17|2001-09-13|Micronas Gmbh|Device for treating malignant tissue changes|
US5863502A|1996-01-24|1999-01-26|Sarnoff Corporation|Parallel reaction cassette and associated devices|
CN1096185C|1996-01-27|2002-12-11|三星电子株式会社|Interlaced-to-progressive conversion apparatus and method using motion and spatial correlation|
US5670375A|1996-02-21|1997-09-23|Biomerieux Vitek, Inc.|Sample card transport method for biological sample testing machine|
DE69721242T2|1996-03-01|2004-01-29|Biota Scient Management|METHOD FOR DETECTING INFLUENZA VIRUS AND COMPOUNDS THAT CAN BE USED|
JP2988362B2|1996-03-11|1999-12-13|株式会社日立製作所|Multi-sample analysis system|
JPH09244055A|1996-03-14|1997-09-19|Hitachi Ltd|Liquid crystal display device|
US5801057A|1996-03-22|1998-09-01|Smart; Wilson H.|Microsampling device and method of construction|
JP2783277B2|1996-03-27|1998-08-06|日本電気株式会社|Patient monitoring device and patient monitoring system|
US5716852A|1996-03-29|1998-02-10|University Of Washington|Microfabricated diffusion-based chemical sensor|
JPH09281078A|1996-04-09|1997-10-31|Hitachi Electron Eng Co Ltd|Dna base sequence determining apparatus|
JP3213566B2|1996-04-26|2001-10-02|アークレイ株式会社|Sample analysis tool, sample analysis method and sample analyzer using the same|
US5980830A|1996-05-20|1999-11-09|Sendx Medical, Inc.|Portable modular blood analyzer with simplified fluid handling sequence|
US6108635A|1996-05-22|2000-08-22|Interleukin Genetics, Inc.|Integrated disease information system|
IL118432A|1996-05-27|1999-12-31|Yissum Res Dev Co|Electrochemical and photochemical electrodes and their use|
US5939291A|1996-06-14|1999-08-17|Sarnoff Corporation|Microfluidic method for nucleic acid amplification|
FI2607U1|1996-06-17|1996-09-27|Nokia Mobile Phones Ltd|An additional unit designed to be connected to a digital cordless telephone|
JP3788519B2|1996-06-28|2006-06-21|カリパー・ライフ・サイエンシズ・インコーポレーテッド|High-throughput screening assay system for microscale fluid devices|
US5942443A|1996-06-28|1999-08-24|Caliper Technologies Corporation|High throughput screening assay systems in microscale fluidic devices|
US5797898A|1996-07-02|1998-08-25|Massachusetts Institute Of Technology|Microchip drug delivery devices|
EP0818547A1|1996-07-10|1998-01-14|Autoliv ASP, Inc.|Recovery of metals values from air bag inflators|
US5874046A|1996-10-30|1999-02-23|Raytheon Company|Biological warfare agent sensor system employing ruthenium-terminated oligonucleotides complementary to target live agent DNA sequences|
GB9624096D0|1996-11-20|1997-01-08|Microbial Systems Ltd|Apparatus and method of use thereof|
US6071249A|1996-12-06|2000-06-06|Abbott Laboratories|Method and apparatus for obtaining blood for diagnostic tests|
US6514461B1|1997-02-14|2003-02-04|Escreen, Inc.|System for automatically testing a fluid specimen|
JPH10239240A|1997-02-25|1998-09-11|Hitachi Ltd|Automatic dna probing apparatus|
US7255833B2|2000-07-25|2007-08-14|Cepheid|Apparatus and reaction vessel for controlling the temperature of a sample|
US6369893B1|1998-05-19|2002-04-09|Cepheid|Multi-channel optical detection system|
DK2333520T3|1997-02-28|2014-09-29|Cepheid|Heat-exchanging, requested chemical reaction device|
US5951300A|1997-03-10|1999-09-14|Health Hero Network|Online system and method for providing composite entertainment and health information|
JP3393361B2|1997-03-24|2003-04-07|国立身体障害者リハビリテーションセンター総長|Biosensor|
US6235471B1|1997-04-04|2001-05-22|Caliper Technologies Corp.|Closed-loop biochemical analyzers|
US5961451A|1997-04-07|1999-10-05|Motorola, Inc.|Noninvasive apparatus having a retaining member to retain a removable biosensor|
US5885470A|1997-04-14|1999-03-23|Caliper Technologies Corporation|Controlled fluid transport in microfabricated polymeric substrates|
DE19717023C2|1997-04-23|2003-02-06|Micronas Gmbh|Device for treating malignant, tumorous tissue areas|
CN1105914C|1997-04-25|2003-04-16|卡钳技术有限公司|Microfluidic devices incorporating improved channel geometries|
US6406845B1|1997-05-05|2002-06-18|Trustees Of Tuft College|Fiber optic biosensor for selectively detecting oligonucleotide species in a mixed fluid sample|
JPH10305016A|1997-05-08|1998-11-17|Casio Comput Co Ltd|Behavior information providing system|
WO1999004043A1|1997-07-14|1999-01-28|Abbott Laboratories|Telemedicine|
US7475965B2|1997-07-15|2009-01-13|Silverbrook Research Pty Ltd|Inkjet printer with low droplet to chamber volume ratio|
US5876675A|1997-08-05|1999-03-02|Caliper Technologies Corp.|Microfluidic devices and systems|
US7914994B2|1998-12-24|2011-03-29|Cepheid|Method for separating an analyte from a sample|
US6368871B1|1997-08-13|2002-04-09|Cepheid|Non-planar microstructures for manipulation of fluid samples|
JPH1157560A|1997-08-27|1999-03-02|Shin Meiwa Ind Co Ltd|Sprinkler truck|
US6174675B1|1997-11-25|2001-01-16|Caliper Technologies Corp.|Electrical current for controlling fluid parameters in microchannels|
WO1999017119A1|1997-09-26|1999-04-08|University Of Washington|Simultaneous particle separation and chemical reaction|
US5842787A|1997-10-09|1998-12-01|Caliper Technologies Corporation|Microfluidic systems incorporating varied channel dimensions|
FI107080B|1997-10-27|2001-05-31|Nokia Mobile Phones Ltd|measuring device|
WO1999033559A1|1997-12-24|1999-07-08|Cepheid|Integrated fluid manipulation cartridge|
US6887693B2|1998-12-24|2005-05-03|Cepheid|Device and method for lysing cells, spores, or microorganisms|
US6074616A|1998-01-05|2000-06-13|Biosite Diagnostics, Inc.|Media carrier for an assay device|
US6200814B1|1998-01-20|2001-03-13|Biacore Ab|Method and device for laminar flow on a sensing surface|
US6979424B2|1998-03-17|2005-12-27|Cepheid|Integrated sample analysis device|
US7188001B2|1998-03-23|2007-03-06|Cepheid|System and method for temperature control|
US7635588B2|2001-11-29|2009-12-22|Applied Biosystems, Llc|Apparatus and method for differentiating multiple fluorescence signals by excitation wavelength|
US6287765B1|1998-05-20|2001-09-11|Molecular Machines, Inc.|Methods for detecting and identifying single molecules|
EP0962773A1|1998-06-03|1999-12-08|Mark Howard Jones|Electrochemical based assay processes instrument and labels|
US6503231B1|1998-06-10|2003-01-07|Georgia Tech Research Corporation|Microneedle device for transport of molecules across tissue|
JP3389106B2|1998-06-11|2003-03-24|松下電器産業株式会社|Electrochemical analysis element|
US6743605B1|1998-06-24|2004-06-01|Enzo Life Sciences, Inc.|Linear amplification of specific nucleic acid sequences|
US7799521B2|1998-06-24|2010-09-21|Chen & Chen, Llc|Thermal cycling|
US6344333B2|1998-09-08|2002-02-05|Synectig Corporation|Reagent-free immunoassay monitoring electrode assembly|
US20020035459A1|1998-09-14|2002-03-21|George M. Grass|Pharmacokinetic-based drug design tool and method|
US6602469B1|1998-11-09|2003-08-05|Lifestream Technologies, Inc.|Health monitoring and diagnostic device and network-based health assessment and medical records maintenance system|
US6542717B1|1999-01-20|2003-04-01|International Business Machines Corporation|System and method for optimizing personal area network electrostatic communication|
CA2363833C|1999-02-23|2007-04-03|Oralscan Laboratories, Inc.|Minimally invasive apparatus and method for testing lesions of the oral cavity and similar epithelium|
US6215894B1|1999-02-26|2001-04-10|General Scanning, Incorporated|Automatic imaging and analysis of microarray biochips|
US8636648B2|1999-03-01|2014-01-28|West View Research, Llc|Endoscopic smart probe|
EP1106244A3|1999-03-03|2001-11-21|Symyx Technologies, Inc.|Chemical processing microsystems and controlling reaction conditions in same|
US6464687B1|1999-03-09|2002-10-15|Ball Semiconductor, Inc.|Implantable drug delivery system|
JP4085514B2|1999-04-30|2008-05-14|株式会社島津製作所|Electrophoresis chip|
US7209835B1|1999-04-30|2007-04-24|Centralized Laboratory Services, Inc.|Algorithmic testing in laboratory medicine|
US7001725B2|1999-04-30|2006-02-21|Aclara Biosciences, Inc.|Kits employing generalized target-binding e-tag probes|
AU4709400A|1999-05-11|2000-11-21|Ortho-Mcneil Pharmaceutical, Inc.|Pharmacokinetic and pharmacodynamic modeling of erythropoietin administration|
US6582964B1|1999-05-12|2003-06-24|Cme Telemetrix Inc.|Method and apparatus for rapid measurement of HbA1c|
US6544732B1|1999-05-20|2003-04-08|Illumina, Inc.|Encoding and decoding of array sensors utilizing nanocrystals|
DE60014676T2|1999-05-28|2005-11-17|Cepheid, Sunnyvale|DEVICE AND METHOD FOR THE ANALYSIS OF LIQUID SAMPLES|
AU5291600A|1999-06-01|2000-12-18|Caliper Technologies Corporation|Microscale assays and microfluidic devices for transporter, gradient induced, and binding activities|
US6256533B1|1999-06-09|2001-07-03|The Procter & Gamble Company|Apparatus and method for using an intracutaneous microneedle array|
EP1187677B1|1999-06-22|2003-11-26|Agilent Technologies, Inc. |Apparatus for the operation of a microfluidic device|
US7395216B2|1999-06-23|2008-07-01|Visicu, Inc.|Using predictive models to continuously update a treatment plan for a patient in a health care location|
US7195670B2|2000-06-27|2007-03-27|California Institute Of Technology|High throughput screening of crystallization of materials|
US6353475B1|1999-07-12|2002-03-05|Caliper Technologies Corp.|Light source power modulation for use with chemical and biochemical analysis|
US20040053290A1|2000-01-11|2004-03-18|Terbrueggen Robert Henry|Devices and methods for biochip multiplexing|
US6514769B2|1999-07-29|2003-02-04|Jin Po Lee|Multiple analyte assay device with sample integrity monitoring system|
JP4627395B2|1999-08-11|2011-02-09|旭化成株式会社|Analytical cartridge and liquid feeding control device|
EP1210064B1|1999-08-18|2005-03-09|Microchips, Inc.|Thermally-activated microchip chemical delivery devices|
JP2001065458A|1999-08-25|2001-03-16|Matsushita Electric Ind Co Ltd|Compressor|
ES2283153T3|1999-09-09|2007-10-16|Nokia Corporation|DETERMINATION OF DATA SPEED, BASED ON ESTIMATES OF SPECTRAL POWER DENSITY.|
US6835184B1|1999-09-24|2004-12-28|Becton, Dickinson And Company|Method and device for abrading skin|
US6368275B1|1999-10-07|2002-04-09|Acuson Corporation|Method and apparatus for diagnostic medical information gathering, hyperthermia treatment, or directed gene therapy|
JP3481578B2|1999-10-12|2003-12-22|松下電器産業株式会社|Electron-emitting device, electron source using the same, field-emission-type image display device, fluorescent lamp, and manufacturing method thereof|
CA2360194C|2000-10-25|2008-10-07|Micronix, Inc.|A solid state microcuvette using dry films|
US6471916B1|1999-11-09|2002-10-29|Packard Instrument Company|Apparatus and method for calibration of a microarray scanning system|
US6361958B1|1999-11-12|2002-03-26|Motorola, Inc.|Biochannel assay for hybridization with biomaterial|
US20050009101A1|2001-05-17|2005-01-13|Motorola, Inc.|Microfluidic devices comprising biochannels|
DE60041825D1|1999-11-17|2009-04-30|Boston Scient Ltd|MINIATURIZED DEVICES FOR DISPENSING MOLECULES IN A CARRIER FLUID|
JP3441058B2|1999-12-03|2003-08-25|理化学研究所|Microchip for capillary gel electrophoresis and method for producing the same|
JP2001165752A|1999-12-06|2001-06-22|Hitachi Ltd|Instrument and method for measuring serum quantity|
GB9930000D0|1999-12-21|2000-02-09|Phaeton Research Ltd|An ingestible device|
WO2001054813A2|2000-01-11|2001-08-02|Clinical Micro Sensors, Inc.|Devices and methods for biochip multiplexing|
WO2001053792A2|2000-01-19|2001-07-26|Given Imaging Ltd.|A system for detecting substances|
US6558320B1|2000-01-20|2003-05-06|Medtronic Minimed, Inc.|Handheld personal data assistant with a medical device and method of using the same|
AU3320001A|2000-01-31|2001-08-07|Univ Texas|Portable sensor array system|
JP4216434B2|2000-02-02|2009-01-28|大塚製薬株式会社|Test strip measuring device|
US7039453B2|2000-02-08|2006-05-02|Tarun Mullick|Miniature ingestible capsule|
KR100542386B1|2000-02-15|2006-01-10|주식회사 신한은행|System and method for managing a payment relation between the enterprises|
US7194395B2|2000-02-23|2007-03-20|The United States Of America As Represented By The Secretary Of The Army|System and method for hazardous incident decision support and training|
AU2001252890B2|2000-03-02|2005-06-02|Microchips, Inc.|Microfabricated devices for the storage and selective exposure of chemicals and devices|
US7041206B2|2000-03-09|2006-05-09|Clinical Analysis Corporation|Medical diagnostic system|
AU7681501A|2000-03-23|2001-10-08|Univ Johns Hopkins|Method and system for bio-surveillance detection and alerting|
US20100138160A1|2000-03-29|2010-06-03|Jacquez Geoffrey M|Model transition sensitivity analysis system and method|
US6927851B2|2000-03-31|2005-08-09|Neogen Corporation|Methods and apparatus to improve the sensitivity and reproducibility of bioluminescent analytical methods|
US6716620B2|2000-04-17|2004-04-06|Purdue Research Foundation|Biosensor and related method|
US6413213B1|2000-04-18|2002-07-02|Roche Diagnostics Corporation|Subscription based monitoring system and method|
US20030211618A1|2001-05-07|2003-11-13|Patel Gordhandhai Nathalal|Color changing steam sterilization indicator|
US7006858B2|2000-05-15|2006-02-28|Silver James H|Implantable, retrievable sensors and immunosensors|
IL143418A|2000-05-31|2004-09-27|Given Imaging Ltd|Measurement of electrical characteristics of tissue|
US6887202B2|2000-06-01|2005-05-03|Science Applications International Corporation|Systems and methods for monitoring health and delivering drugs transdermally|
EP1289417A4|2000-06-07|2005-06-15|Healthetech Inc|Breath ketone analyzer|
US6494830B1|2000-06-22|2002-12-17|Guidance Interactive Technologies, Inc.|Handheld controller for monitoring/using medical parameters|
US6699188B2|2000-06-22|2004-03-02|Guidance Interactive Technologies|Interactive reward devices and methods|
US6540675B2|2000-06-27|2003-04-01|Rosedale Medical, Inc.|Analyte monitor|
JP2002024401A|2000-07-06|2002-01-25|Takeda Chem Ind Ltd|Guidance/support system for treatment and prevention of disease|
US6603987B2|2000-07-11|2003-08-05|Bayer Corporation|Hollow microneedle patch|
JP2002031055A|2000-07-14|2002-01-31|Matsushita Electric Ind Co Ltd|Hermetic compressor|
US20020059030A1|2000-07-17|2002-05-16|Otworth Michael J.|Method and apparatus for the processing of remotely collected electronic information characterizing properties of biological entities|
US20030208113A1|2001-07-18|2003-11-06|Mault James R|Closed loop glycemic index system|
AU7716301A|2000-07-24|2002-02-05|Motorola Inc|Ingestible electronic capsule|
JP2002044007A|2000-07-26|2002-02-08|Ricoh Elemex Corp|Portable telephone|
US20040005582A1|2000-08-10|2004-01-08|Nanobiodynamics, Incorporated|Biospecific desorption microflow systems and methods for studying biospecific interactions and their modulators|
US20020026111A1|2000-08-28|2002-02-28|Neil Ackerman|Methods of monitoring glucose levels in a subject and uses thereof|
WO2002019246A2|2000-09-01|2002-03-07|The Government Of The United States Of America, As Represented By The Secretary Of The Navy|Early warning outbreak and recognition system|
US7349808B1|2000-09-06|2008-03-25|Egenomics, Inc.|System and method for tracking and controlling infections|
AT363922T|2000-09-08|2007-06-15|Insulet Corp|INFUSION DEVICE AND SYSTEM|
US7052831B2|2000-09-29|2006-05-30|Becton Dickinson And Company|Detection of multiple analytes from a single sample using a multi-well, multi-analyte flow-through diagnostic test device|
US20040030672A1|2001-08-01|2004-02-12|Garwin Jeffrey L|Dynamic health metric reporting method and system|
US6929636B1|2000-11-08|2005-08-16|Hewlett-Packard Development Company, L.P.|Internal drug dispenser capsule medical device|
JP2002161856A|2000-11-28|2002-06-07|Matsushita Electric Ind Co Ltd|Shaft and manufacturing method therefor|
JP2004537713A|2000-12-01|2004-12-16|セテクコーポレイション|High-throughput capillary electrophoresis system|
AU3322502A|2000-12-06|2002-06-18|Ecole Polytech|Bioanalytical reagent, method for production thereof, sensor platforms and detection methods based on use of said bioanalytical reagent|
GB0030929D0|2000-12-19|2001-01-31|Inverness Medical Ltd|Analyte measurement|
US6312929B1|2000-12-22|2001-11-06|Cepheid|Compositions and methods enabling a totally internally controlled amplification reaction|
AU2002243360A1|2000-12-26|2002-08-06|C. Frederick Battrell|Microfluidic cartridge with integrated electronics|
US6870797B2|2001-01-04|2005-03-22|Hewlett-Packard Development Company, L.P.|Media storage system using a transponder for transmitting data signal|
US7435384B2|2001-01-08|2008-10-14|Leonard Fish|Diagnostic instrument with movable electrode mounting member and methods for detecting analytes|
CA2366802A1|2001-01-17|2002-07-17|Gary E. Rehm|Method and apparatus for using infrared readings to detect misidentification of a diagnostic test strip in a reflectance spectrometer|
AU2002226655B2|2001-01-22|2006-09-07|Spectrum Dymanics Medical Limited|Ingestible device|
US6878755B2|2001-01-22|2005-04-12|Microgen Systems, Inc.|Automated microfabrication-based biodetector|
US20040109793A1|2002-02-07|2004-06-10|Mcneely Michael R|Three-dimensional microfluidics incorporating passive fluid control structures|
US6484104B2|2001-02-15|2002-11-19|Klaus Abraham-Fuchs|Network for evaluating data obtained in a biochip measurement device|
US7315784B2|2001-02-15|2008-01-01|Siemens Aktiengesellschaft|Network for evaluating data obtained in a biochip measurement device|
US6612985B2|2001-02-26|2003-09-02|University Of Rochester|Method and system for monitoring and treating a patient|
US6949377B2|2001-03-05|2005-09-27|Ho Winston Z|Chemiluminescence-based microfluidic biochip|
JP2002266762A|2001-03-07|2002-09-18|Matsushita Electric Ind Co Ltd|Refrigerating cycle device|
JP2002263185A|2001-03-12|2002-09-17|Sanyo Electric Co Ltd|Medicine administration system and method and medicine administration device|
JP3995424B2|2001-03-16|2007-10-24|株式会社パスコ|Infectious disease transmission analysis system and its transmission simulation system|
JP4323806B2|2001-03-19|2009-09-02|ユィロス・パテント・アクチボラグ|Characterization of reaction variables|
JP2002282217A|2001-03-27|2002-10-02|Sysmex Corp|Measuring device and result of measurement management system including the device|
US7010391B2|2001-03-28|2006-03-07|Handylab, Inc.|Methods and systems for control of microfluidic devices|
US20030009088A1|2001-04-04|2003-01-09|Uwe Korth|Monitoring system for patients|
KR100797458B1|2001-04-17|2008-01-24|엘지전자 주식회사|System for performing a medical diagnosis, mobile telephone and method for the same|
US20040142496A1|2001-04-23|2004-07-22|Nicholson Jeremy Kirk|Methods for analysis of spectral data and their applications: atherosclerosis/coronary heart disease|
US7563255B2|2001-05-03|2009-07-21|Massachusetts Eye And Ear Infirmary|Implantable drug delivery device and use thereof|
AU2002253388B2|2001-05-09|2006-09-28|Axis-Shield Asa|Assay system|
US6591124B2|2001-05-11|2003-07-08|The Procter & Gamble Company|Portable interstitial fluid monitoring system|
US20020173991A1|2001-05-18|2002-11-21|Boaz Avitall|Health care information management system and method|
DE60214375T2|2001-05-18|2007-08-30|Polymer Technology Systems, Inc., Indianapolis|DEVICE FOR EXAMINING BODY FLUIDS WITH SOLVENTLY FIXED, PORTABLE TEST DEVICE|
US6919046B2|2001-06-07|2005-07-19|Nanostream, Inc.|Microfluidic analytical devices and methods|
US6845327B2|2001-06-08|2005-01-18|Epocal Inc.|Point-of-care in-vitro blood analysis system|
JP2002371955A|2001-06-15|2002-12-26|Sanuki Kogyo Kk|Reciprocating drive unit and liquid transfer pump using the reciprocating drive unit|
US20030117491A1|2001-07-26|2003-06-26|Dov Avni|Apparatus and method for controlling illumination in an in-vivo imaging device|
JP3775263B2|2001-08-10|2006-05-17|ニプロ株式会社|Recording medium and blood glucose measurement system using the recording medium|
EP2309253A2|2001-09-05|2011-04-13|Life Technologies Corporation|Apparatus for reading signals generated from resonance light scattered particle labels|
US7807197B2|2002-09-28|2010-10-05|Mcneil-Ppc, Inc.|Composite dosage forms having an inlaid portion|
US6966880B2|2001-10-16|2005-11-22|Agilent Technologies, Inc.|Universal diagnostic platform|
JP2003222611A|2001-11-20|2003-08-08|Nec Corp|Separating apparatus and method therefor, and manufacturing method thereof|
WO2003043494A1|2001-11-23|2003-05-30|Medit As|A cluster system for remote monitoring and diagnostic support|
IL146776A|2001-11-27|2010-11-30|Yoram Alroy|Device for blood sampling under vacuum conditions|
JP2003166910A|2001-11-30|2003-06-13|Asahi Kasei Corp|Liquid-feeding mechanism and analyzer provided with the same|
US7266484B2|2001-12-04|2007-09-04|The Johns Hopkins University|Techniques for early detection of localized exposure to an agent active on a biological population|
JP2003167960A|2001-12-04|2003-06-13|Ikuo Kondo|Health control system|
US7457731B2|2001-12-14|2008-11-25|Siemens Medical Solutions Usa, Inc.|Early detection of disease outbreak using electronic patient data to reduce public health threat from bio-terrorism|
JP2005513439A|2001-12-17|2005-05-12|パウダージェクトリサーチリミテッド|Diagnostic sensing device|
US20050027182A1|2001-12-27|2005-02-03|Uzair Siddiqui|System for monitoring physiological characteristics|
JP2003207454A|2002-01-15|2003-07-25|Minolta Co Ltd|Transmission light-detecting apparatus|
US20030143551A1|2002-01-30|2003-07-31|Cattell Herbert F.|Reading multiple chemical arrays|
AU2003217306A1|2002-02-07|2003-09-02|Eastern Virginia Medical School Of The Medical College Of Hampton Roads|Diagnostic microarray and method of use thereof|
US7004928B2|2002-02-08|2006-02-28|Rosedale Medical, Inc.|Autonomous, ambulatory analyte monitor or drug delivery device|
US7343302B2|2002-02-11|2008-03-11|Puresense Environmental Inc.|System and method for emergency response|
US20030212379A1|2002-02-26|2003-11-13|Bylund Adam David|Systems and methods for remotely controlling medication infusion and analyte monitoring|
CA2419200C|2002-03-05|2015-06-30|Bayer Healthcare Llc|Fluid collection apparatus having an integrated lance and reaction area|
US7024370B2|2002-03-26|2006-04-04|P) Cis, Inc.|Methods and apparatus for early detection of health-related events in a population|
US6787108B2|2002-04-02|2004-09-07|Cmc Daymark Corporation|Plural intrinsic expiration initiation application indicators|
US7797033B2|2002-04-08|2010-09-14|Smart Pill Corporation|Method of using, and determining location of, an ingestible capsule|
US6832296B2|2002-04-09|2004-12-14|Ip-First, Llc|Microprocessor with repeat prefetch instruction|
JP2003315348A|2002-04-22|2003-11-06|Hitachi High-Technologies Corp|Specimen processing system and specimen inspection automating system using it|
US7112444B2|2002-04-24|2006-09-26|Wisconsin Alumni Research Foundation|Method of performing gradient-based assays in a microfluidic device|
WO2003090614A1|2002-04-25|2003-11-06|Matsushita Electric Industrial Co., Ltd.|Dosage determination supporting device, injector, and health management supporting system|
JP2003322653A|2002-05-07|2003-11-14|Toshiba Corp|Support and carrier for fixing probe|
US6883957B2|2002-05-08|2005-04-26|Cytonome, Inc.|On chip dilution system|
US20030143113A2|2002-05-09|2003-07-31|Lifescan, Inc.|Physiological sample collection devices and methods of using the same|
US7125510B2|2002-05-15|2006-10-24|Zhili Huang|Microstructure fabrication and microsystem integration|
JP3839349B2|2002-05-15|2006-11-01|株式会社堀場製作所|Chemiluminescent enzyme immunoassay device|
US7303921B2|2002-05-23|2007-12-04|Gian Paolo Littarru|Method to assay coenzyme Q10 in blood plasma or blood serum|
WO2003104770A2|2002-06-01|2003-12-18|Chempaq A/S|A disposable cartridge for characterizing particles suspended in a liquid|
JP4106977B2|2002-06-21|2008-06-25|株式会社日立製作所|Analysis chip and analyzer|
EP1376131A1|2002-06-27|2004-01-02|Inverness Medical Switzerland GmbH|Assay device for liquid sample|
US20040005247A1|2002-07-03|2004-01-08|Nanostream, Inc.|Microfluidic closed-end metering systems and methods|
US7470533B2|2002-12-20|2008-12-30|Acea Biosciences|Impedance based devices and methods for use in assays|
US7840421B2|2002-07-31|2010-11-23|Otto Carl Gerntholtz|Infectious disease surveillance system|
WO2004014227A1|2002-08-13|2004-02-19|Given Imaging Ltd.|System for in vivo sampling and analysis|
CN2559986Y|2002-08-23|2003-07-09|上海博昇微晶科技有限公司|Integrated microfluid and microchip of microarray probe|
JP2004101381A|2002-09-10|2004-04-02|Nittec Co Ltd|Double path cell for automatic analyzer, and analysis method using the double path cell|
US6874000B2|2002-10-08|2005-03-29|Food Security Systems, Llc|System and method for identifying a food event, tracking the food product, and assessing risks and costs associated with intervention|
US7177767B2|2002-10-18|2007-02-13|Abaxis, Inc.|Systems and methods for the detection of short and long samples|
US7390457B2|2002-10-31|2008-06-24|Agilent Technologies, Inc.|Integrated microfluidic array device|
US20040086872A1|2002-10-31|2004-05-06|Childers Winthrop D.|Microfluidic system for analysis of nucleic acids|
CN1173182C|2002-12-18|2004-10-27|陕西超英生物医学研究开发有限公司|Albumen chip for detecting autoimmunity antibody of diabetes, as well as preparation and detection method|
US20040122787A1|2002-12-18|2004-06-24|Avinash Gopal B.|Enhanced computer-assisted medical data processing system and method|
US20040122486A1|2002-12-18|2004-06-24|Stahmann Jeffrey E.|Advanced patient management for acquiring, trending and displaying health-related parameters|
US20040121305A1|2002-12-18|2004-06-24|Wiegand Roger Charles|Generation of efficacy, toxicity and disease signatures and methods of use thereof|
US20040122708A1|2002-12-18|2004-06-24|Avinash Gopal B.|Medical data analysis method and apparatus incorporating in vitro test data|
US20040236604A1|2002-12-20|2004-11-25|Mcnair Douglas S.|System and method for detecting spatiotemporal clusters|
US20060094108A1|2002-12-20|2006-05-04|Karl Yoder|Thermal cycler for microfluidic array assays|
EP2711415B1|2002-12-26|2022-02-16|Meso Scale Technologies, LLC.|Assay cartridges and methods of using the same|
US20050221281A1|2003-01-08|2005-10-06|Ho Winston Z|Self-contained microfluidic biochip and apparatus|
SE0300823D0|2003-03-23|2003-03-23|Gyros Ab|Preloaded Microscale Devices|
JP4464172B2|2003-03-31|2010-05-19|キヤノン株式会社|Biochemical reaction cartridge and method of using the same|
US7178386B1|2003-04-10|2007-02-20|Nanostream, Inc.|Parallel fluid processing systems and methods|
US7415299B2|2003-04-18|2008-08-19|The Regents Of The University Of California|Monitoring method and/or apparatus|
US7611480B2|2003-04-24|2009-11-03|Levy Mark M|Gastrointestinal bioreactor|
JP2004355616A|2003-05-06|2004-12-16|Hiroshi Sato|Information providing system and information processing system|
US7399276B1|2003-05-08|2008-07-15|Health Hero Network, Inc.|Remote health monitoring system|
US20060206010A1|2003-05-09|2006-09-14|Nec Corporation|Diagnostic support system and mobile terminal|
JP4260541B2|2003-05-12|2009-04-30|旭化成ファーマ株式会社|Test piece for measuring glycated protein|
US20040228766A1|2003-05-14|2004-11-18|Witty Thomas R.|Point of care diagnostic platform|
JP2007516446A|2003-12-23|2007-06-21|ファストラックインコーポレイテッド|Point-of-care diagnostic platform|
US6789510B1|2003-05-30|2004-09-14|Ru-Ping Lee|Animal restraining apparatus and animal experiment for using the same|
US7258673B2|2003-06-06|2007-08-21|Lifescan, Inc|Devices, systems and methods for extracting bodily fluid and monitoring an analyte therein|
US20060288253A1|2003-06-18|2006-12-21|Tatsurou Kawamura|Biological information utilization system, biological information utilization method, program and recording medium|
JP3918178B2|2003-06-23|2007-05-23|大阪瓦斯株式会社|Manufacturing method of high purity nanoscale carbon tube containing carbonaceous material|
US20050009191A1|2003-07-08|2005-01-13|Swenson Kirk D.|Point of care information management system|
JP2005030983A|2003-07-09|2005-02-03|Matsushita Electric Ind Co Ltd|Measuring instrument|
US8615282B2|2004-07-13|2013-12-24|Dexcom, Inc.|Analyte sensor|
US8346482B2|2003-08-22|2013-01-01|Fernandez Dennis S|Integrated biosensor and simulation system for diagnosis and therapy|
KR101165102B1|2003-09-03|2012-07-12|라이프 패치 인터내셔널, 인크.|Personal diagnostic devices and related methods|
JPWO2005024437A1|2003-09-05|2007-11-08|日本電気株式会社|Measuring system|
US7682833B2|2003-09-10|2010-03-23|Abbott Point Of Care Inc.|Immunoassay device with improved sample closure|
NZ580449A|2003-09-11|2011-06-30|Theranos Inc|Ingestible medical device with biocompatible polymer coating, device with microarray to interact with disease marker|
JP4933258B2|2003-09-22|2012-05-16|クイデルコーポレーション|Device for detecting multiple analytes in a sample|
US7524464B2|2003-09-26|2009-04-28|Ahn Chong H|Smart disposable plastic lab-on-a-chip for point-of-care testing|
JP2005104750A|2003-09-29|2005-04-21|Matsushita Electric Ind Co Ltd|Method for refining nanotube|
JP4441618B2|2003-10-06|2010-03-31|独立行政法人産業技術総合研究所|How to detect influenza virus|
JP4073023B2|2003-11-07|2008-04-09|財団法人新産業創造研究機構|Microchannel device and manufacturing method thereof|
EP1697872A2|2003-11-12|2006-09-06|Draeger Medical Systems, Inc.|A modular medical care system|
GB0329288D0|2003-12-18|2004-01-21|Inverness Medical Switzerland|Monitoring method and apparatus|
US7447333B1|2004-01-22|2008-11-04|Siemens Corporate Research, Inc.|Video and audio monitoring for syndromic surveillance for infectious diseases|
KR100814545B1|2004-01-27|2008-03-17|알티베라, 엘엘씨|Diagnostic radio frequency identification sensors and applications thereof|
US7105183B2|2004-02-03|2006-09-12|The Regents Of The University Of California|Chlorite in the treatment of neurodegenerative disease|
WO2005076957A2|2004-02-06|2005-08-25|Huttin Christine C|Cost sensitivity decision tool for predicting and/or guiding health care decisions|
WO2005079122A1|2004-02-11|2005-08-25|Cstar Technologies Inc.|Method and apparatus for cataloguing and poling movement in an environment for purposes of tracking and/or containment of infectious diseases|
US20060257941A1|2004-02-27|2006-11-16|Mcdevitt John T|Integration of fluids and reagents into self-contained cartridges containing particle and membrane sensor elements|
CN1667418B|2004-03-10|2010-10-06|马杰|Multifunctional portable unit for measurement, analysis and diagnosis|
JP2005291954A|2004-03-31|2005-10-20|Olympus Corp|Disposable reagent pack and analyzer using the reagent pack|
US7887750B2|2004-05-05|2011-02-15|Bayer Healthcare Llc|Analytical systems, devices, and cartridges therefor|
US7641856B2|2004-05-14|2010-01-05|Honeywell International Inc.|Portable sample analyzer with removable cartridge|
WO2007075920A2|2005-12-22|2007-07-05|Honeywell International Inc.|Hematological analyzer system with removable cartridge|
US8071051B2|2004-05-14|2011-12-06|Honeywell International Inc.|Portable sample analyzer cartridge|
EP1766400B1|2004-06-03|2009-04-15|Meso Scale Technologies, LLC|Methods for conducting whole blood assays|
EP1761645A4|2004-06-10|2008-12-31|Agency Science Tech & Res|Diagnostic primers and method for detecting avian influenza virus subtype h5 and h5n1|
EP1772736B1|2004-07-22|2013-07-03|Wako Pure Chemical Industries, Ltd.|Analysis assisting method, analyzer, remote computer, data analyzing method, program, and reagent container|
US20060036619A1|2004-08-09|2006-02-16|Oren Fuerst|Method for accessing and analyzing medically related information from multiple sources collected into one or more databases for deriving illness probability and/or for generating alerts for the detection of emergency events relating to disease management including HIV and SARS, and for syndromic surveillance of infectious disease and for predicting risk of adverse events to one or more drugs|
CN1311239C|2004-09-07|2007-04-18|李人|Immune chromatograph testing strip and production thereof|
AT391294T|2004-09-27|2008-04-15|Analytical Developments Ltd|liquid analyzer|
AU2005290314A1|2004-09-28|2006-04-06|Singulex, Inc.|System and method for spectroscopic analysis of single particles|
DE102004048864A1|2004-10-07|2006-04-13|Roche Diagnostics Gmbh|Analytical test element with wireless data transmission|
US20060218010A1|2004-10-18|2006-09-28|Bioveris Corporation|Systems and methods for obtaining, storing, processing and utilizing immunologic information of individuals and populations|
US8234129B2|2005-10-18|2012-07-31|Wellstat Vaccines, Llc|Systems and methods for obtaining, storing, processing and utilizing immunologic and other information of individuals and populations|
US20080020379A1|2004-11-05|2008-01-24|Agan Brian K|Diagnosis and prognosis of infectious diseases clinical phenotypes and other physiologic states using host gene expression biomarkers in blood|
US7396689B2|2005-02-04|2008-07-08|Decision Biomarkers Incorporated|Method of adjusting the working range of a multi-analyte assay|
CA2600934A1|2005-03-07|2006-09-14|Novx Systems Inc.|Automated analyzer|
EP3026874B1|2005-04-01|2019-11-06|Polymer Technology Systems, Inc.|Body fluid testing component for analyte detection|
US20070026426A1|2005-04-26|2007-02-01|Applera Corporation|System for genetic surveillance and analysis|
US8741230B2|2006-03-24|2014-06-03|Theranos, Inc.|Systems and methods of sample processing and fluid control in a fluidic system|
EP3763824A1|2005-05-09|2021-01-13|Labrador Diagnostics LLC|Point-of-care fluidic systems and uses thereof|
ES2752000T3|2005-05-09|2020-04-02|Biofire Diagnostics Llc|Autonomous biological analysis|
JP2006047305A|2005-07-08|2006-02-16|Nec Corp|Gas-specifying device, gas-specifying method, gas-coping support system and gas-coping support method|
JP4182227B2|2005-09-09|2008-11-19|国立大学法人岡山大学|Test method and test kit for latent virus infection|
WO2007035350A2|2005-09-15|2007-03-29|University Of Utah Research Foundation|Bioluminescence-based sensor with centrifugal separation and enhanced light collection|
GB0521851D0|2005-10-26|2005-12-07|Genial Genetic Solutions Ltd|Biological apparatus|
US7406453B2|2005-11-04|2008-07-29|Microsoft Corporation|Large-scale information collection and mining|
US20070179349A1|2006-01-19|2007-08-02|Hoyme Kenneth P|System and method for providing goal-oriented patient management based upon comparative population data analysis|
WO2007092713A2|2006-02-02|2007-08-16|Trustees Of The University Of Pennsylvania|Microfluidic system and method for analysis of gene expression in cell-containing samples and detection of disease|
US7705723B2|2006-03-15|2010-04-27|Dp Technologies, Inc.|Method and apparatus to provide outbreak notifications based on historical location data|
US10410308B2|2006-04-14|2019-09-10|Fuzzmed, Inc.|System, method, and device for personal medical care, intelligent analysis, and diagnosis|
US7727723B2|2006-04-18|2010-06-01|Advanced Liquid Logic, Inc.|Droplet-based pyrosequencing|
WO2007123908A2|2006-04-18|2007-11-01|Advanced Liquid Logic, Inc.|Droplet-based multiwell operations|
US8007999B2|2006-05-10|2011-08-30|Theranos, Inc.|Real-time detection of influenza virus|
US20100009352A1|2006-05-24|2010-01-14|Gough Albert H|Method for Modeling a Disease|
US7598092B2|2006-06-08|2009-10-06|Eric Toone|Methods, devices, systems and computer program products for stochastic, competitive, force-based analyte detection|
EP3745408A1|2006-07-13|2020-12-02|Abbott Point Of Care Inc|Medical data acquisition and patient management system and method|
US20090319295A1|2006-07-25|2009-12-24|Kass-Hout Taha A|Global disease surveillance platform, and corresponding system and method|
WO2008036381A2|2006-09-20|2008-03-27|Spadac Inc.|Method and system for global consolidated risk, threat and opportunity assessment|
US20080177571A1|2006-10-16|2008-07-24|Rooney James H|System and method for public health surveillance and response|
WO2008140557A2|2006-10-24|2008-11-20|Samuel Bogoch|A method of predicting influenza outbreaks|
US20080113391A1|2006-11-14|2008-05-15|Ian Gibbons|Detection and quantification of analytes in bodily fluids|
US8168121B2|2006-12-05|2012-05-01|Elkins Earthworks, Llc|Portable gas monitor|
WO2008098147A1|2007-02-07|2008-08-14|Core Microsolutions, Inc.|Remotely controlled real-time and virtual lab experimentation systems and methods|
WO2008103827A1|2007-02-22|2008-08-28|Welldoc Communications, Inc.|System and method for providing treatment recommendations based on models|
BRPI0809174A2|2007-04-02|2014-09-16|Kamran Khan|"SYSTEM AND METHOD FOR PREDICTING THE TRANSMISSION OF AN INFECTIOUS AGENT THROUGH AIR TRAVEL"|
EP2147115A4|2007-04-16|2010-05-05|Cardibioindex/cardibioscore and utility of salivary proteome in cardiovascular diagnostics|
US20080270200A1|2007-04-30|2008-10-30|International Business Machines Corporation|System and methods for pandemic planning|
KR100876764B1|2007-06-18|2009-01-07|한국정보통신대학교 산학협력단|Diagnosis system and method for cardiovascular disorders|
US8158430B1|2007-08-06|2012-04-17|Theranos, Inc.|Systems and methods of fluidic sample processing|
WO2009029599A1|2007-08-25|2009-03-05|Quantum Leap Research, Inc.|A scalable, computationally efficient and rapid simulation suited to decision support, analysis and planning|
CN103323610B|2007-10-02|2016-12-28|赛拉诺斯股份有限公司|Modular point-of-care devices and application thereof|
US20090112541A1|2007-10-26|2009-04-30|Joel Anderson|Virtual reality tools for development of infection control solutions|
US20110275079A1|2007-11-13|2011-11-10|Palma John F|Diagnostic biomarkers of diabetes|
US9489495B2|2008-02-25|2016-11-08|Georgetown University|System and method for detecting, collecting, analyzing, and communicating event-related information|
JP5511095B2|2008-03-20|2014-06-04|アバクシス,インコーポレイテッド|Multiwavelength analysis of sol particle specific binding assays|
CN102047255B|2008-03-26|2016-08-03|赛拉诺斯股份有限公司|Medical information system|
US8538778B2|2008-05-15|2013-09-17|Soar Biodynamics, Ltd.|Methods and systems for integrated health systems|
US8021850B2|2008-07-14|2011-09-20|Ribo Guo|Universal tandem solid-phases based immunoassay|
US8617873B2|2008-08-08|2013-12-31|Neal Solomon|Intelligent medical device system for personalized medicine applications|
KR101875858B1|2009-10-19|2018-07-06|테라노스, 인코포레이티드|Integrated health data capture and analysis system|
US9061283B2|2010-12-03|2015-06-23|Abbott Point Of Care Inc.|Sample metering device and assay device with integrated sample dilution|
US10828636B2|2016-10-25|2020-11-10|Fannin Partners Llc|Automated remotely instructed driving of an assay|CN103323610B|2007-10-02|2016-12-28|赛拉诺斯股份有限公司|Modular point-of-care devices and application thereof|
LT2476223T|2009-09-08|2021-07-12|Abbott Diabetes Care, Inc.|Methods and articles of manufacture for hosting a safety critical application on an uncontrolled data processing device|
KR101875858B1|2009-10-19|2018-07-06|테라노스, 인코포레이티드|Integrated health data capture and analysis system|
US9470584B2|2010-05-27|2016-10-18|Exergen Corporation|Method and apparatus for accurate detection of fever|
EP2590134A4|2010-06-30|2016-04-27|Nikon Corp|Infection spread prevention support system, infection spread prevention support server, examination terminal, mobile terminal and program|
US8762167B2|2010-07-27|2014-06-24|Segterra Inc.|Methods and systems for generation of personalized health plans|
BR112013018656B1|2011-01-21|2021-03-02|Labrador Diagnostics Llc|method for detecting the presence or concentration of an analyte in a sample of fluid contained in a container, and, method of measuring the concentration of analyte in a sample of fluid|
WO2012174230A1|2011-06-14|2012-12-20|Sickweather, Llc|Social networking aggregator to track illnesses|
US9619627B2|2011-09-25|2017-04-11|Theranos, Inc.|Systems and methods for collecting and transmitting assay results|
US8475739B2|2011-09-25|2013-07-02|Theranos, Inc.|Systems and methods for fluid handling|
US9632102B2|2011-09-25|2017-04-25|Theranos, Inc.|Systems and methods for multi-purpose analysis|
US9268915B2|2011-09-25|2016-02-23|Theranos, Inc.|Systems and methods for diagnosis or treatment|
US20140170735A1|2011-09-25|2014-06-19|Elizabeth A. Holmes|Systems and methods for multi-analysis|
US9250229B2|2011-09-25|2016-02-02|Theranos, Inc.|Systems and methods for multi-analysis|
US9664702B2|2011-09-25|2017-05-30|Theranos, Inc.|Fluid handling apparatus and configurations|
US10012664B2|2011-09-25|2018-07-03|Theranos Ip Company, Llc|Systems and methods for fluid and component handling|
EA201490948A1|2011-11-07|2014-11-28|Деб Груп Лтд.|METHOD FOR DETERMINING COMPLIANCE WITH HYGIENE OF HANDS|
US8645538B2|2011-11-20|2014-02-04|Yang Pan|System and method for monitoring outbreak of contagious diseases|
US9075909B2|2011-11-20|2015-07-07|Flurensics Inc.|System and method to enable detection of viral infection by users of electronic communication devices|
IN2014CN04748A|2011-12-16|2015-09-18|Koninkl Philips Nv|
WO2013138811A1|2012-03-16|2013-09-19|Life Of Purpose, Llc|Niche-specific treatment infrastructure continuum|
US20130262060A1|2012-03-29|2013-10-03|General Electric Company|Systems and methods for managing an infrastructure using a virtual modeling platform|
WO2014190231A1|2013-05-23|2014-11-27|Iphenotype Llc|Methods and systems for assisting persons, product providers and/or service providers|
US9946887B2|2012-06-04|2018-04-17|Nokia Technologies Oy|Method and apparatus for determining privacy policy based on data and associated values|
US20130332236A1|2012-06-08|2013-12-12|Ipinion, Inc.|Optimizing Market Research Based on Mobile Respondent Behavior|
US8984932B2|2012-07-18|2015-03-24|Theranos, Inc.|Rapid measurement of formed blood component sedimentation rate from small sample volumes|
US9500639B2|2012-07-18|2016-11-22|Theranos, Inc.|Low-volume coagulation assay|
CN110187089B|2012-07-18|2021-10-29|赛拉诺斯知识产权有限责任公司|Rapid measurement of sedimentation rate of formed blood components in small volume samples|
EP2882333A1|2012-08-09|2015-06-17|Koninklijke Philips N.V.|Device for home monitoring of haematological parameters of patients|
US9892155B2|2012-09-06|2018-02-13|Beyond Verbal Communication Ltd|System and method for selection of data according to measurement of physiological parameters|
US20140095417A1|2012-10-01|2014-04-03|Frederick S.M. Herz|Sdi |
CN104937414A|2012-10-24|2015-09-23|夏洛特-梅克伦堡医院|Biomarkers for the identification of liver damage|
US20160210881A9|2012-11-01|2016-07-21|Tyrone Ralph Smith|Scientific Instrument Trainer|
WO2014085794A1|2012-11-30|2014-06-05|Kinsa, Inc.|Mobile-enabled health system|
US20140164015A1|2012-12-06|2014-06-12|Oracle International Corporation|Clinical trial adverse event data change reporting system|
JPWO2014097598A1|2012-12-17|2017-01-12|日本電気株式会社|Information processing apparatus for performing risk analysis and risk analysis method|
US9599610B2|2012-12-19|2017-03-21|Dnae Group Holdings Limited|Target capture system|
CN103077321A|2013-01-25|2013-05-01|上海市浦东新区疾病预防控制中心|Hospital-based infectious disease symptom data acquisition system|
WO2014127204A2|2013-02-15|2014-08-21|Battelle Memorial Institue|Use of web-based symptom checker data to predict incidence of a disease or disorder|
US10332637B2|2013-02-15|2019-06-25|Battelle Memorial Institute|Use of web-based symptom checker data to predict incidence of a disease or disorder|
US9810704B2|2013-02-18|2017-11-07|Theranos, Inc.|Systems and methods for multi-analysis|
EP2787352A1|2013-04-05|2014-10-08|F. Hoffmann-La Roche AG|Analysis system for a biological sample|
CN104111322A|2013-04-18|2014-10-22|镇江太阳圣华医疗科技有限公司|Wireless-integrated and module-management coagulometer|
US9305257B2|2013-05-20|2016-04-05|International Business Machines Corporation|Adaptive cataclysms in genetic algorithms|
US10422806B1|2013-07-25|2019-09-24|Theranos Ip Company, Llc|Methods for improving assays of biological samples|
GB2516865A|2013-08-02|2015-02-11|Nokia Corp|Method, apparatus and computer program product for activity recognition|
CN105683751B|2013-09-06|2020-01-17|赛拉诺斯知识产权有限责任公司|Device, system, method and kit for receiving a swab|
BR112016004994A2|2013-09-06|2021-05-11|Theranos Ip Company, Llc|SYSTEMS AND METHODS TO DETECT INFECTIOUS DISEASES|
US20150100330A1|2013-10-08|2015-04-09|Assaf Shpits|Method and system of identifying infectious and hazardous sites, detecting disease outbreaks, and diagnosing a medical condition associated with an infectious disease|
EP3058331A4|2013-10-14|2018-01-17|Theranos, Inc.|Methods and systems for obtaining clinical samples|
CN103593554B|2013-11-01|2017-08-08|王浩锴|Remote emergency system|
MX2016005944A|2013-11-12|2016-08-17|Deb Ip Ltd|Reducing health care-associated infections based on hand hygiene.|
US20150186617A1|2013-12-27|2015-07-02|International Business Machines Corporation|System and method for probabilistic evaluation of contextualized reports and personalized recommendation in travel health personal assistants|
US9691046B1|2014-01-13|2017-06-27|Joel Adler|System and method for risk analyzed program planning |
RU2572227C2|2014-03-31|2015-12-27|Николай Александрович Контаров|Method for analysing and predicting development of epidemical situation caused by socially minded airborne infections|
USD745423S1|2014-05-12|2015-12-15|Cue Inc.|Automated analyzer test cartridge and sample collection device for analyte detection|
CN103954778A|2014-05-22|2014-07-30|江苏金标世纪生物科技有限公司|Myocardial infarction triple rapid detection kit and preparation method for same|
US20150350040A1|2014-06-02|2015-12-03|Hcl Technologies Ltd.|Enabling an early response to a disease based on data analytics|
EP3158526A4|2014-06-17|2018-03-07|University Of Virginia Patent Foundation|Continuous monitoring of event trajectories system and related method|
CA2952630A1|2014-06-28|2015-12-30|Relevance Health|System for assessing global wellness|
US20160012193A1|2014-07-09|2016-01-14|Gal Almogy|System, method and computer program product for disease monitoring and identification|
KR101609816B1|2014-09-11|2016-04-06|경희대학교 산학협력단|The data fusion apparatus based healthcare data integration model and method therefor|
US10541936B1|2015-04-06|2020-01-21|EMC IP Holding Company LLC|Method and system for distributed analysis|
US10528875B1|2015-04-06|2020-01-07|EMC IP Holding Company LLC|Methods and apparatus implementing data model for disease monitoring, characterization and investigation|
US10812341B1|2015-04-06|2020-10-20|EMC IP Holding Company LLC|Scalable recursive computation across distributed data processing nodes|
US10515097B2|2015-04-06|2019-12-24|EMC IP Holding Company LLC|Analytics platform for scalable distributed computations|
US10706970B1|2015-04-06|2020-07-07|EMC IP Holding Company LLC|Distributed data analytics|
US10505863B1|2015-04-06|2019-12-10|EMC IP Holding Company LLC|Multi-framework distributed computation|
US10348810B1|2015-04-06|2019-07-09|EMC IP Holding Company LLC|Scalable distributed computations utilizing multiple distinct clouds|
US10496926B2|2015-04-06|2019-12-03|EMC IP Holding Company LLC|Analytics platform for scalable distributed computations|
US10270707B1|2015-04-06|2019-04-23|EMC IP Holding Company LLC|Distributed catalog service for multi-cluster data processing platform|
US10791063B1|2015-04-06|2020-09-29|EMC IP Holding Company LLC|Scalable edge computing using devices with limited resources|
US10776404B2|2015-04-06|2020-09-15|EMC IP Holding Company LLC|Scalable distributed computations utilizing multiple distinct computational frameworks|
US10425350B1|2015-04-06|2019-09-24|EMC IP Holding Company LLC|Distributed catalog service for data processing platform|
US10656861B1|2015-12-29|2020-05-19|EMC IP Holding Company LLC|Scalable distributed in-memory computation|
US10860622B1|2015-04-06|2020-12-08|EMC IP Holding Company LLC|Scalable recursive computation for pattern identification across distributed data processing nodes|
US10509684B2|2015-04-06|2019-12-17|EMC IP Holding Company LLC|Blockchain integration for scalable distributed computations|
US10366111B1|2015-04-06|2019-07-30|EMC IP Holding Company LLC|Scalable distributed computations utilizing multiple distinct computational frameworks|
US10541938B1|2015-04-06|2020-01-21|EMC IP Holding Company LLC|Integration of distributed data processing platform with one or more distinct supporting platforms|
US10404787B1|2015-04-06|2019-09-03|EMC IP Holding Company LLC|Scalable distributed data streaming computations across multiple data processing clusters|
US10331380B1|2015-04-06|2019-06-25|EMC IP Holding Company LLC|Scalable distributed in-memory computation utilizing batch mode extensions|
US10511659B1|2015-04-06|2019-12-17|EMC IP Holding Company LLC|Global benchmarking and statistical analysis at scale|
US20160300146A1|2015-04-12|2016-10-13|Behzad Nejat|Method of Design for Identifying Fixed and Removable Medical Prosthetics Using a Dynamic Anatomic Database|
CN104730260B|2015-04-14|2017-04-12|苏州国科芯感医疗科技有限公司|Portable mobile blood clotting analyzing system and blood clotting analyzing method|
CA2984888A1|2015-05-05|2016-11-10|ShoCard, Inc.|Identity management service using a block chain|
JP6493006B2|2015-06-18|2019-04-03|富士通株式会社|Population estimation method, population estimation program, and population estimation device|
EP3689466A1|2015-07-17|2020-08-05|Cue Health Inc.|System for enhanced detection and quantification of analytes|
CN106342717A|2015-07-18|2017-01-25|吉林农业大学|Intelligent standardized rabbit breeding monitoring system based on Internet of Things|
US20170024539A1|2015-07-23|2017-01-26|PrioBio, LLC|Enhancing blood cell estimation|
US20180233233A1|2015-08-17|2018-08-16|Siemens Healthcare Gmbh|Method and System for Disease Progression Modeling and Therapy Optimizing for Individual Patients|
WO2017079938A1|2015-11-12|2017-05-18|Coyote Bioscience Co., Ltd.|Methods and systems for disease monitoring and assessment|
KR101822404B1|2015-11-30|2018-01-26|임욱빈|diagnostics system for cell using Deep Neural Network learning|
US10803993B2|2016-01-14|2020-10-13|Stuart Tin Fah Huang|Proximity tracing methods and systems|
US10614537B2|2016-01-26|2020-04-07|The Mitre Corporation|Systems and method for implementing biomedical innovation datametrics dashboard|
TWI691929B|2016-02-17|2020-04-21|原相科技股份有限公司|Interactive service platform and operating method thereof|
WO2017152150A1|2016-03-04|2017-09-08|ShoCard, Inc.|Method and system for authenticated login using static or dynamic codes|
US10007826B2|2016-03-07|2018-06-26|ShoCard, Inc.|Transferring data files using a series of visual codes|
US10509932B2|2016-03-07|2019-12-17|ShoCard, Inc.|Large data transfer using visual codes with feedback confirmation|
CN105956378A|2016-04-26|2016-09-21|成都聚恒康科技有限公司|Clinical decision supporting system of tumor diagnosis and treatment|
US20170316324A1|2016-04-27|2017-11-02|Virginia Polytechnic Institute And State University|Computerized Event-Forecasting System and User Interface|
US11195128B2|2016-08-02|2021-12-07|Baidu Usa Llc|Systems and methods for estimating healthcare resource demand|
WO2018026931A1|2016-08-02|2018-02-08|Eccrine Systems, Inc.|Biosensor disease and infection screening|
CN106066348A|2016-08-23|2016-11-02|武汉和生科技有限公司|Hospital blood sugar test instrument and noninvasive management system|
US10726957B2|2016-09-26|2020-07-28|Vitralogy Ip, Llc|Systems and methods for predicting and detecting hazardous conditions and facilitating regulatory compliance through automated communication platforms|
US20180107802A1|2016-10-17|2018-04-19|Sneez, Llc|Computer implemented method and system, and computer program product, for collecting and displaying health status information|
US10937548B2|2016-10-25|2021-03-02|Wisconsin Alumni Research Foundation|Computerized system for efficient augmentation of data sets|
CN106548103A|2016-11-03|2017-03-29|王力安防产品有限公司|Line production system based on RFID|
JP2022008719A|2016-11-23|2022-01-14|セルヴァス エーアイ インコーポレイテッド|Disease onset prediction method and equipment|
US10979305B1|2016-12-29|2021-04-13|Wells Fargo Bank, N.A.|Web interface usage tracker|
US10374968B1|2016-12-30|2019-08-06|EMC IP Holding Company LLC|Data-driven automation mechanism for analytics workload distribution|
WO2018140540A1|2017-01-25|2018-08-02|Cue Health Inc.|Systems and methods for enhanced detection and quantification of analytes|
RU2636857C1|2017-02-13|2017-11-28|Марина Николаевна Суворова|Modern method for teeth cleaning for persons with hearing disabilities|
CN110312935A|2017-02-15|2019-10-08|富士胶片株式会社|Immunologic test device and its operating method, information processing unit and its operating method and operation sequence and information processing system|
WO2018165354A1|2017-03-08|2018-09-13|Optofluidic Bioassay, Llc|Optofluidic diagnostics system|
US10404635B2|2017-03-21|2019-09-03|Bank Of America Corporation|Optimizing data replication across multiple data centers|
US10140835B2|2017-04-05|2018-11-27|Cisco Technology, Inc.|Monitoring of vectors for epidemic control|
US10978203B2|2017-06-20|2021-04-13|International Business Machines Corporation|Power-efficient health affliction classification|
CN108009390A|2017-09-02|2018-05-08|成都市双流区贸园医院有限公司|A kind of chronic disease medical order compliance management method and system|
CN107590805A|2017-09-15|2018-01-16|湖南友哲科技有限公司|Trichomonad detection method based on motion vector|
KR20190046459A|2017-10-26|2019-05-07|계명대학교 산학협력단|movable Intensive Care apparatus and system|
RU2658600C1|2017-11-20|2018-06-21|Федеральное государственное бюджетное образовательное учреждение высшего образования "Российский национальный исследовательский медицинский университет им. Н.И. Пирогова" Министерства здравоохранения Российской Федерации |Construction of the cartridge working area for carrying out a polymerase chain reaction|
WO2019110706A1|2017-12-08|2019-06-13|INSERM |Methods for predicting the risk of developing a sepsis or a systemic inflammatory response syndrome |
US11206133B2|2017-12-08|2021-12-21|Ping Identity Corporation|Methods and systems for recovering data using dynamic passwords|
CA3085195A1|2017-12-12|2019-06-20|VFD Consulting, Inc.|Reference interval generation|
US11010449B1|2017-12-12|2021-05-18|VFD Consulting, Inc.|Multi-dimensional data analysis and database generation|
WO2019152657A1|2018-02-03|2019-08-08|Simple Healthkit, Inc.|Reliable, comprehensive, and rapid sexual health assessment|
US11031134B2|2018-02-05|2021-06-08|International Business Machines Corporation|Monitoring individuals for water retention management|
WO2019156254A1|2018-02-09|2019-08-15|アクシオンリサーチ株式会社|System that estimates state of complex system to be inspected|
EP3764882A4|2018-03-14|2021-05-12|Siemens Healthcare Diagnostics Inc.|Predictive quality control apparatus and methods in diagnostic testing systems|
US10362769B1|2018-03-23|2019-07-30|International Business Machines Corporation|System and method for detection of disease breakouts|
CN108682457B|2018-04-17|2022-01-25|中国医学科学院阜外医院|Patient long-term prognosis quantitative prediction and intervention system and method|
CN108597618B|2018-04-20|2021-09-03|杭州恒生数字设备科技有限公司|Influenza prediction camera with automatic learning function|
CN108762780B|2018-05-09|2021-09-21|四川华创世纪科技有限公司|Method for realizing service software program separation operation|
CN108830034B|2018-05-25|2021-07-20|陈育聪|Medicine clinical research and development information processing system and method|
US11029300B2|2018-06-14|2021-06-08|International Business Machines Corporation|Detecting contamination sources in liquid distribution systems|
US20210233606A1|2018-06-20|2021-07-29|The Chinese University Of Hong Kong|Measurement and prediction of virus genetic mutation patterns|
KR102150602B1|2018-06-28|2020-09-01|케어마인드|Method, apparatus and computer program for postoperative diagnosis|
CN109065179B|2018-08-22|2021-10-22|昆明理工大学|Space-time dynamic simulation method for disease infection process|
CN109411015B|2018-09-28|2020-12-22|深圳裕策生物科技有限公司|Tumor mutation load detection device based on circulating tumor DNA and storage medium|
US10979227B2|2018-10-17|2021-04-13|Ping Identity Corporation|Blockchain ID connect|
US11082221B2|2018-10-17|2021-08-03|Ping Identity Corporation|Methods and systems for creating and recovering accounts using dynamic passwords|
JPWO2020116012A1|2018-12-04|2021-09-02|株式会社村田製作所|Virus detection system, virus detection method, and virus detection program|
CN109614430B|2018-12-05|2022-03-04|云南大学|Configurable data acquisition and data verification system and method based on WLAN|
CN109670541B|2018-12-07|2020-09-29|中国科学院软件研究所|Mosquito-borne infectious disease fever crowd range marking method based on space aggregation|
CN109859854A|2018-12-17|2019-06-07|中国科学院深圳先进技术研究院|Prediction Method of Communicable Disease, device, electronic equipment and computer-readable medium|
CN109740970B|2019-01-31|2020-09-22|杭州智乎物联科技有限公司|Management system for electric welding element temperature and welding personnel management applied to factory|
WO2020198511A1|2019-03-27|2020-10-01|Juno Diagnostics, Inc.|Digital health ecosystem|
WO2020205345A1|2019-03-29|2020-10-08|President And Fellows Of Harvard College|Influenza virus biomarkers and treatments|
US11250062B2|2019-04-04|2022-02-15|Kpn Innovations Llc|Artificial intelligence methods and systems for generation and implementation of alimentary instruction sets|
WO2020208912A1|2019-04-09|2020-10-15|株式会社Fronteo|Risk response analysis system, risk response analysis method, and risk response analysis program|
WO2020236763A1|2019-05-18|2020-11-26|Rpi Consulting Llc|Systems and methods for aspirating, dispensing, and transporting samples for analysis|
IT201900010719A1|2019-07-02|2021-01-02|Marco Visconti|STATION AND DATA ACQUISITION SYSTEM FOR THE ENROLLMENT OF SUBJECTS IN PROTOCOLS OF MEDICAL AND RESEARCH INTEREST|
US11131473B2|2019-07-12|2021-09-28|Johnson Controls Tyco IP Holdings LLP|HVAC system design and operational tool for building infection control|
CN110507294B|2019-08-07|2022-02-08|北京安龙脉德医学科技有限公司|First-aid system based on internet information transmission|
CN110705894A|2019-10-11|2020-01-17|秒针信息技术有限公司|Method and system for controlling virus risk of farm|
WO2021151191A1|2020-01-31|2021-08-05|Apt International Business Sciences Inc.|System and method for consistent user data acquisition and monitoring|
EP3911769A1|2020-02-18|2021-11-24|Life Technologies Corporation|Compositions, kits and methods for detection of viral sequences|
US11094413B1|2020-03-13|2021-08-17|Kairoi Healthcare Strategies, Inc.|Time-based resource allocation for long-term integrated health computer system|
US20210296007A1|2020-03-18|2021-09-23|Kinsa Inc.|Systems and methods for contagious illness surveillance and outbreak detection|
KR102329416B1|2020-03-18|2021-11-22|연세대학교 산학협력단|First aid guideline providing method and system based on Artificial Intelligence|
WO2021188942A1|2020-03-20|2021-09-23|WellDoc, Inc.|Systems and methods for analyzing, interpreting, and acting on continuous glucose monitoring data|
CN111430041A|2020-03-26|2020-07-17|北京懿医云科技有限公司|Infectious disease epidemic situation prediction method and device, storage medium and electronic equipment|
WO2021205435A1|2020-04-05|2021-10-14|Rapid Bio Pass Ltd.|Home testing with closed feedback loop and central epidemiological monitoring|
US11200987B2|2020-04-10|2021-12-14|Ix Innovation Llc|Virtual telemedicine mechanism|
WO2021211739A1|2020-04-15|2021-10-21|Healthpointe Solutions, Inc.|Evaluation of comprehensive clinical risk profiles of infectious disease in real-time|
CN111540477B|2020-04-20|2021-04-30|中国科学院地理科学与资源研究所|Respiratory infectious disease close contact person identification method based on mobile phone data|
US11197634B2|2020-04-28|2021-12-14|Wayne C. A. Wright|Geospatial bioimpedance biosurveillance tool|
US10777325B1|2020-04-29|2020-09-15|Zhaoyang Li|Self-governed individual social safe determination and confirmation method and system|
WO2021222978A1|2020-05-04|2021-11-11|Mark Andrew Radford|Health passport systems and methods of its use|
WO2021225780A1|2020-05-08|2021-11-11|Healthpointe Solutions, Inc.|Method for controlled and trust-aware contact tracing with active involvement of contact actors|
CN111601242A|2020-05-20|2020-08-28|深圳市本示科技有限公司|Epidemic prevention system for efficiently tracing close splicer by using Bluetooth technology|
WO2021235949A1|2020-05-22|2021-11-25|Juanico Drandreb Earl|System and methods for surveillance of epidemic networks by telemetry of infectious node estimate location|
WO2021242911A1|2020-05-26|2021-12-02|Pangolin Llc|Wastewater system to monitor pathogens and methods of use|
WO2021247682A1|2020-06-02|2021-12-09|Ross Peter M|Methods for mitigating or diminishing spread of pathogenic infections|
WO2021247905A1|2020-06-04|2021-12-09|Xcures, Inc.|Methods and systems for precision oncology using a multilevel bayesian model|
EP3929938A1|2020-06-25|2021-12-29|LeeLoo Medical GmbH|System, method and device for vehicle-based, mobile acquisition of health data|
US20210407692A1|2020-06-26|2021-12-30|Meso Scale Technologies, Llc.|Global bio-surveillance and response system|
US10991190B1|2020-07-20|2021-04-27|Abbott Laboratories|Digital pass verification systems and methods|
US20220028563A1|2020-07-22|2022-01-27|Pandemic Insights, Inc.|Geolocation pathogen-risk assessment with pandemic-bio-surveillance multi pathogen systems|
US11127506B1|2020-08-05|2021-09-21|Vignet Incorporated|Digital health tools to predict and prevent disease transmission|
US11056242B1|2020-08-05|2021-07-06|Vignet Incorporated|Predictive analysis and interventions to limit disease exposure|
US11107588B2|2020-08-11|2021-08-31|Gal EHRLICH|Methods and systems of prioritizing treatments, vaccination, testing and/or activities while protecting the privacy of individuals|
US11255762B1|2020-08-11|2022-02-22|Specialty DiagnosticLaboratories, Inc.|Method and system for classifying sample data for robotically extracted samples|
US11011278B1|2020-09-21|2021-05-18|Biolytical Laboratories Inc.|Methods and rapid test kits facilitating epidemiological surveillance|
US11232870B1|2020-12-09|2022-01-25|Neura Labs Ltd.|Communicable disease prediction and control based on behavioral indicators derived using machine learning|
RU2743687C1|2020-12-11|2021-02-24|федеральное государственное автономное образовательное учреждение высшего образования "Казанскийфедеральный университет" |Method of monitoring covid-19 morbidity with the use of wastewater analysis|
CN112562867A|2021-02-22|2021-03-26|天津迈德新医药科技有限公司|Device, storage medium and electronic device for predicting very early HIV infection risk|
US11170130B1|2021-04-08|2021-11-09|Aster Key, LLC|Apparatus, systems and methods for storing user profile data on a distributed database for anonymous verification|
CN113470770B|2021-06-30|2022-03-01|北京大学第三医院(北京大学第三临床医学院)|Platelet detection system based on error prediction model|
法律状态:
2018-05-29| B25A| Requested transfer of rights approved|Owner name: THERANOS IP COMPANY, LLC (US) |
2019-01-08| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]|
2019-08-20| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]|
2020-09-24| B25D| Requested change of name of applicant approved|Owner name: LABRADOR DIAGNOSTICS LLC (US) |
2020-10-27| B07A| Application suspended after technical examination (opinion) [chapter 7.1 patent gazette]|
2021-02-17| B09A| Decision: intention to grant [chapter 9.1 patent gazette]|
2021-03-30| B16A| Patent or certificate of addition of invention granted [chapter 16.1 patent gazette]|Free format text: PRAZO DE VALIDADE: 10 (DEZ) ANOS CONTADOS A PARTIR DE 30/03/2021, OBSERVADAS AS CONDICOES LEGAIS. |
优先权:
申请号 | 申请日 | 专利标题
US25301509P| true| 2009-10-19|2009-10-19|
US61/253,015|2009-10-19|
PCT/US2010/053088|WO2011049886A1|2009-10-19|2010-10-18|Integrated health data capture and analysis system|
[返回顶部]