WO2002070035A2 - Multi-purpose, automated blood and fluid processing systems and methods - Google Patents

Multi-purpose, automated blood and fluid processing systems and methods Download PDF

Info

Publication number
WO2002070035A2
WO2002070035A2 PCT/US2002/005244 US0205244W WO02070035A2 WO 2002070035 A2 WO2002070035 A2 WO 2002070035A2 US 0205244 W US0205244 W US 0205244W WO 02070035 A2 WO02070035 A2 WO 02070035A2
Authority
WO
WIPO (PCT)
Prior art keywords
blood
chamber
separation
patient
separated
Prior art date
Application number
PCT/US2002/005244
Other languages
French (fr)
Other versions
WO2002070035A3 (en
Inventor
Kyungyoon Min
Richard I. Brown
Rohit Vishnoi
Alan Armer
Tom Westberg
Original Assignee
Baxter International Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Baxter International Inc. filed Critical Baxter International Inc.
Priority to BR0204451-0A priority Critical patent/BR0204451A/en
Priority to EP02717481.2A priority patent/EP1365831B1/en
Priority to CA002407485A priority patent/CA2407485C/en
Priority to AU2002248479A priority patent/AU2002248479B2/en
Priority to JP2002569206A priority patent/JP2004519281A/en
Publication of WO2002070035A2 publication Critical patent/WO2002070035A2/en
Publication of WO2002070035A3 publication Critical patent/WO2002070035A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3687Chemical treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • A61M1/16Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis with membranes
    • A61M1/26Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis with membranes and internal elements which are moving
    • A61M1/262Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis with membranes and internal elements which are moving rotating
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • A61M1/30Single needle dialysis ; Reciprocating systems, alternately withdrawing blood from and returning it to the patient, e.g. single-lumen-needle dialysis or single needle systems for hemofiltration or pheresis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • A61M1/30Single needle dialysis ; Reciprocating systems, alternately withdrawing blood from and returning it to the patient, e.g. single-lumen-needle dialysis or single needle systems for hemofiltration or pheresis
    • A61M1/301Details
    • A61M1/302Details having a reservoir for withdrawn untreated blood
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • A61M1/30Single needle dialysis ; Reciprocating systems, alternately withdrawing blood from and returning it to the patient, e.g. single-lumen-needle dialysis or single needle systems for hemofiltration or pheresis
    • A61M1/301Details
    • A61M1/303Details having a reservoir for treated blood to be returned
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • A61M1/30Single needle dialysis ; Reciprocating systems, alternately withdrawing blood from and returning it to the patient, e.g. single-lumen-needle dialysis or single needle systems for hemofiltration or pheresis
    • A61M1/301Details
    • A61M1/305Control of inversion point between collection and re-infusion phase
    • A61M1/308Volume control, e.g. with open or flexible containers, by counting the number of pump revolutions, weighing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3621Extra-corporeal blood circuits
    • A61M1/3622Extra-corporeal blood circuits with a cassette forming partially or totally the blood circuit
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3621Extra-corporeal blood circuits
    • A61M1/3622Extra-corporeal blood circuits with a cassette forming partially or totally the blood circuit
    • A61M1/36225Extra-corporeal blood circuits with a cassette forming partially or totally the blood circuit with blood pumping means or components thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3621Extra-corporeal blood circuits
    • A61M1/3622Extra-corporeal blood circuits with a cassette forming partially or totally the blood circuit
    • A61M1/36226Constructional details of cassettes, e.g. specific details on material or shape
    • A61M1/362261Constructional details of cassettes, e.g. specific details on material or shape at least one cassette surface or portion thereof being flexible, e.g. the cassette having a rigid base portion with preformed channels and being covered with a foil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3621Extra-corporeal blood circuits
    • A61M1/3622Extra-corporeal blood circuits with a cassette forming partially or totally the blood circuit
    • A61M1/36226Constructional details of cassettes, e.g. specific details on material or shape
    • A61M1/362265Details of valves
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3672Means preventing coagulation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3672Means preventing coagulation
    • A61M1/3675Deactivation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3679Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits by absorption
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3692Washing or rinsing blood or blood constituents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3693Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits using separation based on different densities of components, e.g. centrifuging
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3693Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits using separation based on different densities of components, e.g. centrifuging
    • A61M1/3696Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits using separation based on different densities of components, e.g. centrifuging with means for adding or withdrawing liquid substances during the centrifugation, e.g. continuous centrifugation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/34Filtering material out of the blood by passing it through a membrane, i.e. hemofiltration or diafiltration
    • A61M1/342Adding solutions to the blood, e.g. substitution solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/34Filtering material out of the blood by passing it through a membrane, i.e. hemofiltration or diafiltration
    • A61M1/342Adding solutions to the blood, e.g. substitution solutions
    • A61M1/3441Substitution rate control as a function of the ultrafiltration rate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3621Extra-corporeal blood circuits
    • A61M1/3622Extra-corporeal blood circuits with a cassette forming partially or totally the blood circuit
    • A61M1/36222Details related to the interface between cassette and machine
    • A61M1/362227Details related to the interface between cassette and machine the interface providing means for actuating on functional elements of the cassette, e.g. plungers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3621Extra-corporeal blood circuits
    • A61M1/3622Extra-corporeal blood circuits with a cassette forming partially or totally the blood circuit
    • A61M1/36226Constructional details of cassettes, e.g. specific details on material or shape
    • A61M1/362266Means for adding solutions or substances to the blood
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2202/00Special media to be introduced, removed or treated
    • A61M2202/04Liquids
    • A61M2202/0413Blood
    • A61M2202/0427Platelets; Thrombocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/12General characteristics of the apparatus with interchangeable cassettes forming partially or totally the fluid circuit

Definitions

  • the present invention relates, in general, to a highly versatile, automated system for processing blood, blood components, and other fluids included in such processing. More particularly, the present invention relates to an automated system that can separate blood into two or more blood components ("apheresis") , and then perform a further procedure involving one or more of the separated components .
  • a separated component can be collected from a healthy donor, and later transfused to a patient in need of the component. Apheresis is also used in therapeutic applications to treat illness by removing diseased or otherwise undesirable components from a patient .
  • a basic apheresis procedure blood is withdrawn from a donor through a needle inserted into the vein of a donor.
  • the needle is attached to one end of a plastic tube which provides a flow path for the blood.
  • the other end of the tube terminates in a container for collecting the blood.
  • the collected blood is then separated in a separator, such as a centrifuge, into its components.
  • the desired blood component which, depending on the procedure, can be red blood cells, platelets, plasma, white blood cells or stem cells may be collected and stored for later transfusion to a patient in need of the blood component .
  • automated apheresis systems have come into widespread use. These automated systems utilize disposable, pre-sterilized fluid circuits (i.e., tubing sets) through which the blood flows.
  • the fluid circuits are mounted on re-usable hardware devices or modules that have pumps, valves, sensors and the like.
  • These automated systems further include an internal computer and associated software programs (controller) which control many of the processing functions.
  • blood flow through the fluid circuit, the operation of valves and pumps may be monitored and regulated by the system.
  • An automated system can be programmed to initiate, terminate or otherwise control certain functions based on patient or donor data (e.g., height, weight, sex, hematocrit) .
  • patient or donor data e.g., height, weight, sex, hematocrit
  • an automated system may monitor certain functions with the aid of sensors which can, for example, sense the amount of the collected or withdrawn component.
  • Optical sensors are used to measure the clarity or content of a fluid, or sense the presence or absence of certain components .
  • Automated apheresis systems are available from several different manufacturers. Examples of commercially available apheresis systems include the Autopheresis C ® Cell Separator and the Amicus ® Cell Separator, sold by Baxter Healthcare Corporation of Deerfield, Illinois.
  • the Autopheresis C ® utilizes a separator that includes a chamber and rotating membrane. Blood is introduced into the chamber and the membrane separates the blood into (at least) plasma and red blood cells, or other plasma-depleted blood.
  • the Amicus ® Cell Separator utilizes a centrifugal separation principle.
  • whole blood is introduced into a dual-chambered or single- chambered container mounted on a rotatable centrifuge.
  • Whole blood is introduced into the first chamber where red blood cells are separated from platelet-rich plasma (PRP) .
  • PRP platelet-rich plasma
  • the PRP flows into a second chamber where it is further separated into platelets and platelet-poor plasma.
  • the disposable fluid circuit of Amicus ® uses preformed cassettes with flow paths defined therein, which is mounted on the Amicus ® device. Flow through the flow path is assisted by peristaltic pumps.
  • a more detailed description of the Amicus ® Separator is provided in U.S. Patent No.
  • Another application where automated separating and processing blood may be desirable is in the salvaging of red blood cells during surgery on a patient.
  • blood from a wound area or from the body cavity i.e., extra-vascular or "shed” blood
  • the cleaned blood is returned to the donor. Examples of systems and apparatus used for cell salvage are described in U.S. Patent No. 5,976,388, which is incorporated herein by reference.
  • Another application where separating and processing blood may be desirable is in the removal of unwanted substances from blood or a separated blood component such as plasma. For example, the role of cholesterol and low density lipids (LDL) in cardiovascular disease has been well documented.
  • LDL low density lipids
  • the present invention is directed to an automated system for processing blood and a blood component.
  • the system includes a sterile, pre-assembled, disposable fluid circuit.
  • the fluid circuit includes means for withdrawing blood from a patient or donor and a separation chamber.
  • the circuit further includes means for treating the separated blood component.
  • the fluid circuit also includes a flow control cassette having pre-formed flow path segments that are separated by valve stations for controlling communication between the flow path segments.
  • the cassette also includes pump stations for pumping fluid through the flow path segments.
  • the automated system further includes a re-usable component or module adapted to cooperatively receive the fluid circuit.
  • the re-usable component includes means cooperatively associated with the separation chamber of the fluid circuit and for effecting separation of the blood component from blood.
  • the re-usable component also includes means for cooperating with the valve stations of the cassette to allow the flow of fluid through the preformed flow paths of the cassette.
  • the re-usable component also includes a programmable controller for selectively controlling fluid flow through the valve stations and for selectively establishing flow communication between the flow segments.
  • Figure 1 is a diagram showing the re-usable hardware component or module of the present invention and some of the available disposable fluid circuits for use therewith.
  • Figure 2 is a perspective view of a automated system that may be employed with the present invention, including the re-usable component and the disposable fluid circuit .
  • Figure 2A is an enlarged, perspective view of the separation chamber of the fluid circuit of Figure 2 which can be employed in the automated system of the present invention.
  • Figure 3 is a perspective view of an automated system that may be employed with the present invention with a disposable fluid circuit mounted on the re-usable device.
  • Figure 4 is a plane view of the front side of a cassette of the fluid circuit of Figure 2.
  • Figure 5 is a plane view of the back side of the cassette shown in Figure 4.
  • Figure 6 is a flow diagram showing the steps performed in the operation of the automated system of the present invention.
  • Figure 7 depicts the fluid circuit for a system and procedure embodying the present invention.
  • Figure 8 depicts the fluid circuit for an automated hemodilution system and procedure embodying the present invention.
  • Figure 9 depicts the fluid circuit for an automated plasma treatment system and procedure embodying the present invention.
  • Figure 10 depicts the fluid circuit for an automated cell treatment system and procedure embodying the present invention.
  • Figure 11 depicts the fluid circuit for an automated cell salvage system and procedure embodying the present invention cell salvage procedure.
  • Figure 12 depicts the fluid circuit for an alternative automated plasma treatment system and procedure embodying the present invention.
  • Figure 12A is a perspective view of an automated system that may be employed with the present invention with a disposable fluid circuit including a separation column mounted on the re-usable component.
  • Figure 13 is a perspective view of the re-usable component of an alternative automated system that may be employed with the present invention.
  • Figure 14 is a perspective view of a fluid circuit for use with the re-usable device of Figure 13.
  • Figure 15 is a perspective view of the fluid circuit shown in Figure 14 mounted on the re-usable component .
  • Figure 16 is an enlarged perspective view of a separation chamber of the fluid circuit of Figure 14. DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • Figure 1 diagrammatically shows a multi-purpose blood and fluid processing system 10 embodying the present invention.
  • automated system 10 includes a re-usable hardware component or module 12.
  • the re-usable hardware component 12 is particularly versatile and may be used with a variety of disposable fluid circuits.
  • hardware component 12 can be used with fluid circuits for red blood cell collection, plasma collection, platelet collection, white blood cell (leukocyte) collection, stem cell collection, hemodilution, cell salvage, lipid removal from plasma, conversion of red blood cells, cell washing, red blood cell exchange, leukoreduction, other therapeutic plasma treatments and, as will be seen, combinations of such procedures .
  • automated system 10 includes a re-usable module 12 and a disposable fluid circuit 50 for use in association with re-usable component 12.
  • Fluid circuit 50 includes an array of tubing and interconnected containers typically made of a sterilizable, plastic material. Fluid circuit 50 is intended for a single use (i.e., disposable, not re-usable). As shown in Figure 2, fluid circuit 50 includes a venipuncture needle 54 for insertion into the vein of the donor or patient. This needle 54 is attached to tubing, which provides a flow path for the blood withdrawn from the donor or patient . Needle 54 can also be used to return selected components to a donor or patient in a so-called "single-needle procedure. Alternatively, circuit 50 may use a "double-needle" configuration, known to those of skill in the art, where separate needles are used for withdrawal and return.
  • fluid circuit 50 includes several containers for temporary and/or longer-term storage of the separated components, and for holding fluids used during the procedure, such as an anticoagulant, saline, and any other treatment or replacement fluids required for the procedure.
  • Containers 56, 57, 58, 60, 62, 64, and 66 are also typically made of a sterilizable, plastic material.
  • Fluid circuit 50 further includes separation chamber 68.
  • Separation chamber 68 is intended for mounting on the separator of the re-usable device 12.
  • separation chamber 68 may be pre-formed by injection molding from a rigid, biocompatible plastic material, such as a non-plasticized medical grade acrilonitrite-butadiene-styrene (ABS) .
  • ABS acrilonitrite-butadiene-styrene
  • separation chamber 68 includes a base 388 with a center hub 120. Hub 120 is surrounded radially by inside and outside annular walls 122 and 134, which define a circumferential blood separation channel 126.
  • chamber 68 may include first and second sub-chambers.
  • the bottom of channel 126 is closed by a molded annular wall.
  • the top of channel 126 is closed by a separately molded flat lid (not shown) , which can be secured to the top of chamber 68 by welding or other securing means .
  • Chamber 68 also includes passageways 142, 144 and 146, which extend from hub 120 and communicate with channel 126.
  • blood is introduced into passageway 146 at the underside of base 388 via an attached multi-lumened tube or umbilicus 69 (shown as in Figure 2) .
  • Blood enters the channel 126 where it is separated into heavier and lighter components. The heavier components occupy the outer periphery of the channel, while the lighter component occupies the channel interior. The separated components are withdrawn through passageways 142 and 144.
  • Introduction and separation of blood using chamber 68 is described in more detail in U.S. Patent Application Serial No. 09/390,489, previously incorporated by reference.
  • Fluid circuit 50 further includes a cassette 70 which provides a network of flow path segments in fluid communication with and in association with numerous valving and pumping stations.
  • Cassette 70 provides a centralized, programmable, integrated platform for all the pumping and valving functions required for a given blood processing procedure.
  • a more detailed view of cassette 70 is provided in Figures 4 and 5.
  • Cassette 70 interacts with the pneumatically actuated pump and valve station 30 on re-usable module 12 described below.
  • cassette 70 has an array of interior cavities formed on both the front and back sides.
  • the interior cavities define the valve stations and flow paths.
  • Pump stations PP1 to PP4 are formed as wells that are open on the front side of the cassette 70.
  • the valves VI to V23 are likewise formed as wells that are open on the front side of cassette 70.
  • the liquid flow paths FI to F38 are formed as elongated channels that are open on the back side of cassette 70, except for liquid paths at F15, F23, and F24, which are formed as elongated channels that are open on the front side of the cassette 70.
  • the pre-molded ports PI to P13 extend out along two side edges of the cassette 70. As shown in Figure 3, the cassette 70 is vertically mounted for use in the pump and valve station 30 described below. In this orientation, ports P8 to P13 face downward, and the ports PI to P7 are vertically stacked one above the other and face inward.
  • Cassette 70 is preferably made of a rigid, medical grade plastic material. Flexible diaphragms overlay both of the front side and back side of cassette 70. The diaphragms are preferably made of flexible sheets of medical grade plastic. The diaphragms are sealed about their peripheries to the peripheral edges of the front and back sides of cassette 70. Interior regions of the diaphragms can also be sealed to interior regions of the cassette body.
  • a pneumatic pressure source which supplies negative and positive air pressure.
  • a pneumatic pressure source selectively distributes the different pressure and vacuum levels to the pump and valve stations. These levels of pressure and vacuum are systematically applied to the cassette 70 to route blood and processing fluids.
  • the details of the cassette, the pump and valve station 30, and the transport of blood and processing fluids through the cassette are set forth and described in U.S. Patent Application Serial No. 09/390,489, previously incorporated by reference.
  • re-usable component 12 includes (at least one) means for effecting separation of blood components or "separator" 20.
  • separator 20 is cooperatively associated with the chamber 68 of the fluid circuit.
  • separator 20 is adapted to receive the separation chamber of the fluid circuit (described above) and effects separation of whole blood or a blood fraction into two or more components .
  • separator 20 may be a rotatable centrifuge.
  • separator 20 is not limited to a separator that utilizes a centrifugal separation principle.
  • separator 20 may employ a different separation principle, such as a magnetic drive for receiving a spinning membrane as described, for example, in U.S. Patent No. 5,194,145.
  • separator 20 may also be a separation column with its own integral chamber or passageway. Separator 20 may also be a filter.
  • the spinning action of the centrifuge separates the blood components (within separation chamber 68) by density. For example, the spinning action of the centrifuge can separate whole blood into the more dense red cell component and a less dense plasma component.
  • Re-usable component 12 also houses the internal computer or controller 11.
  • the programmable controller includes pre-programmed instructions for carrying out several different blood and fluid processing procedures, allowing the operator to select from a menu, the particular procedure or procedures desired.
  • the controller also includes pre-programmed instructions which selectively activate pumping of fluid and the opening and closing of valves in the fluid circuit described above.
  • the controller may also include data storage capability for storing donor/patient information, processing or collection information and other data for later downloading or compilation.
  • re-usable device 12 includes a control panel such as flat screen display 24 for displaying the status of the procedure as well as providing a touch panel screen to allow for operator interface with the system.
  • Data output capability may also include standard parallel or serial ports or other network connection capability, as desired, for communication with other computers or networks .
  • Device 12 further includes pump and valve station 30.
  • pump and valve station 30 is designed to mate with corresponding structures of cassette 70.
  • Pump and valve station 30 contains four pump actuators PA1 to PA4 and twenty-three valve actuators VA1 to VA23. The pump and valve actuators are oriented to form a mirror image of the pump stations PP1 to PP4 and valve stations VI to V23 of cassette 70.
  • pump and valve station 30 (and, more specifically, pump and valve actuators PA(N) and VA(N)), apply positive and negative pressure upon cassette 70 and the corresponding pump and valve stations therein, to direct liquid flow through the flow path segments defined therein. Access to pump and valve station 30 is obtained through door 34.
  • Case 38 is suited for setup and operation upon a table top or other smooth and flat surface.
  • Case 38 includes a base 39 and hinged lid 40 which opens and closes.
  • Lid 40 includes latch 42 for releasably locking the lid.
  • Lid 40 further includes a handle 44, which the operator can grasp for easy transport of case 38 to a collection site, hospital, etc.
  • Case 38 is made by molding and, preferably, of a light-weight, durable plastic material.
  • lid 40 For supporting containers in a hanging position, lid 40 includes hooks (not shown) for hanging containers of saline, anticoagulant or other treatment or processing fluid. Similarly, a retractable hanger 45 is provided for supporting one or more collection containers in which whole blood and/or separated blood components are (at least temporarily) stored. Hanger 45 and hooks are preferably mounted on a scale 47 within lid 45 to allow automated measurement of the amount of whole blood or blood component collected.
  • Inclined container support surface 48 provides additional areas within the case for supporting containers associated with the disposable circuit 50. One or more areas of the support surface 48 may be heated, if desired, to warm the solution of the container prior to infusion to the donor or patient .
  • controller 11 includes a micro-processor and pre-programmed software. Although some interface and involvement by the operator is required, many of the functions of the automated system 10 are automatically controlled by controller 11.
  • the system automatically undergoes a system check procedure to confirm that all electrical and mechanical components of the device 12 are functioning properly and within preset parameters. If during the system check the controller detects a problem, the system may generate an audible alarm which prompts the operator to intervene. If the system successfully completes the system check, the system may prompt the operator to select the desired procedure.
  • the automated system allows the operator to select from a variety of procedures. For example, the operator may select a red cell collection
  • a plasma collection procedure a platelet collection procedure, a white blood cell collection procedure, a stem cell collection procedure.
  • the operator may select from a one or more other additional procedures.
  • the operator can select a first procedure to separate whole blood into two or more components, and also instruct the system to perform another additional treatment or other processing procedure including the separated componen (s) .
  • the operator may directly select one of the additional procedures, which already combines aspects of the above-mentioned apheresis procedures with additional "downstream" blood processing and/or treatment protocols.
  • these additional procedures include procedures for hemodilution, plasma treatment, such as lipid removal, the conversion of cells, cell salvage and other procedures including, but not limited to, therapeutic plasma treatments, removal of certain compounds from plasma using monoclonal antibodies, magnetic, para-magnetic, and other beads. Additional supplemental procedures involving the separated components of blood may also be performed.
  • the system prompts the operator to load the appropriate fluid circuit.
  • re-usable device 12 is adapted to receive any one of a variety of disposable fluid circuits.
  • Each of the enumerated procedures may require its own unique disposable fluid circuit or, more preferably, a disposable fluid circuit will be suitable for two or more procedures.
  • Most of the disposable fluid circuits will have many common elements such as a venipuncture needle, anticoagulant container, saline container, storage containers for red blood cells, plasma or whole blood, a separation chamber and the cassette 70.
  • each procedure may have its own unique disposable fluid circuit.
  • a universal fluid processing circuit can be used and any additional required containers of fluid (e.g., treating agents) , additional separators or other components can be easily attached at the time of use.
  • Additional fluid circuit components can be connected to the universal set in a sterile manner in ways that are well known to those of skill in the art.
  • Any additional tubes or flow paths can be attached to existing ports (e.g., P13) on the cassette and the system programmed to perform the additional procedures .
  • the system will verify that the correct disposable has been loaded and/or that it has been loaded properly. Once proper loading of the disposable processing circuit has been confirmed, the system will automatically initiate a priming sequence based on the selected protocol .
  • the priming sequence will include priming the fluid circuit with anticoagulant and/or saline.
  • the system may also prime the disposable fluid circuit with such fluid or the patient's or donor's blood.
  • the system may allow the operator to enter desired patient or donor data, such as height, weight, gender, hematocrit, or any other donor or patient characteristic that the controller may utilize during the course of the procedure . Entry of donor data may occur before or after prime. For example, the system may use the above-described donor data to determine flow rates, and/or duration of a particular step. After prime and entry of any required donor data, the system prompts the operator to begin the procedure .
  • desired patient or donor data such as height, weight, gender, hematocrit, or any other donor or patient characteristic that the controller may utilize during the course of the procedure . Entry of donor data may occur before or after prime. For example, the system may use the above-described donor data to determine flow rates, and/or duration of a particular step. After prime and entry of any required donor data, the system prompts the operator to begin the procedure .
  • Figure 7 shows an automated system embodying the present invention including a typical fluid circuit. As shown in Figure 7, whole blood is withdrawn from donor 100 and introduced into cassette 70 via line (tubing) 74. Anticoagulant from container 56 is likewise drawn into cassette 70. Anticoagulant enters through one of the ports
  • Controller opens the selected valve (s) to allow anticoagulant flow through the fluid segment, and establishes flow communication between the anticoagulant line 75 and line 74 to combine anticoagulant with the whole blood being withdrawn from the patient.
  • Anticoagulated whole blood is introduced into container 58, which serves as an interim whole blood source.
  • the hardware component 12 can include weight scales.
  • container 58 is suspended from a weight scale so that when the required amount of whole blood is collected a sensor attached to the weight scale prompts the controller and the draw cycle is terminated. The controller then initiates pumping (by controlling the pump and valve station) of whole blood from container 58 into the separator.
  • the separator separates whole blood into two or more components.
  • the separator may be cooperatively associated with separator chamber 68 of the fluid circuit, either physically or as described in, for example, U.S. Patent No. 5,194,195, incorporated herein by reference.
  • separation of whole blood results in a plasma component and a red blood cell component.
  • the separated plasma and red cell components may be withdrawn from separation chamber 68 and collected in separate containers 60 and 62 for temporary storage.
  • further processing of one or both of the separated components may be initiated by the system.
  • fluid circuit 50 may include a treatment fluid container 64.
  • container 64 may include a replacement fluid.
  • further processing or treatment of the separated component may take place in separation chamber 68, one of the containers 60 or 62, or, if required, a second and separate separator (shown in dashed lines in Figure 7 and labeled "Separator 2").
  • the second separator may utilize the same separation principle as Separator 1.
  • both Separators 1 and 2 may be centrifuges.
  • one of the separators may be a centrifuge while the other separator may be a drive mechanism for cooperation with a rotating member and separation membrane of the type described in U.S. Patent No. 5,194,145.
  • one of the separators may be a centrifuge or a drive for a rotating membrane and the other separator can be a filter medium or a separation column.
  • the blood component separated in Separator 1 can be directly introduced into Separator 2 for further treatment and/or processing.
  • blood component can be introduced into cassette 70 from where it can be directed and/or pumped into Separator 2.
  • the separated (and/or treated) component can be directly reinfused back to the donor, as shown by dashed line 87, or through cassette 70 and return line 84.
  • Figure 7 shows a single vein access point (i.e., single needle) for withdrawal of blood and return of blood component
  • a single vein access point i.e., single needle
  • the fluid circuit shown in Figure 7 may also utilize a so-called “double-needle" configuration described above.
  • Figure 8 shows an automated system and procedure for the automated pre- surgical withdrawal of blood, separation into plasma and red blood cells, followed by the return of plasma and infusion of one or more replacement fluids (i.e., "hemodilution" ) .
  • a system of the type shown in Figure 8 is particularly useful in the collection of autologous blood from a patient just prior to a surgical procedure.
  • the plasma component is returned and is supplemented with a volume replacement fluid (such as saline) and a blood substitute which can provide the same biological function (i.e., oxygen transport) as the collected red blood cells.
  • a volume replacement fluid such as saline
  • a blood substitute which can provide the same biological function (i.e., oxygen transport) as the collected red blood cells.
  • whole blood is withdrawn from a patient just prior to the surgical procedure.
  • the whole blood is withdrawn through line 74 and introduced into cassette 70 in the manner generally described above.
  • Whole blood is combined with anticoagulant and the anticoagulated whole blood may be introduced into container 58 or immediately introduced into the separator. Once inside the separator and, more particularly, the separation chamber 68 associated with the separator, whole blood is separated into a red blood cell component and a plasma component.
  • the red cell component is removed from the separator (by pumping of the pump stations in the cassette 70) and collected in container 60 where it is stored until needed (if needed) during or after surgery. If long-term storage of red cells is required, the collected red cells may be combined with a red blood cell preservative solution such as Adsol ® or Erythro-Sol, available from Baxter Healthcare Corporation of Deerfield Illinois. Administration of a preservative (stored, for example, in container 57 ( Figure 2) can also be controlled by controller 11.
  • a preservative stored, for example, in container 57 ( Figure 2) can also be controlled by controller 11.
  • the separated plasma can be introduced into container 62 from where it can be metered back to the patient during surgery or, in the alternative, immediately returned to the patient. In another alternative, plasma (with platelets) may be returned to the patient after surgery, at or around the time that the red blood cells are returned.
  • a volume replacement fluid such as saline may also be administered to the patient.
  • red blood cells include hemoglobin, an oxygen carrying compound in blood, a blood substitute or other synthetic oxygen carrying compound that can perform the same oxygen transport function as the red blood cells may also be administered to the patient.
  • Such blood substitutes and/or oxygen carrying compounds are known and are available from Alliance Pharmaceutical Corporation of San Diego, California, and are described in U.S. Patent No. 5,865,784. Other blood substitutes known to those of skill may also be used.
  • the blood substitute may be combined with the saline or administered separately either before or after administration of the saline.
  • other volume replacement fluids in lieu of saline (which is a crystalline solution) may also be used as the volume replacement fluid.
  • the system 10 can automatically determine the amount and flow rate of the fluids, i.e., saline and a blood substitute required.
  • the controller can be preprogrammed to administer the selected amount of saline or other fluid and a replacement fluid having a known biological function, such as a blood substitute, based on the amount of the red blood cells collected as measured by the weight scale 47 in hardware component 12.
  • the system can determine the amount (and flow rate) of the replacement fluid to be administered based on the amount of whole blood withdrawn.
  • the system can determine the amount of replacement fluid and blood substitute to be administered based on donor data entered at the beginning of the procedure.
  • the automated system of the present invention provides benefits that manual hemodilution cannot achieve.
  • the hemoglobin concentration of the blood is reduced from approximately 12 mg/dl to 9 mg/dl by withdrawing blood and administrating support fluid (3 times the saline or albumin) .
  • Three times the volume of saline is necessary due to saline's limited ability to stay within the vascular space.
  • Albumin being a molecule of larger size, can stay within the vascular space and will not be as quickly excreted as saline.
  • the volume of fluid removed would be limited to the red blood cell volume which would be approximately 400 ml (based on an average, hematocrit of 40%) .
  • the total volume of red blood cells removed is 400 ml, with the remaining 600 ml consisting of plasma.
  • To remove an equal amount of red blood cells using the automated system and procedure would require the removal of only 400 ml of concentrated red blood cells with all of the plasma processed by the system being returned to the patient. This reduces the volume removed by 60%. To replace this, only 400 ml of albumin or 1,200 ml of saline would be necessary. This is substantially less than the typical manual hemodilution procedure .
  • any potential fluid complication caused by saline can be reduced.
  • Saline can cause fluid overload and tissue edema in patients with renal insufficiency.
  • a large volume saline infusion and associated increase in tissue fluid can necessitate the need for diuretic administration to assist in fluid removal after the surgical procedure.
  • Another advantage of the automated system of the present invention is that the system can be programmed by the anesthesiologist and the procedure accomplished automatically.
  • the system can add the appropriate amount of anticoagulant to the blood to prevent clotting in the blood storage container and red cell additive solutions can be used as necessary.
  • Citrate anticoagulation can cause some citrate reactions in patients during apheresis procedures. Citrate reactions are usually controlled by infusion of calcium containing solutions. Using the automated system of the present invention, when the collected blood products are transfused back to the patient, the minimum amount of anticoagulant will be present in the collected blood which, upon transfusion, should cause fewer complications due to citrate transfusion compared to the manual method.
  • the time until blood is to be administered may be prolonged. More importantly, this automated system and procedure can reduce or eliminate the need for non-autologous blood. By eliminating or reducing the need for non-autologous blood, the patient can have limited exposure to non-autologous homologous blood. This can reduce the possibility of post transfusion immunosuppression or inflammatory response due to transfusion of stored blood (cytokine generation during storage) .
  • the automated system of the present invention will allow for the plasma (and platelets) to be returned to the patient. By returning the plasma (and platelets), the patient can more easily maintain normal hemostasis. (In the standard manual hemodilution procedure, severe dilution can cause hemostasis problems which may require infusion of cryoprecipitated clotting proteins (cryoprecipitate) or fresh frozen plasma (FFP) . This also occurs during the manual procedure because whole blood is removed which removes platelets and plasma as well as RBC's.) [0091] Figure 9 shows another application of the automated system of the present invention. In particular, Figure 9 shows a procedure that results in removal of undesired compounds from blood plasma.
  • the fluid circuit and flow system shown in Figure 9 can be used for removal of lipids from the plasma of the patient.
  • whole blood is withdrawn from a patient 100 via venipuncture and allowed to flow through line 74 into cassette 70, and combined with anticoagulant as previously described.
  • the anticoagulated whole blood may be collected in container 58 until a selected weight is attained.
  • the system introduces whole blood into the separator, which can include or is otherwise cooperatively associated with separation chamber 68, where it is separated into red blood cells and plasma.
  • the separated red blood cells may then be returned to the patient immediately, or temporarily stored in container 60 for later return.
  • the separated plasma may then be further treated to remove lipids (or any other undesirable compounds) .
  • plasma may be combined with a solvent contained in container 64.
  • the solvent is capable of extracting lipids from the plasma.
  • solvents are described in, for example, U.S. Patent Nos. 4,895,558, 5,744,558 and 5,911,698, which are incorporated herein by reference.
  • solvents are DIPE (di- isopropylether) .
  • DIPE di- isopropylether
  • Plasma and the solvent may be combined in, for example, container 62 or inside separator 68. If combined outside of the separator, the plasma and solvent may then be reintroduced into the separator to further separate plasma from the lipid containing solvent.
  • the separator is a centrifugal separator of the type shown in Figures 2-3 and/or Figures 13-16. Centrifugal action results in the separation into a two- phase solution, an upper organic phase that includes the solvent and extracted lipid, and a lower lipid-depleted plasma phase. Under control of the controller, the lipid containing solvent may then be pumped to a separate waste container. The lipid-depleted plasma may be returned to the patient.
  • the plasma may be treated with a further washing solution from container 69. Treatment in the washing solution can take place in the separation chamber before return of the plasma to the patient.
  • system 10 may include a second separator (Separator 2) for the cleansing and/or washing step.
  • Separator 2 may employ the same separation principle (e.g., centrifugation) as Separator 1, or more preferably, may employ a different separation principle.
  • Separator 2 may be a column packed with coated beads that have an affinity for the solvent.
  • plasma may be removed from Separator 1 and introduced into column Separator 2 (either directly or via cassette 70) to remove any residual solvent. Plasma that has been passed through Separator 2 may then be suitable for return to the patient.
  • Separator 2 may be filter medium.
  • the system can include an optical detector 83, which is capable of detecting lipids in the plasma being returned. Such detectors are described in U.S. Patent No. 5,958,250, incorporated herein by reference.
  • removal of undesired compounds from plasma may be achieved without resort to a solvent-based system.
  • plasma that has been separated in the separator may be treated or contacted with another material for removing lipids from plasma.
  • a blood component that has been separated from whole blood can be further treated with particles or beads that have a specific affinity for the compound to be removed.
  • container 64 may include the beads or particles.
  • the beads may be lightweight, simple, hollow (or solid) sphere-like structures.
  • the beads are coated with an affinity material, such as monoclonal antibodies.
  • the beads may have a specific affinity for lipids, sickled cells, immunoglobulins, Factor VIII or other proteins.
  • the beads preferably, have a density less than the density of plasma.
  • the beads may be of the type described in U.S. Patent Nos. 5,916,743 and 5,641,622, which are incorporated herein by reference.
  • whole blood is withdrawn from the donor (or patient) through line 74 and combined with anticoagulant as previously described.
  • Anticoagulated whole blood is collected and temporarily stored in container 58.
  • whole blood is introduced in the separator where it is separated into a cellular component and a plasma component.
  • the cellular component can be removed from the separator and collected in, for example, container 60.
  • the plasma component can be combined with beads in container 60 or, to ensure greater contact between the beads and the compound to be removed, in the separator.
  • the controller will cause beads to be pumped into either container 62 or the separator.
  • the bound particle can then be collected in container 62 and the plasma returned to the donor.
  • plasma may be passed through a filter or other type of medium that has attached to its surface monoclonal antibodies that have a specific affinity for lipids.
  • the filter medium may be a flat sheet or a packed column of the type described above.
  • the separation medium e.g., separator 80
  • Another application for the automated system of the present invention is in the treatment of blood cells, such as red cells, white cells or platelets.
  • the automated system can be used to treat red blood cells with an enzyme to convert, for example, Type-A, Type-B, or Type-AB red blood cells to Type-0 red blood cells.
  • whole blood is withdrawn from a patient 100, anticoagulated in the manner described above, and separated in separator 68 to provide a red blood cell component and a plasma component.
  • the plasma component can be collected in container 60 or can be returned to the donor immediately.
  • the red blood cell component can be temporarily collected in container 60.
  • the red blood cell component can be combined with a solution (stored in container 64) that includes a particular enzyme suitable for the red blood cell conversion. Examples of such enzymes are included in U.S. Patent Nos. 6,175,420 and 5,671,135, which are incorporated by reference herein.
  • the treated red blood component may then be collected and stored in container 62.
  • Type-0 blood cells are to be stored long term (e.g., up to 42 days)
  • a preservative solution of the type described above can be added to the red blood cells.
  • red blood cells, platelets or even plasma may be treated to eradicate or inactivate pathogens present in these components.
  • Another application of the automated system of the present invention can be the salvage of blood during a surgical procedure.
  • whole blood can be collected from the body cavity of a patient 100 undergoing surgery.
  • fluid circuit will include a suction device 120 instead of a venipuncture needle.
  • Suction device 120 maybe of the type shown in, for example, U.S. Patent No. 5,976,388, which is incorporated herein by reference. Blood that is removed by suction device 120 is introduced into the separator where it is separated into a red blood cell component and supernatant. The red cell product may then the returned to patient .
  • FIG. 12 an alternative, automated system for treating separated plasma is shown.
  • the system includes a first separator and a second separator.
  • the second separator is a separation column 80 that can be used to remove the above-described compounds from plasma.
  • Columns that can be used for such separation are generally disclosed in U.S. Patent Nos. 5,733,254 and 5,782,792, which are incorporated herein by reference.
  • separation of plasma from whole blood proceeds as generally described above, i.e. , in the separator.
  • the separated plasma may be introduced into column 80.
  • plasma can be directly introduced into column 80 via line 81, or can be conveyed by the pumps and valves of cassette 70 (under the direction of the controller) to column 80.
  • plasma that has passed through column 80 can be returned via cassette 70, can be directly introduced into line 74 for direct return to the donor, or can be introduced into container 62 from where it is pumped (through cassette 70) back to the donor. Red cells in container 60 may be returned to the donor during processing of plasma.
  • Column 80 may be provided as part of the fluid circuit 50.
  • re-usable component 12 can be equipped with clips 13 and 15 for holding column 80, as generally shown in Figure 12A.
  • Figures 13-16 show an alternative embodiment of another re-usable hardware device and fluid circuit that can be used in the automated system and procedure of the present invention.
  • the embodiment shown in Figures 13-16 include a centrifuge assembly 200 and a fluid processing circuit 50 for use in association with the centrifuge assembly.
  • the centrifuge assembly includes a re-usable hardware device capable of long-term use.
  • the disposable fluid circuits like the fluid circuits described above, are intended to be a single-use, disposable item.
  • the fluid circuits shown in Figures 13-16 include a processing chamber, shown in Figure 16, that can be loaded onto a separator of the re-usable device, to centrifugally separate blood components.
  • the separator may separate whole blood into a red blood cell component, a plasma component, a white blood cell component, stem cells or a platelet component.
  • the disposable fluid circuit also includes an array of flexible tubing to convey liquid to and from the processing chamber, described in more detail below.
  • Fluid circuit 50 includes one or more cassettes 222A, B and C, generally of the type described above.
  • the cassettes shown in Figures 14-15 include inter-connectable flow segments and valving stations .
  • the cassettes of this embodiment do not include internal pumping stations. Instead, the cassettes of this embodiment include external tubing loops 223 which engage peristaltic pump rotors 250, which effect movement of fluid through the tubing and the fluid circuit.
  • the details of this embodiment of the automated system are described in U.S. Patent No. 5,868,696, which is incorporated herein by reference.
  • fluid circuit 50 includes a "two-staged" separation chamber 68.
  • the first sub-chamber 226 can be used to perform a first separation step and the second sub-chamber 224 can be used to perform a second separation step.
  • a blood component such as plasma or red blood cell
  • plasma can be separated from red cells in the first "sub-chamber" 226 and the treatment carried out in the second "sub-chamber" 224.
  • a treating agent can be directly introduced into the second subchamber or can be combined with the component elsewhere, such as in one of the containers .
  • the second subchamber can also be used to remove undesirable solvents, compounds, treating agents from the separated component.
  • the blood and fluid processing procedures described above are applicable to the automated system described and shown in Figures 13- 16.
  • the chamber 68 may have only a single chamber .
  • circuit 50 shown in Figure 14 is adapted for single needle platelet collection.
  • Circuit 50 includes processing chamber 68 having separation and collection chambers 34 and 36.
  • the ports of processing chamber 68 communicate with multi-lumen umbilicus 240 which, in turn, communicates with donor needle 14 and containers 220a-g, either directly or through cassettes 222a-c.
  • processing circuit 50 is initially primed with saline withdrawn from container 220a.
  • the donor's blood is mixed with anticoagulant from container 220e.
  • a portion stored in reservoir container 220b and the remainder is conveyed through umbilicus 240 to separation chamber 68 where it is separated into red cells and platelet rich plasma.
  • the red blood cells are conveyed through umbilicus 240 to red cell storage container 220d.
  • the platelet rich plasma is conveyed through umbilicus 240 to cassette 222c and then back through umbilicus 240 to collection chamber 68 where the platelets are sedimented onto the hi-g wall for subsequent processing.
  • the platelet poor plasma is conveyed through umbilicus 240 to plasma reservoir container 220c.
  • plasma from container 220c and red cells from container 220d are returned to the donor, while blood held in reserve in container 220b is being processed.
  • processing chamber 68 is removed from the centrifuge, the platelets are resuspended and conveyed to platelet storage containers 220f and 220g along with sufficient plasma to provide adequate storage for up to five days .
  • the circuit is again primed with saline withdrawn from container 220a.
  • blood is again mixed with anticoagulant from container 22Oe with a portion stored in container 220b and the remainder supplied to separation chamber 68.
  • Separated, packed red blood cells are again stored in container 220d with separated plasma stored in container 220c.
  • sequestered blood from container 220b is conveyed to separation chamber 68, the separated red blood cells collected, while instantaneously separated plasma, along with that plasma previously sequestered in container 220c are returned to the patient along with replacement solution from saline container 220a.
  • Supplemental oxygen carrier held in containers 220f and 220g can also be administered to the patient during the return cycle in a predetermined quantity based upon the amount of red cells collected.
  • An additional tubing section can be provided between saline container 220a and an unused port on cassette 222a to facilitate metered control of saline administration during the return cycle.
  • Disposable circuit 50 can again be reconfigured to accomplish cell salvaging.
  • a reconfigured circuit 50 would again be primed with saline from container 220a.
  • Needle 14 would be replaced by a suction wand, not shown and of known construction, and the extra-vascular or shed blood mixed with anticoagulant from container 220e and stored in blood reservoir 220b until a sufficient quantity is obtained.
  • the stored blood is mixed with saline from container 220a, conveyed to separation chamber 68 and separated into now washed, packed red blood cells and a supernatant fluid containing blood plasma and washing solution saline.
  • the packed red blood cells are stored in container 220d until required, while the supernatant fluid is collected in waste container 220c.
  • An administration set can be provided to return the packed cells stored in container 220d to the patient by known gravity means or a separate return line (not shown) could be provided so that the washed red blood cells could be pumped directly to the patient.
  • extra- vascular or shed blood could be drawn into a stand-alone vacuum cannister (not shown, but of known construction) and withdrawn through needle 14 when processing is desired.
  • an addition tubing segment can be supplied between saline container 220a and cassette 222a to provide metered control of saline during the washing process .
  • Circuit 50 can again be reconfigured to effect such a removal .
  • the circuit can again be primed with saline from container 220a.
  • blood is again mixed with anticoagulant from container 220e with a portion stored in container 220b and the remainder supplied to separation chamber 68.
  • Separated, packed red blood cells are again stored in container 220d.
  • the separated plasma is mixed with a solvent held in containers 220f and 220g and conveyed to secondary separation stage 224 where lipid reduced plasma is produced and conveyed to plasma container 220c.
  • the solvent agglutinated lipids can be sequestered in secondary separation chamber 221, or, alternatively, an additional lumen can be provided in umbilicus 240 so that the lipids could be continuously pumped into a waste container connected into an unused port in cassette 222c (not shown) .
  • affinity based * materials could be used in place of solvents to affect removal of lipids, as described above.
  • red blood cells having Type-A, Type-B, or Type-AB antigens can be converted to Type-0 red cells by certain enzymatic treatments.
  • Disposable circuit 50 can again be reconfigured to affect such a treatment .
  • the circuit can again be primed with saline withdrawn from container 220a, or, if desired, primed with blood.
  • blood is again mixed with anticoagulant from container 220e with a portion stored in container 220b and the remainder supplied to separation chamber 68.
  • packed red blood cells are again stored in container 22Od and the separated plasma stored in container 220c.
  • the plasma is returned during the return cycle.
  • the red cells then undergo enzymatic conversion in a post processing step.
  • the packed red cells are transferred from container 220d to container 220b and mixed with enzymes from containers 220f and 220g.
  • the treated red cells are then admixed with saline from container 220a and conveyed to separation chamber 68.
  • the washed and treated red cells are again stored in container 220d, while the separated supernatant is conveyed to the now unused plasma container 220c for subsequent disposal.
  • the process of transferring the red cells from container 220d to container 220b, admixing the saline from container 220a and separated into washed, packed cells and supernatant solution in separation chamber 68 can be repeated as many times as desired.
  • a normal platelet collection procedure could be performed using disposable circuit 50 with the collected platelets stored in containers 220f and 220g, as described above.
  • a concurrent red cell product can be collected and stored in container 22Od.
  • a new container holding the enzymes would be provided and connected into the unused port on cassette 222c, so that the collected red cells could be converted to Type-O, as discussed above.
  • an additional tubing segment can be supplied between saline container 220a and cassette 222a to provide metered control of saline during the washing process.

Abstract

Automated systems and method for processing blood and other fluids are disclosed. The system and methods utilize a disposable fluid circuit (fig. 1) mounted on a re-usable hardware component or module (12). The system withdraws blood from a donor or patient (100), separates the blood into two or more components and further processes or treats the separated component.

Description

MULTI-PURPOSE, AUTOMATED BLOOD AND FLUID PROCESSING SYSTEMS AND METHODS
[0001] The present invention relates, in general, to a highly versatile, automated system for processing blood, blood components, and other fluids included in such processing. More particularly, the present invention relates to an automated system that can separate blood into two or more blood components ("apheresis") , and then perform a further procedure involving one or more of the separated components .
[0002] The term "apheresis" means removing whole blood from a patient or donor and separating the blood into two or more components. A separated component can be collected from a healthy donor, and later transfused to a patient in need of the component. Apheresis is also used in therapeutic applications to treat illness by removing diseased or otherwise undesirable components from a patient .
[0003] In a basic apheresis procedure, blood is withdrawn from a donor through a needle inserted into the vein of a donor. The needle is attached to one end of a plastic tube which provides a flow path for the blood. The other end of the tube terminates in a container for collecting the blood. The collected blood is then separated in a separator, such as a centrifuge, into its components. The desired blood component which, depending on the procedure, can be red blood cells, platelets, plasma, white blood cells or stem cells may be collected and stored for later transfusion to a patient in need of the blood component .
[0004] More recently, "automated" apheresis systems have come into widespread use. These automated systems utilize disposable, pre-sterilized fluid circuits (i.e., tubing sets) through which the blood flows. The fluid circuits are mounted on re-usable hardware devices or modules that have pumps, valves, sensors and the like. These automated systems further include an internal computer and associated software programs (controller) which control many of the processing functions.
[0005] For example, in an automated system, blood flow through the fluid circuit, the operation of valves and pumps, may be monitored and regulated by the system. An automated system can be programmed to initiate, terminate or otherwise control certain functions based on patient or donor data (e.g., height, weight, sex, hematocrit) . Likewise, an automated system may monitor certain functions with the aid of sensors which can, for example, sense the amount of the collected or withdrawn component. Optical sensors are used to measure the clarity or content of a fluid, or sense the presence or absence of certain components .
[0006] Automated apheresis systems are available from several different manufacturers. Examples of commercially available apheresis systems include the Autopheresis C® Cell Separator and the Amicus® Cell Separator, sold by Baxter Healthcare Corporation of Deerfield, Illinois. The Autopheresis C® utilizes a separator that includes a chamber and rotating membrane. Blood is introduced into the chamber and the membrane separates the blood into (at least) plasma and red blood cells, or other plasma-depleted blood.
[0007] The Amicus® Cell Separator utilizes a centrifugal separation principle. In the Amicus® Separator, whole blood is introduced into a dual-chambered or single- chambered container mounted on a rotatable centrifuge. Whole blood is introduced into the first chamber where red blood cells are separated from platelet-rich plasma (PRP) . The PRP flows into a second chamber where it is further separated into platelets and platelet-poor plasma. The disposable fluid circuit of Amicus® uses preformed cassettes with flow paths defined therein, which is mounted on the Amicus® device. Flow through the flow path is assisted by peristaltic pumps. A more detailed description of the Amicus® Separator is provided in U.S. Patent No. 5,868,696, which is incorporated herein by reference. [0008] Recently, a more, portable automated apheresis system has been developed by Baxter Healthcare Corporation. As described in U.S. Patent Application Serial No. 09/390,489, filed September 3, 1999, entitled "Self- Contained Transportable Blood Processing Device, " which is incorporated herein by reference, the portable apheresis system is also based on the principle of centrifugal separation. It includes a re-usable hardware module and a disposable fluid circuit. The fluid circuit includes a cassette with pre-formed flow paths, valving stations and pumping stations.
[0009] Other manufacturers such as Gambro BCT, Haemonetics, Dideco and Fresenius also provide automated apheresis systems based on centrifugal or other separation principles .
[0010] While efforts continue to develop and provide more efficient, economical and easy-to-use apheresis systems, concerns about the availability and safety of the blood supply, as well as an increased understanding of the role of certain blood components and blood related diseases, have led to the development of additional blood related procedures . These additional procedures often include treatment of the blood component so as to provide a safer or more viable component. Some of the additional procedures may involve eradication or removal of undesired compounds or other substances from blood. Some of these additional procedures may involve replacement of a component with another solution. In any case, these procedures often involve many manual steps, several different pieces of equipment or complex fluid circuits. Thus, there exists a need for an automated system that, in addition to separating blood into its components, can carry out one or more other procedures involving the separated components and/or the treatment thereof .
[0011] Thus, it would be desirable to provide an automated system that can perform additional procedures using a single piece of re-usable hardware and an easy-to- load, easy-to-use disposable that eliminates the need for many tubing connections and complex routing of tubing. It would also be desirable to provide a single system that does not require regular operator intervention to perform the selected separation and other treatment or processing steps. It would also be desirable to provide a system where all desired separation and processing steps are performed within a single integrated system, and "off-line" treatment using separate devices is not required. It would also be desirable to provide a system that can perform multiple fluid separation, processing and/or treatment steps through automated control of flow through the fluid circuit .
[0012] One application where automated separating and processing of blood may be desirable is in the automated pre-surgical donation of blood and administration of a replacement fluid such as a blood substitute and/or oxygen carrier. A manual version of this process is described in U.S. Patent No. 5,865,784, incorporated herein by reference .
[0013] Another application where automated separating and processing blood may be desirable is in the salvaging of red blood cells during surgery on a patient. In cell salvage, blood from a wound area or from the body cavity (i.e., extra-vascular or "shed" blood) that would otherwise be lost, is collected, processed (or cleaned), and the cleaned blood is returned to the donor. Examples of systems and apparatus used for cell salvage are described in U.S. Patent No. 5,976,388, which is incorporated herein by reference. [0014] Another application where separating and processing blood may be desirable is in the removal of unwanted substances from blood or a separated blood component such as plasma. For example, the role of cholesterol and low density lipids (LDL) in cardiovascular disease has been well documented. Methods for lipid removal from the plasma of a patient have been developed and are disclosed in U.S. Patent Nos. 4,895,558, 5,744,038 and 5,911,698, which are incorporated herein by reference. [0015] Still another application where separating and processing blood may be desirable is in the treatment of blood cells. In a particular application, it may be desirable to treat separated red blood cells with enzymes to, for example, convert Type A, B and AB blood cells to the universally acceptable Type 0 blood cells. Examples of such methods are described in U.S. patent No. 6,175,420 and 5,671,135, which are incorporated by reference herein. [0016] As described below, there may be additional applications where it may be desired to separate blood into its components for further treatment and/or processing. [0017] Thus, it would be desirable to provide a single system that, in addition to having the ability of withdrawing whole blood and separating it into two or more components, is programmed for, adaptable for, and capable of carrying out at least two or more applications.
SUMMARY OF THE INVENTION [0018] In one aspect, the present invention is directed to an automated system for processing blood and a blood component. The system includes a sterile, pre-assembled, disposable fluid circuit. The fluid circuit includes means for withdrawing blood from a patient or donor and a separation chamber. The circuit further includes means for treating the separated blood component. The fluid circuit also includes a flow control cassette having pre-formed flow path segments that are separated by valve stations for controlling communication between the flow path segments. The cassette also includes pump stations for pumping fluid through the flow path segments.
[0019] The automated system further includes a re-usable component or module adapted to cooperatively receive the fluid circuit. The re-usable component includes means cooperatively associated with the separation chamber of the fluid circuit and for effecting separation of the blood component from blood. The re-usable component also includes means for cooperating with the valve stations of the cassette to allow the flow of fluid through the preformed flow paths of the cassette. The re-usable component also includes a programmable controller for selectively controlling fluid flow through the valve stations and for selectively establishing flow communication between the flow segments.
BRIEF DESCRIPTION OF THE DRAWINGS [0020] Figure 1 is a diagram showing the re-usable hardware component or module of the present invention and some of the available disposable fluid circuits for use therewith.
[0021] Figure 2 is a perspective view of a automated system that may be employed with the present invention, including the re-usable component and the disposable fluid circuit .
[0022] Figure 2A is an enlarged, perspective view of the separation chamber of the fluid circuit of Figure 2 which can be employed in the automated system of the present invention.
[0023] Figure 3 is a perspective view of an automated system that may be employed with the present invention with a disposable fluid circuit mounted on the re-usable device. [0024] Figure 4 is a plane view of the front side of a cassette of the fluid circuit of Figure 2. [0025] Figure 5 is a plane view of the back side of the cassette shown in Figure 4. [0026] Figure 6 is a flow diagram showing the steps performed in the operation of the automated system of the present invention.
[0027] Figure 7 depicts the fluid circuit for a system and procedure embodying the present invention. [0028] Figure 8 depicts the fluid circuit for an automated hemodilution system and procedure embodying the present invention.
[0029] Figure 9 depicts the fluid circuit for an automated plasma treatment system and procedure embodying the present invention.
[0030] Figure 10 depicts the fluid circuit for an automated cell treatment system and procedure embodying the present invention.
[0031] Figure 11 depicts the fluid circuit for an automated cell salvage system and procedure embodying the present invention cell salvage procedure.
[0032] Figure 12 depicts the fluid circuit for an alternative automated plasma treatment system and procedure embodying the present invention.
[0033] Figure 12A is a perspective view of an automated system that may be employed with the present invention with a disposable fluid circuit including a separation column mounted on the re-usable component.
[0034] Figure 13 is a perspective view of the re-usable component of an alternative automated system that may be employed with the present invention.
[0035] Figure 14 is a perspective view of a fluid circuit for use with the re-usable device of Figure 13. [0036] Figure 15 is a perspective view of the fluid circuit shown in Figure 14 mounted on the re-usable component .
[0037] Figure 16 is an enlarged perspective view of a separation chamber of the fluid circuit of Figure 14. DESCRIPTION OF THE PREFERRED EMBODIMENTS
[0038] Turning now to the drawings, Figure 1 diagrammatically shows a multi-purpose blood and fluid processing system 10 embodying the present invention.
[0039] As generally shown in Figure 1, automated system 10 includes a re-usable hardware component or module 12. The re-usable hardware component 12 is particularly versatile and may be used with a variety of disposable fluid circuits. Thus, for example, hardware component 12 can be used with fluid circuits for red blood cell collection, plasma collection, platelet collection, white blood cell (leukocyte) collection, stem cell collection, hemodilution, cell salvage, lipid removal from plasma, conversion of red blood cells, cell washing, red blood cell exchange, leukoreduction, other therapeutic plasma treatments and, as will be seen, combinations of such procedures .
[0040] One embodiment of the automated, multi-purpose blood and fluid processing system that may incorporate the present invention is shown in Figure 2. As shown in Figure 2, automated system 10 includes a re-usable module 12 and a disposable fluid circuit 50 for use in association with re-usable component 12.
[0041] Fluid circuit 50 includes an array of tubing and interconnected containers typically made of a sterilizable, plastic material. Fluid circuit 50 is intended for a single use (i.e., disposable, not re-usable). As shown in Figure 2, fluid circuit 50 includes a venipuncture needle 54 for insertion into the vein of the donor or patient. This needle 54 is attached to tubing, which provides a flow path for the blood withdrawn from the donor or patient . Needle 54 can also be used to return selected components to a donor or patient in a so-called "single-needle procedure. Alternatively, circuit 50 may use a "double-needle" configuration, known to those of skill in the art, where separate needles are used for withdrawal and return. [0042] As shown in Figure 2, fluid circuit 50 includes several containers for temporary and/or longer-term storage of the separated components, and for holding fluids used during the procedure, such as an anticoagulant, saline, and any other treatment or replacement fluids required for the procedure. Containers 56, 57, 58, 60, 62, 64, and 66 are also typically made of a sterilizable, plastic material.
[0043] Fluid circuit 50 further includes separation chamber 68. Separation chamber 68 is intended for mounting on the separator of the re-usable device 12. As shown generally in Figure 2, and in more detail in Fig. 2A, in one embodiment, separation chamber 68 may be pre-formed by injection molding from a rigid, biocompatible plastic material, such as a non-plasticized medical grade acrilonitrite-butadiene-styrene (ABS) .
[0044] As further shown in Figure 2A, separation chamber 68 includes a base 388 with a center hub 120. Hub 120 is surrounded radially by inside and outside annular walls 122 and 134, which define a circumferential blood separation channel 126. Alternatively, chamber 68 may include first and second sub-chambers. The bottom of channel 126 is closed by a molded annular wall. The top of channel 126 is closed by a separately molded flat lid (not shown) , which can be secured to the top of chamber 68 by welding or other securing means .
[0045] Chamber 68 also includes passageways 142, 144 and 146, which extend from hub 120 and communicate with channel 126. During processing, blood is introduced into passageway 146 at the underside of base 388 via an attached multi-lumened tube or umbilicus 69 (shown as in Figure 2) . Blood enters the channel 126 where it is separated into heavier and lighter components. The heavier components occupy the outer periphery of the channel, while the lighter component occupies the channel interior. The separated components are withdrawn through passageways 142 and 144. Introduction and separation of blood using chamber 68 is described in more detail in U.S. Patent Application Serial No. 09/390,489, previously incorporated by reference.
[0046] Fluid circuit 50 further includes a cassette 70 which provides a network of flow path segments in fluid communication with and in association with numerous valving and pumping stations. Cassette 70 provides a centralized, programmable, integrated platform for all the pumping and valving functions required for a given blood processing procedure. A more detailed view of cassette 70 is provided in Figures 4 and 5. Cassette 70 interacts with the pneumatically actuated pump and valve station 30 on re-usable module 12 described below.
[0047] As shown in Figures 4 and 5, cassette 70 has an array of interior cavities formed on both the front and back sides. The interior cavities define the valve stations and flow paths. Pump stations PP1 to PP4 are formed as wells that are open on the front side of the cassette 70. The valves VI to V23 are likewise formed as wells that are open on the front side of cassette 70. The liquid flow paths FI to F38 are formed as elongated channels that are open on the back side of cassette 70, except for liquid paths at F15, F23, and F24, which are formed as elongated channels that are open on the front side of the cassette 70. The pre-molded ports PI to P13 extend out along two side edges of the cassette 70. As shown in Figure 3, the cassette 70 is vertically mounted for use in the pump and valve station 30 described below. In this orientation, ports P8 to P13 face downward, and the ports PI to P7 are vertically stacked one above the other and face inward.
[0048] Cassette 70 is preferably made of a rigid, medical grade plastic material. Flexible diaphragms overlay both of the front side and back side of cassette 70. The diaphragms are preferably made of flexible sheets of medical grade plastic. The diaphragms are sealed about their peripheries to the peripheral edges of the front and back sides of cassette 70. Interior regions of the diaphragms can also be sealed to interior regions of the cassette body.
[0049] The action of the pump and valve stations is controlled by a pneumatic pressure source which supplies negative and positive air pressure. As shown generally in Figure 2 and described in more detailed in U.S. Patent Application Serial No. 09/390,489, under the control of the controller 11, a pneumatic pressure source selectively distributes the different pressure and vacuum levels to the pump and valve stations. These levels of pressure and vacuum are systematically applied to the cassette 70 to route blood and processing fluids. The details of the cassette, the pump and valve station 30, and the transport of blood and processing fluids through the cassette are set forth and described in U.S. Patent Application Serial No. 09/390,489, previously incorporated by reference. [0050] Turning now to the re-usable hardware component or module, re-usable component 12 includes (at least one) means for effecting separation of blood components or "separator" 20. In one embodiment, separator 20 is cooperatively associated with the chamber 68 of the fluid circuit. In a more particular embodiment, separator 20 is adapted to receive the separation chamber of the fluid circuit (described above) and effects separation of whole blood or a blood fraction into two or more components . In a preferred embodiment, separator 20 may be a rotatable centrifuge. However, it will be understood that separator 20 is not limited to a separator that utilizes a centrifugal separation principle. Accordingly, separator 20 may employ a different separation principle, such as a magnetic drive for receiving a spinning membrane as described, for example, in U.S. Patent No. 5,194,145. In another embodiment, separator 20 may also be a separation column with its own integral chamber or passageway. Separator 20 may also be a filter. In the preferred embodiment, where separator 20 is a centrifuge, the spinning action of the centrifuge separates the blood components (within separation chamber 68) by density. For example, the spinning action of the centrifuge can separate whole blood into the more dense red cell component and a less dense plasma component.
[0051] Re-usable component 12 also houses the internal computer or controller 11. The programmable controller includes pre-programmed instructions for carrying out several different blood and fluid processing procedures, allowing the operator to select from a menu, the particular procedure or procedures desired. The controller also includes pre-programmed instructions which selectively activate pumping of fluid and the opening and closing of valves in the fluid circuit described above. The controller may also include data storage capability for storing donor/patient information, processing or collection information and other data for later downloading or compilation.
[0052] As further shown in Figure 2, re-usable device 12 includes a control panel such as flat screen display 24 for displaying the status of the procedure as well as providing a touch panel screen to allow for operator interface with the system. Data output capability may also include standard parallel or serial ports or other network connection capability, as desired, for communication with other computers or networks .
[0053] Device 12 further includes pump and valve station 30. As indicated above, pump and valve station 30 is designed to mate with corresponding structures of cassette 70. Pump and valve station 30 contains four pump actuators PA1 to PA4 and twenty-three valve actuators VA1 to VA23. The pump and valve actuators are oriented to form a mirror image of the pump stations PP1 to PP4 and valve stations VI to V23 of cassette 70. During operation, pump and valve station 30 (and, more specifically, pump and valve actuators PA(N) and VA(N)), apply positive and negative pressure upon cassette 70 and the corresponding pump and valve stations therein, to direct liquid flow through the flow path segments defined therein. Access to pump and valve station 30 is obtained through door 34. [0054] All of the above-described parts of re-usable device 12, such as separator 20, controller 11, pump and valve station 30, and display screen 24 are mounted inside a portable housing or case 38. Case 38 is suited for setup and operation upon a table top or other smooth and flat surface. Case 38 includes a base 39 and hinged lid 40 which opens and closes. Lid 40 includes latch 42 for releasably locking the lid. Lid 40 further includes a handle 44, which the operator can grasp for easy transport of case 38 to a collection site, hospital, etc. Case 38 is made by molding and, preferably, of a light-weight, durable plastic material.
[0055] For supporting containers in a hanging position, lid 40 includes hooks (not shown) for hanging containers of saline, anticoagulant or other treatment or processing fluid. Similarly, a retractable hanger 45 is provided for supporting one or more collection containers in which whole blood and/or separated blood components are (at least temporarily) stored. Hanger 45 and hooks are preferably mounted on a scale 47 within lid 45 to allow automated measurement of the amount of whole blood or blood component collected.
[0056] Inclined container support surface 48 provides additional areas within the case for supporting containers associated with the disposable circuit 50. One or more areas of the support surface 48 may be heated, if desired, to warm the solution of the container prior to infusion to the donor or patient . [0057] As discussed above, controller 11 includes a micro-processor and pre-programmed software. Although some interface and involvement by the operator is required, many of the functions of the automated system 10 are automatically controlled by controller 11.
[0058] For example, as shown in Figure 6 after the operator turns on the power to the re-usable device 12, the system automatically undergoes a system check procedure to confirm that all electrical and mechanical components of the device 12 are functioning properly and within preset parameters. If during the system check the controller detects a problem, the system may generate an audible alarm which prompts the operator to intervene. If the system successfully completes the system check, the system may prompt the operator to select the desired procedure.
[0059] As shown in Figure 6, the automated system allows the operator to select from a variety of procedures. For example, the operator may select a red cell collection
(apheresis) procedure, a plasma collection procedure a platelet collection procedure, a white blood cell collection procedure, a stem cell collection procedure.
[0060] In addition, the operator may select from a one or more other additional procedures. Thus, the operator can select a first procedure to separate whole blood into two or more components, and also instruct the system to perform another additional treatment or other processing procedure including the separated componen (s) . Alternatively, the operator may directly select one of the additional procedures, which already combines aspects of the above-mentioned apheresis procedures with additional "downstream" blood processing and/or treatment protocols.
[0061] As shown in Figure 6, these additional procedures include procedures for hemodilution, plasma treatment, such as lipid removal, the conversion of cells, cell salvage and other procedures including, but not limited to, therapeutic plasma treatments, removal of certain compounds from plasma using monoclonal antibodies, magnetic, para-magnetic, and other beads. Additional supplemental procedures involving the separated components of blood may also be performed. [0062] In any event, once the operator has selected the desired procedure, under the control of the controller 11, the system prompts the operator to load the appropriate fluid circuit. Referring back to Figure 1, it is shown that re-usable device 12 is adapted to receive any one of a variety of disposable fluid circuits. Each of the enumerated procedures may require its own unique disposable fluid circuit or, more preferably, a disposable fluid circuit will be suitable for two or more procedures. [0063] Most of the disposable fluid circuits will have many common elements such as a venipuncture needle, anticoagulant container, saline container, storage containers for red blood cells, plasma or whole blood, a separation chamber and the cassette 70. As mentioned above, each procedure may have its own unique disposable fluid circuit. However, it is also possible that a universal fluid processing circuit can be used and any additional required containers of fluid (e.g., treating agents) , additional separators or other components can be easily attached at the time of use. These additional fluid circuit components can be connected to the universal set in a sterile manner in ways that are well known to those of skill in the art. Any additional tubes or flow paths can be attached to existing ports (e.g., P13) on the cassette and the system programmed to perform the additional procedures .
[0064] The versatility of the cassette, with its flow path segments that can be interconnected in a variety of ways through selective opening and closing of valves coupled with the programmable microprocessor in the controller lends itself particularly well to the automated system of the present invention. It allows the system to be used with different fluid circuits, to perform a variety of different procedures or protocols, and allows the system to combine aspects of apheresis with additional downstream treatment or processing of blood components.
[0065] Once the fluid circuit 50 has been mounted on the re-usable device 12, the system, under the control of the controller 11, will verify that the correct disposable has been loaded and/or that it has been loaded properly. Once proper loading of the disposable processing circuit has been confirmed, the system will automatically initiate a priming sequence based on the selected protocol . Typically, the priming sequence will include priming the fluid circuit with anticoagulant and/or saline. In addition, if a particular treatment or replacement fluid is intended for use in a particular procedure, the system may also prime the disposable fluid circuit with such fluid or the patient's or donor's blood.
[0066] The system may allow the operator to enter desired patient or donor data, such as height, weight, gender, hematocrit, or any other donor or patient characteristic that the controller may utilize during the course of the procedure . Entry of donor data may occur before or after prime. For example, the system may use the above-described donor data to determine flow rates, and/or duration of a particular step. After prime and entry of any required donor data, the system prompts the operator to begin the procedure .
[0067] Figure 7 shows an automated system embodying the present invention including a typical fluid circuit. As shown in Figure 7, whole blood is withdrawn from donor 100 and introduced into cassette 70 via line (tubing) 74. Anticoagulant from container 56 is likewise drawn into cassette 70. Anticoagulant enters through one of the ports
(e.g., P10) of cassette 70. Controller opens the selected valve (s) to allow anticoagulant flow through the fluid segment, and establishes flow communication between the anticoagulant line 75 and line 74 to combine anticoagulant with the whole blood being withdrawn from the patient. [0068] Anticoagulated whole blood is introduced into container 58, which serves as an interim whole blood source. As described above, the hardware component 12 can include weight scales. Thus, container 58 is suspended from a weight scale so that when the required amount of whole blood is collected a sensor attached to the weight scale prompts the controller and the draw cycle is terminated. The controller then initiates pumping (by controlling the pump and valve station) of whole blood from container 58 into the separator.
[0069] The separator separates whole blood into two or more components. (It will be understood that the separator may be cooperatively associated with separator chamber 68 of the fluid circuit, either physically or as described in, for example, U.S. Patent No. 5,194,195, incorporated herein by reference.) In one embodiment, separation of whole blood results in a plasma component and a red blood cell component. The separated plasma and red cell components may be withdrawn from separation chamber 68 and collected in separate containers 60 and 62 for temporary storage. [0070] At this point, depending on the procedure or protocol selected (see Figure 6) , further processing of one or both of the separated components may be initiated by the system. Thus, for example, if the additional procedure involves treatment of the separated component, fluid circuit 50 may include a treatment fluid container 64. In another embodiment, if the additional procedure requires administration of a replacement fluid (to, for example, provide the biological function of the withdrawn component) , container 64 may include a replacement fluid. [0071] In any event, further processing or treatment of the separated component may take place in separation chamber 68, one of the containers 60 or 62, or, if required, a second and separate separator (shown in dashed lines in Figure 7 and labeled "Separator 2"). The second separator may utilize the same separation principle as Separator 1. Thus, in one embodiment, both Separators 1 and 2 may be centrifuges. In another embodiment, one of the separators may be a centrifuge while the other separator may be a drive mechanism for cooperation with a rotating member and separation membrane of the type described in U.S. Patent No. 5,194,145. In another embodiment, one of the separators may be a centrifuge or a drive for a rotating membrane and the other separator can be a filter medium or a separation column. [0072] The blood component separated in Separator 1 can be directly introduced into Separator 2 for further treatment and/or processing. Alternatively, blood component can be introduced into cassette 70 from where it can be directed and/or pumped into Separator 2. Likewise, upon exiting Separator 2, the separated (and/or treated) component can be directly reinfused back to the donor, as shown by dashed line 87, or through cassette 70 and return line 84.
[0073] Although Figure 7 shows a single vein access point (i.e., single needle) for withdrawal of blood and return of blood component, it will be understood that the fluid circuit shown in Figure 7 (or any one of the other Figures 8-12) may also utilize a so-called "double-needle" configuration described above.
[0074] Once treatment is complete, the desired component (red blood cells or plasma) can be returned to the donor or patient via line 84. As will be described below, depending on the procedure, there may be variations to the general separation and processing sequence described above. [0075] A more particular example of a system embodying the present invention is shown in Figure 8. Figure 8 shows an automated system and procedure for the automated pre- surgical withdrawal of blood, separation into plasma and red blood cells, followed by the return of plasma and infusion of one or more replacement fluids (i.e., "hemodilution" ) . [0076] A system of the type shown in Figure 8 is particularly useful in the collection of autologous blood from a patient just prior to a surgical procedure. The plasma component is returned and is supplemented with a volume replacement fluid (such as saline) and a blood substitute which can provide the same biological function (i.e., oxygen transport) as the collected red blood cells. [0077] As shown in Figure 8, whole blood is withdrawn from a patient just prior to the surgical procedure. The whole blood is withdrawn through line 74 and introduced into cassette 70 in the manner generally described above. [0078] Whole blood is combined with anticoagulant and the anticoagulated whole blood may be introduced into container 58 or immediately introduced into the separator. Once inside the separator and, more particularly, the separation chamber 68 associated with the separator, whole blood is separated into a red blood cell component and a plasma component. The red cell component is removed from the separator (by pumping of the pump stations in the cassette 70) and collected in container 60 where it is stored until needed (if needed) during or after surgery. If long-term storage of red cells is required, the collected red cells may be combined with a red blood cell preservative solution such as Adsol® or Erythro-Sol, available from Baxter Healthcare Corporation of Deerfield Illinois. Administration of a preservative (stored, for example, in container 57 (Figure 2) can also be controlled by controller 11.
[0079] The separated plasma can be introduced into container 62 from where it can be metered back to the patient during surgery or, in the alternative, immediately returned to the patient. In another alternative, plasma (with platelets) may be returned to the patient after surgery, at or around the time that the red blood cells are returned. [0080] In order to compensate for the lost volume of red blood cells, a volume replacement fluid such as saline may also be administered to the patient. In addition, because red blood cells include hemoglobin, an oxygen carrying compound in blood, a blood substitute or other synthetic oxygen carrying compound that can perform the same oxygen transport function as the red blood cells may also be administered to the patient. Such blood substitutes and/or oxygen carrying compounds are known and are available from Alliance Pharmaceutical Corporation of San Diego, California, and are described in U.S. Patent No. 5,865,784. Other blood substitutes known to those of skill may also be used.
[0081] The blood substitute may be combined with the saline or administered separately either before or after administration of the saline. In addition, other volume replacement fluids in lieu of saline (which is a crystalline solution) may also be used as the volume replacement fluid. This includes colloidal solutions, such as dextran and albumin.
[0082] In accordance with a present invention, the system 10 can automatically determine the amount and flow rate of the fluids, i.e., saline and a blood substitute required. In one embodiment, the controller can be preprogrammed to administer the selected amount of saline or other fluid and a replacement fluid having a known biological function, such as a blood substitute, based on the amount of the red blood cells collected as measured by the weight scale 47 in hardware component 12. Alternatively, the system can determine the amount (and flow rate) of the replacement fluid to be administered based on the amount of whole blood withdrawn. In still another alternative, the system can determine the amount of replacement fluid and blood substitute to be administered based on donor data entered at the beginning of the procedure. In any event, the automated system of the present invention provides benefits that manual hemodilution cannot achieve.
[0083] For example, by separating whole blood into red cells and plasma, and returning the plasma to the patient, the extra-corporeal volume of blood is reduced as compared with the manual systems where whole blood is withdrawn. This results in several benefits not available in "manual" hemodilution.
[0084] In the manual hemodilution procedure, the hemoglobin concentration of the blood is reduced from approximately 12 mg/dl to 9 mg/dl by withdrawing blood and administrating support fluid (3 times the saline or albumin) . This represents a total whole blood volume removed of approximately IL. To replace this lost volume either IL of albumin or 3L of saline would have to be administered. Three times the volume of saline is necessary due to saline's limited ability to stay within the vascular space. Albumin, being a molecule of larger size, can stay within the vascular space and will not be as quickly excreted as saline.
[0085] In accordance with the present invention, because the plasma component is returned to the patient, the volume of fluid removed would be limited to the red blood cell volume which would be approximately 400 ml (based on an average, hematocrit of 40%) . When IL of whole blood is removed with a 40% hematocrit, the total volume of red blood cells removed is 400 ml, with the remaining 600 ml consisting of plasma. To remove an equal amount of red blood cells using the automated system and procedure would require the removal of only 400 ml of concentrated red blood cells with all of the plasma processed by the system being returned to the patient. This reduces the volume removed by 60%. To replace this, only 400 ml of albumin or 1,200 ml of saline would be necessary. This is substantially less than the typical manual hemodilution procedure .
[0086] By reducing the volume of saline administered, any potential fluid complication caused by saline can be reduced. Saline can cause fluid overload and tissue edema in patients with renal insufficiency. A large volume saline infusion and associated increase in tissue fluid can necessitate the need for diuretic administration to assist in fluid removal after the surgical procedure.
[0087] Another advantage of the automated system of the present invention is that the system can be programmed by the anesthesiologist and the procedure accomplished automatically. The system can add the appropriate amount of anticoagulant to the blood to prevent clotting in the blood storage container and red cell additive solutions can be used as necessary.
[0088] Citrate anticoagulation can cause some citrate reactions in patients during apheresis procedures. Citrate reactions are usually controlled by infusion of calcium containing solutions. Using the automated system of the present invention, when the collected blood products are transfused back to the patient, the minimum amount of anticoagulant will be present in the collected blood which, upon transfusion, should cause fewer complications due to citrate transfusion compared to the manual method.
[0089] By reducing the volume of fluid removed, the time until blood is to be administered may be prolonged. More importantly, this automated system and procedure can reduce or eliminate the need for non-autologous blood. By eliminating or reducing the need for non-autologous blood, the patient can have limited exposure to non-autologous homologous blood. This can reduce the possibility of post transfusion immunosuppression or inflammatory response due to transfusion of stored blood (cytokine generation during storage) .
[0090] The automated system of the present invention will allow for the plasma (and platelets) to be returned to the patient. By returning the plasma (and platelets), the patient can more easily maintain normal hemostasis. (In the standard manual hemodilution procedure, severe dilution can cause hemostasis problems which may require infusion of cryoprecipitated clotting proteins (cryoprecipitate) or fresh frozen plasma (FFP) . This also occurs during the manual procedure because whole blood is removed which removes platelets and plasma as well as RBC's.) [0091] Figure 9 shows another application of the automated system of the present invention. In particular, Figure 9 shows a procedure that results in removal of undesired compounds from blood plasma. More particularly, the fluid circuit and flow system shown in Figure 9 can be used for removal of lipids from the plasma of the patient. [0092] As shown in Figure 9, whole blood is withdrawn from a patient 100 via venipuncture and allowed to flow through line 74 into cassette 70, and combined with anticoagulant as previously described. The anticoagulated whole blood may be collected in container 58 until a selected weight is attained. Once the desired amount of whole blood has been collected, under control of the controller 11, the system introduces whole blood into the separator, which can include or is otherwise cooperatively associated with separation chamber 68, where it is separated into red blood cells and plasma. The separated red blood cells may then be returned to the patient immediately, or temporarily stored in container 60 for later return.
[0093] The separated plasma may then be further treated to remove lipids (or any other undesirable compounds) . In one embodiment, plasma may be combined with a solvent contained in container 64. The solvent is capable of extracting lipids from the plasma. Such solvents are described in, for example, U.S. Patent Nos. 4,895,558, 5,744,558 and 5,911,698, which are incorporated herein by reference. Examples of solvents are DIPE (di- isopropylether) . Of course, other solvents capable of extracting lipids from plasma and known to those of skill in the art may likewise be used.
[0094] Plasma and the solvent may be combined in, for example, container 62 or inside separator 68. If combined outside of the separator, the plasma and solvent may then be reintroduced into the separator to further separate plasma from the lipid containing solvent. In a preferred embodiment, the separator is a centrifugal separator of the type shown in Figures 2-3 and/or Figures 13-16. Centrifugal action results in the separation into a two- phase solution, an upper organic phase that includes the solvent and extracted lipid, and a lower lipid-depleted plasma phase. Under control of the controller, the lipid containing solvent may then be pumped to a separate waste container. The lipid-depleted plasma may be returned to the patient.
[0095] Because some of the solvents that may be useful in removing lipids from the plasma may (in certain concentrations) be harmful to the patient, a further processing step that involves purging or otherwise removing any residual solvent from the plasma may be preferred. Thus, after removal of the organic phase, the plasma may be treated with a further washing solution from container 69. Treatment in the washing solution can take place in the separation chamber before return of the plasma to the patient.
[0096] Alternatively, as shown in Figure 9, system 10 may include a second separator (Separator 2) for the cleansing and/or washing step. As set forth above, Separator 2 may employ the same separation principle (e.g., centrifugation) as Separator 1, or more preferably, may employ a different separation principle. In one embodiment, Separator 2 may be a column packed with coated beads that have an affinity for the solvent. Thus, plasma may be removed from Separator 1 and introduced into column Separator 2 (either directly or via cassette 70) to remove any residual solvent. Plasma that has been passed through Separator 2 may then be suitable for return to the patient. In another alternative, Separator 2 may be filter medium. The system can include an optical detector 83, which is capable of detecting lipids in the plasma being returned. Such detectors are described in U.S. Patent No. 5,958,250, incorporated herein by reference.
[0097] In still another embodiment of the automated system of the present invention, removal of undesired compounds from plasma may be achieved without resort to a solvent-based system. Instead, plasma that has been separated in the separator may be treated or contacted with another material for removing lipids from plasma. For example, in one embodiment, a blood component that has been separated from whole blood can be further treated with particles or beads that have a specific affinity for the compound to be removed. As shown in Figure 10, container 64 may include the beads or particles. In a preferred embodiment, the beads may be lightweight, simple, hollow (or solid) sphere-like structures. The beads are coated with an affinity material, such as monoclonal antibodies. The beads may have a specific affinity for lipids, sickled cells, immunoglobulins, Factor VIII or other proteins. The beads, preferably, have a density less than the density of plasma. Alternatively, the beads may be of the type described in U.S. Patent Nos. 5,916,743 and 5,641,622, which are incorporated herein by reference. [0098] In any event, as shown in Figure 10, whole blood is withdrawn from the donor (or patient) through line 74 and combined with anticoagulant as previously described. Anticoagulated whole blood is collected and temporarily stored in container 58. When a predetermined amount of whole blood has been collected, under the control of the controller, whole blood is introduced in the separator where it is separated into a cellular component and a plasma component. The cellular component can be removed from the separator and collected in, for example, container 60. The plasma component can be combined with beads in container 60 or, to ensure greater contact between the beads and the compound to be removed, in the separator. The controller will cause beads to be pumped into either container 62 or the separator. The bound particle can then be collected in container 62 and the plasma returned to the donor. Alternatively, in another embodiment, plasma may be passed through a filter or other type of medium that has attached to its surface monoclonal antibodies that have a specific affinity for lipids. The filter medium may be a flat sheet or a packed column of the type described above. In addition, the separation medium (e.g., separator 80) may be used to extract or remove lipids or other compounds (through affinity separation) such as IgG, IgM, Factor VIII, and the like from plasma.
[0099] Another application for the automated system of the present invention is in the treatment of blood cells, such as red cells, white cells or platelets. In one specific embodiment, the automated system can be used to treat red blood cells with an enzyme to convert, for example, Type-A, Type-B, or Type-AB red blood cells to Type-0 red blood cells. Accordingly, as shown in Figure 10, whole blood is withdrawn from a patient 100, anticoagulated in the manner described above, and separated in separator 68 to provide a red blood cell component and a plasma component. The plasma component can be collected in container 60 or can be returned to the donor immediately. The red blood cell component can be temporarily collected in container 60. Under the control of controller 11, the red blood cell component can be combined with a solution (stored in container 64) that includes a particular enzyme suitable for the red blood cell conversion. Examples of such enzymes are included in U.S. Patent Nos. 6,175,420 and 5,671,135, which are incorporated by reference herein. The treated red blood component may then be collected and stored in container 62. In addition, if Type-0 blood cells are to be stored long term (e.g., up to 42 days), a preservative solution of the type described above can be added to the red blood cells. In another treatment-type application, red blood cells, platelets or even plasma may be treated to eradicate or inactivate pathogens present in these components. [00100] Another application of the automated system of the present invention can be the salvage of blood during a surgical procedure. As shown, for example, in Figure 11, whole blood can be collected from the body cavity of a patient 100 undergoing surgery. In this embodiment, fluid circuit will include a suction device 120 instead of a venipuncture needle. Suction device 120 maybe of the type shown in, for example, U.S. Patent No. 5,976,388, which is incorporated herein by reference. Blood that is removed by suction device 120 is introduced into the separator where it is separated into a red blood cell component and supernatant. The red cell product may then the returned to patient .
[00101] Turning briefly to Fig. 12, an alternative, automated system for treating separated plasma is shown. The system includes a first separator and a second separator. As shown in Figure 12A, the second separator is a separation column 80 that can be used to remove the above-described compounds from plasma. Columns that can be used for such separation are generally disclosed in U.S. Patent Nos. 5,733,254 and 5,782,792, which are incorporated herein by reference.
[00102] As shown in Figure 12, separation of plasma from whole blood proceeds as generally described above, i.e. , in the separator. The separated plasma may be introduced into column 80. It should be noted that plasma can be directly introduced into column 80 via line 81, or can be conveyed by the pumps and valves of cassette 70 (under the direction of the controller) to column 80. Likewise, plasma that has passed through column 80 can be returned via cassette 70, can be directly introduced into line 74 for direct return to the donor, or can be introduced into container 62 from where it is pumped (through cassette 70) back to the donor. Red cells in container 60 may be returned to the donor during processing of plasma.
[00103] Column 80 may be provided as part of the fluid circuit 50. In one embodiment, re-usable component 12 can be equipped with clips 13 and 15 for holding column 80, as generally shown in Figure 12A.
[00104] Figures 13-16 show an alternative embodiment of another re-usable hardware device and fluid circuit that can be used in the automated system and procedure of the present invention. The embodiment shown in Figures 13-16 include a centrifuge assembly 200 and a fluid processing circuit 50 for use in association with the centrifuge assembly. The centrifuge assembly includes a re-usable hardware device capable of long-term use. The disposable fluid circuits, like the fluid circuits described above, are intended to be a single-use, disposable item. [00105] Like the disposable fluid circuits described above, the fluid circuits shown in Figures 13-16 include a processing chamber, shown in Figure 16, that can be loaded onto a separator of the re-usable device, to centrifugally separate blood components. The separator may separate whole blood into a red blood cell component, a plasma component, a white blood cell component, stem cells or a platelet component. The disposable fluid circuit also includes an array of flexible tubing to convey liquid to and from the processing chamber, described in more detail below.
[00106] Fluid circuit 50 includes one or more cassettes 222A, B and C, generally of the type described above. The cassettes shown in Figures 14-15 include inter-connectable flow segments and valving stations . In contrast to cassettes 70 described above in connection with other embodiments, the cassettes of this embodiment do not include internal pumping stations. Instead, the cassettes of this embodiment include external tubing loops 223 which engage peristaltic pump rotors 250, which effect movement of fluid through the tubing and the fluid circuit. The details of this embodiment of the automated system are described in U.S. Patent No. 5,868,696, which is incorporated herein by reference.
[00107] As shown in Figure 16, in the embodiment of Figures 13-16, fluid circuit 50 includes a "two-staged" separation chamber 68. Thus, the first sub-chamber 226 can be used to perform a first separation step and the second sub-chamber 224 can be used to perform a second separation step. For example, where a blood component such as plasma or red blood cell is to be treated with a treating agent or described above, plasma can be separated from red cells in the first "sub-chamber" 226 and the treatment carried out in the second "sub-chamber" 224. A treating agent can be directly introduced into the second subchamber or can be combined with the component elsewhere, such as in one of the containers .
[00108] The second subchamber can also be used to remove undesirable solvents, compounds, treating agents from the separated component. In most other respects, the blood and fluid processing procedures described above are applicable to the automated system described and shown in Figures 13- 16. Of course, the chamber 68 may have only a single chamber .
[00109] More particularly, disposable circuit 50 shown in Figure 14 is adapted for single needle platelet collection. Circuit 50 includes processing chamber 68 having separation and collection chambers 34 and 36. The ports of processing chamber 68 communicate with multi-lumen umbilicus 240 which, in turn, communicates with donor needle 14 and containers 220a-g, either directly or through cassettes 222a-c.
[00110] In a typical apheresis procedure, processing circuit 50 is initially primed with saline withdrawn from container 220a. During the draw cycle, the donor's blood is mixed with anticoagulant from container 220e. A portion stored in reservoir container 220b and the remainder is conveyed through umbilicus 240 to separation chamber 68 where it is separated into red cells and platelet rich plasma. The red blood cells are conveyed through umbilicus 240 to red cell storage container 220d. The platelet rich plasma is conveyed through umbilicus 240 to cassette 222c and then back through umbilicus 240 to collection chamber 68 where the platelets are sedimented onto the hi-g wall for subsequent processing. The platelet poor plasma is conveyed through umbilicus 240 to plasma reservoir container 220c. During the return cycle, plasma from container 220c and red cells from container 220d are returned to the donor, while blood held in reserve in container 220b is being processed. After the donation is completed, processing chamber 68 is removed from the centrifuge, the platelets are resuspended and conveyed to platelet storage containers 220f and 220g along with sufficient plasma to provide adequate storage for up to five days .
[00111] In accordance with the present invention, many different and additional procedures can be performed with the system shown in figures 13-16, by reconfiguring the interconnections of disposable circuit 50 and providing different containers 220a-g and processing chamber 68. One such reconfiguration provides for the collection of mono- nuclear cells and is described in U.S. Patent No. 5,980,760, which is incorporated herein by reference. The flexibility to reconfigure the functions and characteristics of disposable circuit 50 is provided, in large part, by the versatility of cassettes 222a-c. Several such different procedures are described below. [00112] For example, when the system of Figures 13-16 is used for hemodilution, some red cells are stored for subsequent transfusion, a replacement solution is provided and a supplemental oxygen carrier may be also supplied. As in the mono-nuclear cell procedure of U.S. Patent No. 5,980,760, only a single separation chamber is required. Thus, the system can be supplied with a single chamber, or the dual chambered embodiment may be used, but only utilizing sub-chamber 226.
[00113] The circuit is again primed with saline withdrawn from container 220a. During the draw cycle, blood is again mixed with anticoagulant from container 22Oe with a portion stored in container 220b and the remainder supplied to separation chamber 68. Separated, packed red blood cells are again stored in container 220d with separated plasma stored in container 220c. During return, sequestered blood from container 220b is conveyed to separation chamber 68, the separated red blood cells collected, while instantaneously separated plasma, along with that plasma previously sequestered in container 220c are returned to the patient along with replacement solution from saline container 220a. Supplemental oxygen carrier held in containers 220f and 220g can also be administered to the patient during the return cycle in a predetermined quantity based upon the amount of red cells collected. An additional tubing section can be provided between saline container 220a and an unused port on cassette 222a to facilitate metered control of saline administration during the return cycle.
[00114] During the cell salvaging procedure, a patient's extra-vascular ("shed") blood is withdrawn from the surgical field, washed, and returned to the patient. Disposable circuit 50 can again be reconfigured to accomplish cell salvaging. A reconfigured circuit 50 would again be primed with saline from container 220a. Needle 14 would be replaced by a suction wand, not shown and of known construction, and the extra-vascular or shed blood mixed with anticoagulant from container 220e and stored in blood reservoir 220b until a sufficient quantity is obtained. Upon processing, the stored blood is mixed with saline from container 220a, conveyed to separation chamber 68 and separated into now washed, packed red blood cells and a supernatant fluid containing blood plasma and washing solution saline. The packed red blood cells are stored in container 220d until required, while the supernatant fluid is collected in waste container 220c.
[00115] An administration set can be provided to return the packed cells stored in container 220d to the patient by known gravity means or a separate return line (not shown) could be provided so that the washed red blood cells could be pumped directly to the patient. Alternatively, extra- vascular or shed blood could be drawn into a stand-alone vacuum cannister (not shown, but of known construction) and withdrawn through needle 14 when processing is desired. As with the hemodilution application above, an addition tubing segment can be supplied between saline container 220a and cassette 222a to provide metered control of saline during the washing process .
[00116] During lipid removal, lipid are removed from a patient's blood. Circuit 50 can again be reconfigured to effect such a removal . The circuit can again be primed with saline from container 220a. During the draw cycle, blood is again mixed with anticoagulant from container 220e with a portion stored in container 220b and the remainder supplied to separation chamber 68. Separated, packed red blood cells are again stored in container 220d. The separated plasma is mixed with a solvent held in containers 220f and 220g and conveyed to secondary separation stage 224 where lipid reduced plasma is produced and conveyed to plasma container 220c. The solvent agglutinated lipids can be sequestered in secondary separation chamber 221, or, alternatively, an additional lumen can be provided in umbilicus 240 so that the lipids could be continuously pumped into a waste container connected into an unused port in cassette 222c (not shown) . Alternatively, affinity based * materials could be used in place of solvents to affect removal of lipids, as described above. [00117] As previously discussed, red blood cells having Type-A, Type-B, or Type-AB antigens can be converted to Type-0 red cells by certain enzymatic treatments. Disposable circuit 50 can again be reconfigured to affect such a treatment . The circuit can again be primed with saline withdrawn from container 220a, or, if desired, primed with blood. During the draw cycle, blood is again mixed with anticoagulant from container 220e with a portion stored in container 220b and the remainder supplied to separation chamber 68. Separated, packed red blood cells are again stored in container 22Od and the separated plasma stored in container 220c. The plasma is returned during the return cycle. The red cells then undergo enzymatic conversion in a post processing step. The packed red cells are transferred from container 220d to container 220b and mixed with enzymes from containers 220f and 220g. The treated red cells are then admixed with saline from container 220a and conveyed to separation chamber 68. The washed and treated red cells are again stored in container 220d, while the separated supernatant is conveyed to the now unused plasma container 220c for subsequent disposal. The process of transferring the red cells from container 220d to container 220b, admixing the saline from container 220a and separated into washed, packed cells and supernatant solution in separation chamber 68 can be repeated as many times as desired.
[00118] Alternatively, a normal platelet collection procedure could be performed using disposable circuit 50 with the collected platelets stored in containers 220f and 220g, as described above. A concurrent red cell product can be collected and stored in container 22Od. A new container holding the enzymes would be provided and connected into the unused port on cassette 222c, so that the collected red cells could be converted to Type-O, as discussed above. As with the hemodilution application above, an additional tubing segment can be supplied between saline container 220a and cassette 222a to provide metered control of saline during the washing process.
[00119] The many procedures discussed above have been based upon the single needle disposable circuit 50 of Figure 13, but it should be appreciated by those skilled in the art that a two-needle circuit can also be modified to accomplish the desired procedures as well.
[00120] The various features of the present invention are set forth in the attached claims.

Claims

THAT WHICH IS CLAIMED;
1. An automated system for separating and processing blood and blood components of a donor or patient comprising:
(a) a sterile, pre-asse bled, disposable fluid circuit module comprising:
(i) means for withdrawing blood from a patient or donor,
(ii) a separation chamber,
(iii) means for treating said separated blood component ,
(iv) a flow control cassette having pre- formed flow path segments therein separated by valve stations for controlling communication between said flow path segments and pump stations for pumping fluid through said flow path segments, and
(b) a re-usable module adapted to cooperatively receive said fluid circuit module, said re-usable module including: means cooperatively associated with said chamber for effecting separation of said blood component from the remainder of said blood; means for cooperating with said valve stations to allow the flow of fluid through said pre- formed flow paths of said cassette; and a programmable controller for selectively controlling fluid flow through said valve stations and selectively establishing flow communication between said flow segments.
2. The system of Claim 1 wherein said means for treating comprises a solution for contacting said separated blood component .
3. The system of Claim 1 wherein said means for treating comprises a particle means for contacting said separated blood component .
4. The system of Claim 3 further comprising a sorption column through which said separated component is passed.
5. The system of Claim 2 wherein said solution comprises a solvent for extracting a selected compound from the separated component .
6. The system of Claim 2 wherein said solution comprises a compound for enzymatically treating said separated blood component .
7. The system of Claim 1 wherein said separation chamber is adapted for use with a centrifuge.
8. The system of Claim 1 wherein said separation chamber comprises a rotatable member and a membrane.
9. The system of Claim 1 comprising a first means for effecting separation means and a second means for effecting separation.
10. The system of Claim 9 wherein said first means is based on a first separation principle and said second means is based on a second separation principle.
11. The system of Claim 9 wherein said first and second means are based on the same separation principle.
12. The system of Claim 10 wherein said first means is based on the principle of centrifugal separation and said second means is based on affinity binding.
13. The system of Claim 12 wherein said second means comprises a sorption column adapted for separating selected compounds by affinity binding.
14. The system of Claim 12 wherein said second means comprises particle means adapted for flow within said fluid circuit .
15. The system of Claim 1 wherein said chamber comprises a first sub-chamber and a second sub-chamber.
16. The system of Claim 15 wherein said chamber is adapted for use with a centrifuge.
17. The system of Claim 15 further comprising means for introducing said treatment means into said second sub- chamber without introducing said treatment means into said first sub-chamber.
18. The system of Claim 15 wherein said treatment means comprises a solution.
19. The system of Claim 18 wherein said solution comprises a solvent for extracting a selected compound from said separated blood component .
20. The system of Claim 1 wherein said fluid circuit further comprises means for returning a separated blood component to said patient .
21. The system of Claim 20 wherein said fluid circuit comprises a single means for withdrawing blood from a donor or patient and returning said blood component to said donor or patient.
22. The system of Claim 21 wherein said single means comprises a needle adapted for insertion into a vein of said donor or patient, said needle being attached to tubing defining a flow path for said blood and blood component.
23. The system of Claim 1 further comprising means for collecting said treated blood component.
24. The system of Claim 1 wherein said re-usable module and said fluid circuit are contained in a portable, hand-carryable case.
PCT/US2002/005244 2001-03-06 2002-02-13 Multi-purpose, automated blood and fluid processing systems and methods WO2002070035A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
BR0204451-0A BR0204451A (en) 2001-03-06 2002-02-13 Automated multipurpose blood and fluid processing systems and methods
EP02717481.2A EP1365831B1 (en) 2001-03-06 2002-02-13 Multi-purpose, automated blood and fluid processing system
CA002407485A CA2407485C (en) 2001-03-06 2002-02-13 Multi-purpose, automated blood and fluid processing systems and methods
AU2002248479A AU2002248479B2 (en) 2001-03-06 2002-02-13 Multi-purpose, automated blood and fluid processing systems and methods
JP2002569206A JP2004519281A (en) 2001-03-06 2002-02-13 System and method for versatile automated blood and fluid processing

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/800,129 US6582386B2 (en) 2001-03-06 2001-03-06 Multi-purpose, automated blood and fluid processing systems and methods
US09/800,129 2001-03-06

Publications (2)

Publication Number Publication Date
WO2002070035A2 true WO2002070035A2 (en) 2002-09-12
WO2002070035A3 WO2002070035A3 (en) 2003-07-17

Family

ID=25177558

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/005244 WO2002070035A2 (en) 2001-03-06 2002-02-13 Multi-purpose, automated blood and fluid processing systems and methods

Country Status (8)

Country Link
US (1) US6582386B2 (en)
EP (1) EP1365831B1 (en)
JP (2) JP2004519281A (en)
CN (1) CN100377750C (en)
AU (1) AU2002248479B2 (en)
BR (1) BR0204451A (en)
CA (1) CA2407485C (en)
WO (1) WO2002070035A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017142003A1 (en) * 2016-02-16 2017-08-24 Terumo Kabushiki Kaisha Blood component sampling cassette, method for manufacturing blood component sampling cassette, blood sampling circuit set, and blood component sampling system
EP3195886A4 (en) * 2014-09-19 2018-04-11 Terumo Kabushiki Kaisha Blood component separation device
US11247216B2 (en) 2015-09-14 2022-02-15 Fenwal, Inc. Methods for sterilely connecting a container to a blood processing set
US11478755B2 (en) 2019-08-15 2022-10-25 Fenwal, Inc. Small volume processing systems and methods

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6890291B2 (en) 2001-06-25 2005-05-10 Mission Medical, Inc. Integrated automatic blood collection and processing unit
FR2850581B1 (en) * 2003-02-03 2005-09-09 Maco Pharma Sa PRECAUTIONED LOOP PICKUP SYSTEM
US7001513B2 (en) * 2003-08-06 2006-02-21 Nuvue Technologies, Inc. Automated processing of a biological component
KR101099962B1 (en) * 2003-11-07 2011-12-28 감브로 룬디아 아베 Fluid distribution module and extracorporeal blood circuit including such a module
DE602004004857T2 (en) 2003-11-07 2007-10-25 Gambro Lundia Ab END CAP ASSEMBLY WITH A HOSE FOR A PUMP FOR A FILTER AND FILTER WITH SUCH A FINAL CAP ASSEMBLY
ES2476021T3 (en) * 2003-11-24 2014-07-11 Gambro Lundia Ab Degassing device and end cap assembly for a filter
US7708710B2 (en) * 2003-12-23 2010-05-04 Fenwal, Inc. Method and apparatus for collecting and processing blood
US7354515B2 (en) 2004-02-23 2008-04-08 Millennium Medical Technologies, Inc. Fluid concentrator
US7479130B2 (en) * 2004-12-27 2009-01-20 University Hospitals Of Cleveland Case Western Reserve University Apparatus and method for autologous normovolemic hemodilution
US20060226086A1 (en) * 2005-04-08 2006-10-12 Robinson Thomas C Centrifuge for blood processing systems
JP2010535339A (en) * 2007-08-02 2010-11-18 ミリポア・コーポレイション System and apparatus for processing fluid samples
KR20100061704A (en) * 2007-08-31 2010-06-08 더 리젠츠 오브 더 유니버시티 오브 미시간 Selective cytopheresis devices and related methods thereof
US8685258B2 (en) 2008-02-27 2014-04-01 Fenwal, Inc. Systems and methods for conveying multiple blood components to a recipient
US8075468B2 (en) 2008-02-27 2011-12-13 Fenwal, Inc. Systems and methods for mid-processing calculation of blood composition
US20100158893A1 (en) * 2008-12-19 2010-06-24 Baxter International Inc. Systems and methods for obtaining immunoglobulin from blood
US11864553B2 (en) * 2009-10-23 2024-01-09 Fenwal, Inc. Methods and systems for providing red blood cell products with reduced plasma
US9034280B2 (en) * 2009-12-16 2015-05-19 General Electric Corporation High-throughput methods and systems for processing biological materials
FR2961711B1 (en) * 2010-06-23 2012-08-17 Millipore Corp POCKET FOR CIRCUIT OF A BIOLOGICAL LIQUID TREATMENT FACILITY
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
EP2627369B1 (en) 2010-10-15 2022-01-12 SeaStar Medical, Inc. Cytopheresic cartridge and use thereof
US9011684B2 (en) 2011-03-07 2015-04-21 Spinesmith Holdings, Llc Fluid concentrator with removable cartridge
PT2697768T (en) 2011-04-12 2020-05-18 Applied Science Inc Systems and methods for managing blood donations
EP2696961B1 (en) 2011-04-13 2019-02-13 Fenwal, Inc. Systems for use and control of an automated separator with adsorption columns
CA2860831A1 (en) 2012-01-09 2013-07-18 H. David Humes Cartridge and method for increasing myocardial function
US9399093B2 (en) 2012-01-27 2016-07-26 Fenwal, Inc. Systems and methods for performing online extracorporeal photopheresis
US9388383B2 (en) 2012-09-10 2016-07-12 Fenwal, Inc. Method for collecting red blood cells
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
DE102014103491A1 (en) * 2014-03-14 2015-09-17 Fresenius Medical Care Deutschland Gmbh Blood treatment cassette with foil valve and inelastic spacer as well as blood treatment device
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
AU2015266570B2 (en) 2014-05-30 2020-02-27 Applied Science, Inc. Systems and methods for managing blood donations
US10130752B2 (en) * 2014-08-21 2018-11-20 Fenwal, Inc. Parallel processing of fluid components
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
CN107735120B (en) * 2015-07-13 2021-06-15 美国血液技术公司 System and method for removing fat from salvaged blood
US10434240B2 (en) 2015-08-17 2019-10-08 Fenwal, Inc. Methods and systems for processing and washing a photopheresis mononuclear cell product
US10329530B2 (en) * 2016-01-18 2019-06-25 Fenwal, Inc. Cell washing system and methods for washing small volumes of cells
CN105854104B (en) * 2016-04-22 2020-03-13 四川南格尔生物科技有限公司 Annular separator applied to continuous plasma collection device
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
JP2019532696A (en) * 2016-09-16 2019-11-14 フェンウォール、インコーポレイテッド Blood separation system and method using centrifuge and rotating membrane separation techniques
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
US11191880B2 (en) * 2018-05-16 2021-12-07 Fenwal, Inc. Fill and finish systems and methods for small volume processing
EP3705146A3 (en) 2019-03-05 2020-11-25 Fenwal, Inc. Collection of mononuclear cells and peripheral blood stem cells
US11890399B2 (en) 2019-05-23 2024-02-06 Fenwal, Inc. Centrifugal separation and collection of red blood cells, plasma, or both red blood cells and plasma
EP3741460B1 (en) 2019-05-23 2023-09-27 Fenwal, Inc. Adjustment of target interface location between separated fluid components in a centrifuge
WO2023220209A1 (en) * 2022-05-10 2023-11-16 Fenwal, Inc. Unmanned helicopter for delivering blood product to a target location

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4895558A (en) 1985-07-15 1990-01-23 University Of Queensland Autologous plasma delipidation using a continuous flow system
US5194145A (en) 1984-03-21 1993-03-16 William F. McLaughlin Method and apparatus for separation of matter from suspension
US5865784A (en) 1995-06-07 1999-02-02 Alliance Pharmaceutical Corp. Method of hemodilution facilitated by monitoring oxygenation status
US5868696A (en) 1993-12-22 1999-02-09 Baxter International Inc. Peristaltic pump tube holder with pump tube shield and cover
US5958250A (en) 1995-06-07 1999-09-28 Baxter International Inc. Blood processing systems and methods which optically derive the volume of platelets contained in a plasma constituent
US5976388A (en) 1997-05-20 1999-11-02 Cobe Cardiovascular Operating Co., Inc. Method and apparatus for autologous blood salvage
US6175420B1 (en) 1998-05-20 2001-01-16 Zymequest, Inc. Optical sensors for cell processing systems
US6325775B1 (en) 1999-09-03 2001-12-04 Baxter International Inc. Self-contained, transportable blood processsing device

Family Cites Families (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4370983A (en) 1971-01-20 1983-02-01 Lichtenstein Eric Stefan Computer-control medical care system
NZ180199A (en) 1976-03-04 1978-11-13 New Zealand Dev Finance Method of testing for the presence of elevated plasma liprotein concentration
US4427777A (en) 1980-08-14 1984-01-24 New York Blood Center, Inc. Enzymatic conversion of red cells for transfusion
JPS5810056A (en) 1981-07-10 1983-01-20 株式会社クラレ Blood purifying apparatus
JPS5827559A (en) 1981-08-11 1983-02-18 株式会社クラレ Low density lipoprotein adsorbent
EP0180720B1 (en) 1981-09-10 1990-06-13 B. Braun-SSC AG Device for the selective extracorporeal precipitation of low-density lipoproteins for serum or plasma
US4481827A (en) 1981-12-15 1984-11-13 Baxter Travenol Laboratories, Inc. Blood fractionation apparatus having collection rate display system
JPS5826819A (en) 1982-03-17 1983-02-17 Kuraray Co Ltd Porous glass adsorbent for low-density lipoprotein particles
CA1221307A (en) 1982-12-02 1987-05-05 Nobutaka Tani Adsorbent and process for preparing the same
US4684521A (en) 1982-12-08 1987-08-04 Frederic A. Bourke, Jr. Method and system for externally treating the blood
US4479762A (en) 1982-12-28 1984-10-30 Baxter Travenol Laboratories, Inc. Prepackaged fluid processing module having pump and valve elements operable in response to applied pressures
DE3310727A1 (en) 1983-03-24 1984-10-04 B. Braun Melsungen Ag, 3508 Melsungen METHOD AND DEVICE FOR SELECTIVE, EXTRACORPORAL SEPARATION OF PATHOLOGICAL AND / OR TOXIC BLOOD COMPONENTS
US4683889A (en) 1983-03-29 1987-08-04 Frederic A. Bourke, Jr. Method and system for externally treating the blood
NZ208241A (en) 1984-05-22 1987-09-30 John Stephen Ayers Purification of blood plasma or serum: selective removal of (very) low density lipoproteins (ldl and vldl)
JPS60246765A (en) 1984-05-22 1985-12-06 旭化成工業株式会社 Adsorbing column for purifying body fluids
DE3422435A1 (en) 1984-06-16 1986-01-16 B. Braun Melsungen Ag, 3508 Melsungen METHOD AND DEVICE FOR SELECTIVELY SEPARATING PATHOLOGICAL AND / OR TOXIC SPECIES FROM BLOOD OR BLOOD PLASMA USING FILTER CANDLES
DE3422407A1 (en) 1984-06-16 1986-03-06 B. Braun Melsungen Ag, 3508 Melsungen USE OF HEPARINE DERIVATIVES FOR SELECTIVE EXTRA-CORPORAL PRECIPITATION OF LOW-DENSITY-LIPOPROTEINS FROM FULL SERUM OR PLASMA
DE3422494A1 (en) 1984-06-16 1985-12-19 B. Braun Melsungen Ag, 3508 Melsungen METHOD AND DEVICE FOR SPECIFIC ADSORPTION OF HEPARIN
ATE38846T1 (en) 1984-06-27 1988-12-15 Organon Teknika Bv BINDER FOR LOW DENSITY LIPOPROTEINS.
US4775482A (en) 1985-05-28 1988-10-04 Mederi Medical Systems, Inc. Method for removing fat and for purifying and defoaming liquids
FR2586359B1 (en) 1985-08-23 1989-09-08 Commissariat Energie Atomique SOLID SUPPORT BASED ON POLYVINYL ALCOHOL CAPABLE OF ADSORTING LIPOPROTEINS AND ITS USE FOR THE SEPARATION OF LOW DENSITY LIPOPROTEINS PRESENT IN A LIQUID SUCH AS BLOOD PLASMA
CA1259915A (en) 1985-10-09 1989-09-26 Sailen S. Mookerjea Means to reduce plasma cholesterol
US5354262A (en) 1986-02-18 1994-10-11 Boehringer Laboratories Apparatus for removal of insoluble fat from blood of a patient
US5203778A (en) 1986-02-18 1993-04-20 Boehringer Laboratories Process and apparatus for removal of insoluble fat from blood of a patient
US5133703A (en) 1986-02-18 1992-07-28 Boehringer Laboratories Process and apparatus for collecting blood of a patient for autotransfusion
EP0321597B1 (en) 1986-09-09 1992-11-11 Fraunhofer-Gesellschaft Zur Förderung Der Angewandten Forschung E.V. Selective adsorbent for binding low-density lipoproteins
US4842576A (en) * 1986-10-15 1989-06-27 Baxter International Inc. System for generating substantially constant fluid pressure
US5782792A (en) 1986-11-21 1998-07-21 Cypress Bioscience, Inc. Method for treatment of rheumatoid arthritis
US5104526A (en) 1987-01-30 1992-04-14 Baxter International Inc. Centrifugation system having an interface detection system
JP2543693B2 (en) 1987-03-20 1996-10-16 旭メデイカル株式会社 Adsorbent for low-density lipoprotein and method for producing the same
US5733254A (en) 1987-10-15 1998-03-31 Cypress Bioscience, Inc. Method for treating patients suffering from immune thrombocytopenic purpura
JPH01229878A (en) 1988-03-07 1989-09-13 Teijin Ltd Porous polyester fiber and body fluid cleaning unit
EP0427715A1 (en) 1988-06-20 1991-05-22 E.I. Du Pont De Nemours And Company Biocompatible, substance-specific reagents for treating physiological fluids
US5152743A (en) 1988-08-05 1992-10-06 Healthdyne, Inc. Apparatus and method for selective separation of blood cholesterol
JP2777604B2 (en) 1988-08-29 1998-07-23 旭メディカル株式会社 Adsorbent for body fluid treatment
JPH01315338A (en) 1989-05-01 1989-12-20 Asahi Chem Ind Co Ltd Adsorbent for lipoprotein having low specific gravity
DE3926539A1 (en) 1989-08-11 1991-02-14 Braun Melsungen Ag USE OF TENTACLE CATION EXCHANGERS FOR THE SELECTIVE ELIMINATION OF LOW DENSITY LIPOPROTEINS (LDL), FIBRINOGEN AND / OR UREA FROM LIQUIDS
JPH03114468A (en) 1989-09-29 1991-05-15 Terumo Corp Blood purifier and method of purifying thereof
JPH03289966A (en) 1990-04-06 1991-12-19 Nissho Corp Method for removing low density and very low lipoprotein in blood
US5464634A (en) 1990-06-22 1995-11-07 The Regents Of The University Of California Red blood cell surrogate
US5641622A (en) 1990-09-13 1997-06-24 Baxter International Inc. Continuous centrifugation process for the separation of biological components from heterogeneous cell populations
US5258149A (en) 1990-11-27 1993-11-02 W. R. Grace & Co.-Conn. Process of making a membrane for high efficiency removal of low density lipoprotein-cholesterol from whole blood
US5308320A (en) * 1990-12-28 1994-05-03 University Of Pittsburgh Of The Commonwealth System Of Higher Education Portable and modular cardiopulmonary bypass apparatus and associated aortic balloon catheter and associated method
JPH0557015A (en) 1991-09-04 1993-03-09 Asahi Medical Co Ltd Adsorbent and sterilizing method therefor
US6007725A (en) 1991-12-23 1999-12-28 Baxter International Inc. Systems and methods for on line collection of cellular blood components that assure donor comfort
US5298016A (en) 1992-03-02 1994-03-29 Advanced Haemotechnologies Apparatus for separating plasma and other wastes from blood
US5437624A (en) 1993-08-23 1995-08-01 Cobe Laboratories, Inc. Single needle recirculation system for harvesting blood components
JPH07509153A (en) 1992-07-13 1995-10-12 ポール・コーポレーション Automated systems and methods for processing biological fluids
DE4331358A1 (en) 1992-10-12 1994-04-14 Braun Melsungen Ag Process for the quantitative selective removal or preparation of tumor necrosis factor (TNF) and / or lipopolysaccharides (LPS) from aqueous liquids
DE4239442C2 (en) 1992-11-24 2001-09-13 Sebo Gmbh Use of an adsorbent material modified with polynuclear metal oxide hydroxides for the selective elimination of inorganic phosphate from protein-containing liquids
JPH06233815A (en) 1993-02-10 1994-08-23 Toyobo Co Ltd Hemocatharsis adsorbent
JPH06233816A (en) 1993-02-12 1994-08-23 Toyobo Co Ltd Hemocatharsis adsorbent
JPH06237995A (en) 1993-02-15 1994-08-30 Toyobo Co Ltd Blood purifying/adsorbing material
JPH06237996A (en) 1993-02-17 1994-08-30 Toyobo Co Ltd Blood purifying/adsorbing material
US5391143A (en) 1993-03-12 1995-02-21 Kensey Nash Corporation Method and system for effecting weight reduction of living beings
US5945272A (en) 1993-06-04 1999-08-31 Biotime, Incorporated Plasma expanders and blood substitutes
US5753227A (en) 1993-07-23 1998-05-19 Strahilevitz; Meir Extracorporeal affinity adsorption methods for the treatment of atherosclerosis, cancer, degenerative and autoimmune diseases
EP0710126B1 (en) 1993-07-30 2004-05-06 Aruba International Pty. Ltd. A plasma delipidation system
EP0804734B1 (en) 1994-05-13 2005-05-04 Miltenyi Biotec GmbH Sterile and pyrogen-free columns coupled to protein for binding and removal of substances from blood
RU2079349C1 (en) 1994-06-17 1997-05-20 Малое государственное предприятие "ВИАМ-41" Filtering member for microfiltration and ultrafiltration and method of its production
DE4435612A1 (en) 1994-10-05 1996-04-11 Braun Melsungen Ag Process for the simultaneous removal of tumor necrosis factor alpha and bacterial lipopolysaccharides from an aqueous liquid
US5733253A (en) 1994-10-13 1998-03-31 Transfusion Technologies Corporation Fluid separation system
US5581687A (en) 1994-11-10 1996-12-03 Baxter International Inc. Interactive control systems for medical processing devices
DE19515554C2 (en) 1995-04-27 1999-06-17 Braun Melsungen Ag Use of an agent and device for the simultaneous extracorporeal elimination of tumor necrosis factor alpha and bacterial lipopolysaccharides from whole blood and / or blood plasma
US5738644A (en) 1995-06-07 1998-04-14 Cobe Laboratories, Inc. Extracorporeal blood processing methods and apparatus
US5720716A (en) * 1995-06-07 1998-02-24 Cobe Laboratories, Inc. Extracorporeal blood processing methods and apparatus
US5671135A (en) 1995-06-07 1997-09-23 Zymequest, Inc. Programmable controller module
US5911698A (en) 1995-12-22 1999-06-15 Aruba International Pty. Ltd. Treatment for cardiovascular and related diseases
WO1997027889A1 (en) 1996-01-31 1997-08-07 Kaneka Corporation Adsorbent for disease-related factors in body fluids, method of elimination by adsorption, body fluid purifier, and apparatus for purifying body fluids
US5637082A (en) * 1996-02-22 1997-06-10 Haemonetics Corporation Adaptive apheresis apparatus
US5865785A (en) 1996-02-23 1999-02-02 Baxter International Inc. Systems and methods for on line finishing of cellular blood products like platelets harvested for therapeutic purposes
US5858238A (en) 1996-03-08 1999-01-12 Baxter Research Medical, Inc. Salvage of autologous blood via selective membrane/sorption technologies
EP0796865B1 (en) 1996-03-18 2003-06-04 Kaneka Corporation Adsorbent and method for removing chemokines of the CC subtype from body fluids
EP0842694A4 (en) 1996-03-21 2000-01-05 Kaneka Corp Hollow yarn membrane used for blood purification and blood purifier
JPH1076004A (en) 1996-09-05 1998-03-24 Kanegafuchi Chem Ind Co Ltd Adsorbing material for humor treatment and adsorber for humor treatment
JPH10182694A (en) 1996-12-20 1998-07-07 Asahi Chem Ind Co Ltd Compound for binding to lipoprotein having low specific gravity
US5879624A (en) 1997-01-15 1999-03-09 Boehringer Laboratories, Inc. Method and apparatus for collecting and processing blood
US5980760A (en) 1997-07-01 1999-11-09 Baxter International Inc. System and methods for harvesting mononuclear cells by recirculation of packed red blood cells
JPH1160595A (en) 1997-08-22 1999-03-02 Asahi Chem Ind Co Ltd Peptide for selective adsorption of low density lipoprotein
DE19802321C2 (en) 1998-01-23 2000-05-11 Fresenius Ag Method and device for the preparation of intra- or post-operative blood loss for autotransfusion
JPH11332981A (en) 1998-05-25 1999-12-07 Tosoh Corp Method for removing low-density lipoprotein in blood
US6306346B1 (en) 1999-02-10 2001-10-23 Terumo Cardiovascular Systems Corporation Self-contained pack assembly for an extracorporeal blood circuit
US6254567B1 (en) 1999-02-26 2001-07-03 Nxstage Medical, Inc. Flow-through peritoneal dialysis systems and methods with on-line dialysis solution regeneration
US6284142B1 (en) * 1999-09-03 2001-09-04 Baxter International Inc. Sensing systems and methods for differentiating between different cellular blood species during extracorporeal blood separation or processing
US6315707B1 (en) * 1999-09-03 2001-11-13 Baxter International Inc. Systems and methods for seperating blood in a rotating field
US6296450B1 (en) * 1999-09-03 2001-10-02 Baxter International Inc. Systems and methods for control of pumps employing gravimetric sensing
US6294094B1 (en) * 1999-09-03 2001-09-25 Baxter International Inc. Systems and methods for sensing red blood cell hematocrit
US6270673B1 (en) * 1999-09-03 2001-08-07 Baxter International Inc. Door latching assembly for holding a fluid pressure actuated cassette during use
US6322488B1 (en) * 1999-09-03 2001-11-27 Baxter International Inc. Blood separation chamber with preformed blood flow passages and centralized connection to external tubing
US6261065B1 (en) * 1999-09-03 2001-07-17 Baxter International Inc. System and methods for control of pumps employing electrical field sensing

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5194145A (en) 1984-03-21 1993-03-16 William F. McLaughlin Method and apparatus for separation of matter from suspension
US4895558A (en) 1985-07-15 1990-01-23 University Of Queensland Autologous plasma delipidation using a continuous flow system
US5868696A (en) 1993-12-22 1999-02-09 Baxter International Inc. Peristaltic pump tube holder with pump tube shield and cover
US5865784A (en) 1995-06-07 1999-02-02 Alliance Pharmaceutical Corp. Method of hemodilution facilitated by monitoring oxygenation status
US5958250A (en) 1995-06-07 1999-09-28 Baxter International Inc. Blood processing systems and methods which optically derive the volume of platelets contained in a plasma constituent
US5976388A (en) 1997-05-20 1999-11-02 Cobe Cardiovascular Operating Co., Inc. Method and apparatus for autologous blood salvage
US6175420B1 (en) 1998-05-20 2001-01-16 Zymequest, Inc. Optical sensors for cell processing systems
US6325775B1 (en) 1999-09-03 2001-12-04 Baxter International Inc. Self-contained, transportable blood processsing device

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1365831A4

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3195886A4 (en) * 2014-09-19 2018-04-11 Terumo Kabushiki Kaisha Blood component separation device
US11247216B2 (en) 2015-09-14 2022-02-15 Fenwal, Inc. Methods for sterilely connecting a container to a blood processing set
WO2017142003A1 (en) * 2016-02-16 2017-08-24 Terumo Kabushiki Kaisha Blood component sampling cassette, method for manufacturing blood component sampling cassette, blood sampling circuit set, and blood component sampling system
US11617524B2 (en) 2016-02-16 2023-04-04 Terumo Kabushiki Kaisha Blood component sampling cassette, method for manufacturing blood component sampling cassette, blood component sampling circuit set, and blood component sampling system
US11478755B2 (en) 2019-08-15 2022-10-25 Fenwal, Inc. Small volume processing systems and methods

Also Published As

Publication number Publication date
EP1365831A4 (en) 2010-03-31
CN100377750C (en) 2008-04-02
EP1365831A2 (en) 2003-12-03
BR0204451A (en) 2003-03-25
CN1561243A (en) 2005-01-05
WO2002070035A3 (en) 2003-07-17
US6582386B2 (en) 2003-06-24
AU2002248479B2 (en) 2006-09-21
EP1365831B1 (en) 2013-04-10
CA2407485A1 (en) 2002-09-12
JP2004519281A (en) 2004-07-02
US20020128583A1 (en) 2002-09-12
JP2007301388A (en) 2007-11-22
CA2407485C (en) 2009-10-27

Similar Documents

Publication Publication Date Title
CA2407485C (en) Multi-purpose, automated blood and fluid processing systems and methods
CA2407961C (en) Automated system and method for withdrawing compounds from blood
AU2002248477B2 (en) Automated system adaptable for use with different fluid circuits
AU2002248478B2 (en) Automated system and method for pre-surgical blood donation and fluid replacement
AU2002248479A1 (en) Multi-purpose, automated blood and fluid processing systems and methods
AU2002244108A1 (en) Automated system and method for withdrawing compounds from blood
AU2002248477A1 (en) Automated system adaptable for use with different fluid circuits
AU2002248478A1 (en) Automated system and method for pre-surgical blood donation and fluid replacement
JP5763099B2 (en) Device for the collection of filtered blood components, especially red blood cells
US20220305187A1 (en) System And Method For Plasma Purification Prior To Mononuclear Cell Collection
JP5215412B2 (en) Apparatus for the controlled addition of solvents to blood components

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU BR CA CN IN JP

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR

WWE Wipo information: entry into national phase

Ref document number: IN/PCT/2002/01425/MU

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2002717481

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2002248479

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2407485

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2002 569206

Country of ref document: JP

Kind code of ref document: A

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 028005562

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2002717481

Country of ref document: EP