CA2398614A1 - System for organ and tissue preservation and hypothermic blood substitution - Google Patents

System for organ and tissue preservation and hypothermic blood substitution Download PDF

Info

Publication number
CA2398614A1
CA2398614A1 CA002398614A CA2398614A CA2398614A1 CA 2398614 A1 CA2398614 A1 CA 2398614A1 CA 002398614 A CA002398614 A CA 002398614A CA 2398614 A CA2398614 A CA 2398614A CA 2398614 A1 CA2398614 A1 CA 2398614A1
Authority
CA
Canada
Prior art keywords
solution
organ
tissue
preservation
additive
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002398614A
Other languages
French (fr)
Inventor
Michael J. Taylor
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Organ Recovery Systems Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2398614A1 publication Critical patent/CA2398614A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • A01N1/0221Freeze-process protecting agents, i.e. substances protecting cells from effects of the physical process, e.g. cryoprotectants, osmolarity regulators like oncotic agents

Abstract

A unified solution system for organ and/or tissue preservation and/or hypothermic blood substitution can be used to prepare multiple solutions for use in various stages of organ procurement, preservation and transplantation and bloodless surgery procedures. The base electrolyte solution may comprise an (hyperkalemic) intracellular or extracellular base solution to which various additives are added depending on the specific stages of the preservation or transplantation procedure. Base and additives can be stored in separate containers in a single package or kit.

Description

SYSTEM FOR ORGAN AND TISSUE PRESERVATION AND HYPOTHERMIC
BLOOD SUBSTITUTION
BACKGROUND OF THE INVENTION
1. Field of Invention This invention relates to organ preservation and hypothermic blood substitution. This invention particularly relates to compositions, processes and systems for organ and tissue preservation and/or hypothermic blood substitution.
2. Description of Related Art Hypothermia is the bed rock of all useful methods of organ and tissue preservation, and has proven to be most effectively applied by controlling the extracellular environment of cells directly, and the intracellular environment indirectly, during cold exposure. Control of the extracellular environment of cells to optimise preservation is based upon different strategies that include either static cold storage (or flush preservation), or low temperature continuous perfusion.
These different strategies call for different approaches to interventional control of the extracellular environment in order to optimize preservation, and hence different design elements for the solutions used to effect these strategies.
In principle, cold flush storage or preservation is based upon the premise that temperature reduction to near but not below the ice point (0°C) precludes the need to support metabolism to any significant extent, and that the correct distribution of water and ions between the intracellular and extracellular compartments can be maintained by physical rather than metabolic means. During the period that metabolic pumps are inactivated, the driving force for transmembrane ion flux is the difference in ionic balance between intracellular and extracellular fluid. The driving force for water uptake (cell swelling) is the impermeant intracellular anions. Thus changes can be prevented or restricted by manipulating the extracellular environment to abolish chemical potential gradients. On this basis, a variety of flush, or organ washout, r-= ~ ' ~ , j.rin ivo ma ,..

Z
solutions have been devised and evaluated for cold storage. These solutions are often referred to as "intracellular" solutions due to their resemblance, in some respects, to intracellular fluid.
'fhe principle design elements of the "intracellular" flush solutions has been to adjust the ionic balance (notably of the monovaLent cations) and to raise the osmnlality by including an impermeant solute to balance the intcace11u1ar osmotic prESSUre responsible for water uptake. ~Iowever, the most important factor of the efficacy of cold flush solutions may be the prevention of cellular edema by inelnsion of imperrneant solutes since it has been established that ionic imbalances, especially potassium depletion, are readily and rapidly reversible.
Prior to 1988, the standard solution for clinical preservation of abdominal organs, principally the kidney, was Collies solution, which consists predominantly of potassium phosphate, magnesium sulfate and glucose. Tn recent years, however, this has been superseded either by a modified version called "EuroCollins" in which the magnesium sulfate is omitted, or more extensively by the University of Wisconsin solution (LlW solution) in which much of the phosphate anion hsa been replacod with lactobivnate, and ~in'which glucose has been replaced with ramose. These larger molecules provide better protection against adverse effects of cell swelling during hypothermic storage. The choice of solutions for heart preservation has been strongly influenced by the previous experience of cardiac surgeons with eardioplegic solutions in open.-heart surgery. In this case the primary objective has been to produce rapid cessation of the heartbeat, and solutions were designed more with this in mind than with protection of the cells during preservation in mind. In particular, early studies suggested that the very high potassium levels (>I OOmlvn found in organ preservation ZS solutions might be harmful to the heart. In fact, the solution most often used was St. Thomas's (Plcgisol) with a potassium contest of only 16 mM.
The choice of solution for cardioplegia and myocardial preservation remains controversial and widely varied. While UW solution has emerged as the industry standard for kidney, liver and pancreas, no such standard has been adopted for heart preservation. Moreover, the development of the variety of preservation solutions for organ storage has emphasized the zteed for careful optimization in relation to the specific characteristics of the tissues to be preserved.
Attention to biophysical properties of "intracellular" flush solutions, to restrict passive diffusional processes, has unquestionably led to the development of SUBSTITUTE SHEET
Emvfangszeit 28. Jan. 23:22 techniques that have provided the basis of clinical organ preservation during the past 30 years. Nevertheless, it is recognized that further optimization of cold flush solutions can be achieved by inclusion of biochemical and pharmacological components that will be effective in counteracting the deleterious effects of ischemia and repexfusion injury. To a limited extent this approach has been incorporated in the design of the University of Wisconsin organ preservation solution (LTW
solution marketed as "ViaspanTM"; DuPont) which has become the most widely used solution for cold flush preservation of kidneys, livers and pancreases. With due consideration for the effects of ischemia, hypoxia, hypothermia and reperfusion injury on cells, coupled with the proven efficacy of various existing organ preservation solutions, a general consensus of the most important characteristics in the design of hypothermic storage solutions has emerged. These include: minimizing of hypothermically induced cell swelling; preventing expansion of the interstitial space (especially important during perfusion); restricting ionic imbalances; preventing intracellular acidosis; preventing injury from free radicals; and providing substrates for regeneration of high energy phosphate compounds during reperfusion.
In continuous hypothermic perfusion preservation, the desirable properties of hypothermic solutions listed above are also applicable to controlling the extracellular environment by way of continuous perfusion techniques. In contrast to static cold storage, continuous perfusion is usually controlled at around 10°C and is based upon a different principle: it is generally assumed that a moderate degree of cooling will reduce metabolic needs but that continuous perfusion is required to support the suppressed metabolism and remove catabolic products. Because it is assumed that sufficient metabolic activity remains to actively regulate a near-normal cell volume and ionic gradients, the perfusates are generally acellular, isotonic, well oxygenated solutions having a composition that more closely resembles plasma than intracellular fluid. Such perfusates are therefore designated as "extracellular" solutions, and are perfused through the vascular bed of an organ at a pressure sufficient to achieve uniform tissue distribution (typically 40-60mm Hg). To balance this applied hydrostatic pressure and prevent interstitial edema, oncotic agents such as albumin or synthetic macromolecular colloids are incorporated into the perfusates.
Substrate support of the remaining metabolism at ~10°C is also an important consideration and it has been shown in several organs that high energy adenine nucleotides can be synthesized during hypothermic perfusion preservation.
In addition to the principal objective of supporting metabolism, continuous perfusion also provides other advantages over flush preservation. These include the wash out of accumulated lactate and protons, thereby removing the metabolic block on glycolysis; this is thought to be especially beneficial for organs that have suffered prior warm ischemia. Perfusion also facilitates the removal of erythrocytes from the microcirculation and helps to maintain vascular patency during prolonged storagee' Continuous perfusion has been shown to provide the best means of achieving prolonged hypothermic preservation (e.g., 3-7 days for kidneys), but concerns for damage to the vascular endothelium during prolonged perfusion may be a limiting factor.
Although it has been experimentally verified that cell metabolism continues at temperatures as low as 10°C, and that adenine nucleotides can be resynthesized during hypothermic preservation if appropriate substrates are provided, it is considered unlikely that this level of metabolism can prevent transmembrane ion and water movements: this is due principally to the temperature sensitivity of the active pumps.
Hence, some advocates of continuous perfusion have modified the perfusate accordingly by increasing both the K+ concentration and the osmolality.
Similarly, modification of cold flush solutions can be considered to circumvent some of the identified limitations of that approach. For example, the lack of support of metabolism during ice-storage can be addressed by raising the temperature of storage, by incorporating biochemical substrates and raising the oxygen tension to promote adenine nucleotide repletion. Also, the use of pharmacological agents, such as inhibitors of 5'-nucleotidase (e.g., allopurinol) has been advocated as a means of averting adenine nucleotide depletion.
With respect to specific cell requirements as a function of temperature, it is not necessary to consider incorporating specific oxygen-carrying molecules at temperatures below ~ 10°C since at such low temperatures it is well established that metabolic activity is sufficiently depressed that the 02 demand can be satisfied by dissolved 02 in the aqueous solution without the need for hemoglobin or synthetic OZ-carrying molecules.
Optimum control of the intracellular and extracellular environment of cells during hypothermia depends upon the interaction of a variety of factors that include temperature, oxygen tension, acidity, osmotic pressure and chemical composition of the perfusion fluid or wash-out solution.
It is. now recognized that the successive phases of the transplantation procedure involving organ procurement, storage, transportation, reimplantation and 5 reperfusion may impose different requirements for optimum preservation at the different stages. This is illustrated by evidence that heart preservation with the "intracellular" solution, EuroCollins, was enhanced when the heart was initially arrested and subsequently flushed prior to reperfusion, with an "extracellular"
cardioplegic solution. Therefore, any single formulation of preservation solution is unlikely to provide optimum protection during all the processing stages of a transplantation procedure, or the interventional stages of complex surgeries.
Interest in general or universal tissue preservation techniques is exemplified by the need for methods of protecting multiple vital organs, and even the whole body, for applications in modern day surgery. Multiple organ harvesting for transplantation can be optimized by hypothermic perfusion of the whole cadaver, or donor organ blocks comprising several organs, to minimize warm ischemic injury. The ultimate challenge is perhaps protection of the entire body against the effects of global ischemia during periods of circulatory and/or cardiac arrest for "bloodless" surgery.
Surgeons have developed skills that allow very complex, corrective and life-saving operations to be performed, notably on the heart and brain. Many of these complicated time-consuming procedures have the inherent need for temporary cessation of blood flow and demand protection of the patient against the deleterious effects of ischemia and anoxia. Although hypothermia is routinely used as an adjunctive protective modality for surgical procedures that require a period of cardiac arrest, there are restrictive time constraints (<1 hour at temperatures usually not lower than 18°C) upon the safe interval of cold ischemia if neurological sequelae are to be avoided. It is well recognized that the window of opportunity for safe surgical intervention could be extended by using greater degrees of hypothermic metabolic suppression, but this becomes unacceptably dangerous due principally to the effects of profound hypothermia on the blood, leading to coagulopathies and irreversible microvascular blockage.
U.S. Patents Nos. 5643712, 5699793, 5843024 to Brasile and Nos. 5,599,659, 5,702,881 to Brasile et al., each of which is incorporated herein by reference in its entirety, describe separate resuscitation and preservation solutions for tissues and y o . ~ ~~~~ :,° a ~ ~ a ~ ,_ ,. - f : -~.~501 U2~3 k ' ~ ,~a~~.ra~x~.~~m~.
.°v orgaas. The Brasile patents disclose methods in which some embodiments of the composition of this invention can be applied. Also, the Brasile patents disclose compositions that may be used in methods and kits of this~invention.
U.S. Patent No. 5,130,230 to Segall et al. discloses a method for performing a bloodless hypothermic procedure that uses a plurality of solutions that are systematically administered tv the subject or patient. International Application No. WO 86/00812 discloses a drug for use in preventing and treating ischaemic cell damage that includes at least one plasma volume expander, at least one hydroxyl radical scavenger, at least one magnesium salt and at (cast one calcium blocking organic compound; and a kit that includes all of the components of the drug, individually or in any combination. U.S. Patent No. 5,552,267 to Stern et al.
discloses a method for prcscrving an organ that uses a sohrtion that specifically avoids the use of sodium ions, chloride ions and calcium ions.
The present inventor has explored experimental approaches employing a technique of asanguineous~blood substitution using accllular synthetic solutions designed to protect the heart, brain and visceral organs during several houra of bloodless perfusion. The concept of using ultraprofouad hypothermia (<10°C) and complete blood replacement is appealing for several reasons and is based upon a variety of factors. First, deeper hypothermia can provide more effective suppression of metabolism, thereby extending the tolerance to isehemia and minimizing the demand for oxygen to levels that can be adequately supplied in a cold aqueous solution without the need of special oxygen-carrying molecules. Second, complete exsanguinagon ameliorates a complication associated with increased viscosity, coagulopathies, arid erythrocyte clumping of cooled blood. Third, vascular purging can remove harmful catabolic products and formed elements that might participate in the ischemia and reperfusion injury cascades. A fourth advantage is that total exsanguination provides the opportunity to Control the vascular and cxtracellular compartments directly with fluids designed to be protective under the conditions of ultraprofound hypothermia. For example, solutes can be added to maintain ionic and osmotic balance at the cellular and~tissue levels; biochemical and pharmacological additives can help sustain tissue integrity is a variety of ways including efficient vascular flushing, membrane stabilization, free-za~dical scavenging and providing substrates for the regeneration of high-energy compounds during rewarming and reperfusion. In essence, these are the principles that are embodied, to a greater or SUHSTITtTTE SHEET
Fmnfangszeit 28. Jan. 23:22 ~FAg 703 ~ 2787 r ~f~ ~~ ~iD~~. ~ > ~ ~.
x w lesser extent, in the design of various solutions used fvr ear vlvo organ preservation. ~
this invention, similax principles have been adopted in the design of new hypothermic blood substitutes.
The working hypothesis that was used to evaluate this approach has been that an acellular solution can be designed to act as a universal tissue preservation solution during several hours of hypothermic wholo-body washout involving cardiac arrest, with or without circulatory arrest. Under this hypothesis, 'Taylor et al. have formulated and evaluated two solutions designated Hypothernnosol'~''I-purga (HTS-P) and Hypothermosol ~-maintenance (ITS-IV)7 that fulfill separate requirements during the -asanguineous procedure. Sonic aspects of these solutions are described in U.S.
SU$STITLJTE SI~EEE'T

,.. [Mnf,evnanil 9A ion Patents Nos. 5,405,742 and 5,514,536 to Taylor, both of which are incorporated herein by reference in their entireties. The Taylor patents disclose methods in which some embodiments of the compositions of this invention can be applied. Also, the Taylor patents disclose compositions that may be used in methods and kits of this invention.
The principal solution (HTS-M) is a hyperkalemic "intracellular" solution specifically designed to "maintain" cellular integrity during the hypothermic interval at the lowest temperature. The second solution is designed to interface between the blood and the HTS-M maintenance solution during both cooling and warming. This companion solution is, therefore, an "extracellular" flush solution designed to aid in purging the circulation of blood during cooling since the removal of erythrocytes from the microvasculature is an important objective during ultraprofound hypothermia.
The "purge" solution is also designed to flush the system (vasculature and CPB
circuit) of the hyperkalemic HTS-M solution during warming and possibly help to flush-out accumulated toxins and metabolic byproducts that might promote oxidative stress and free radical injury upon reperfusion.
Based upon the principles that have emerged from isolated organ preservation studies, an attempt was made to incorporate some of the important characteristics in the formulation of the HypothermosolTM solutions and, wherever possible, components that might fulfill multiple roles were selected. This strategy maximizes the intrinsic qualities of the solutions that, by design as universal tissue preservation solutions, would inevitably be a hybrid of other hypothermic perfusates and storage media.
The composition of the HypothermosolTM blood substitutes and the rationale for their formulation are discussed in U.S. Patents Nos. 5,405,742 and 5,514,536 to Taylor, both of which are incorporated herein by reference in their entireties. These solutions have been shown to protect the brain, heart and visceral organs during 3.5 hours of cardiac arrest and global ischemia in an asanguineous canine model during controlled profound hypothermia at <10°C. Successful application of this technique to man would provide more than a 3-fold extension of the current limits of <1 hour for "safe" arrest without a high risk of neurological complications. This novel approach to bloodless surgery would significantly broaden the window of opportunity for surgical intervention in a variety of currently inoperable cases, principally in the areas of cardiovascular surgery, neurosurgery and emergency trauma surgery.
More recently, the HypothermosolTM-maintenance solution has been used for ih vitro hypothermic preservation of a variety of tissues and organs including isolated hearts, fetal spinal cord and engineered skin.
SUMMARY OF THE INVENTION
The present invention is based upon the concept of a unified solution system for preparing multiple solutions designed and optimized for the various stages of organ or tissue procurement-preservation-transplantation and/or bloodless surgery procedures.
While the known systems, including those taught in the Brasile and Taylor patents discussed above, have required completely different compositions for different stages of organ procurement, preservation and transplantation procedures, the present invention provides one or two (or optionally more) base compositions and a number of different additives that can be added to one or more of the base compositions to produce specific compositions useful for specific stages in organ or tissue procurement, preservation and transplantation and/or bloodless surgery procedures. In embodiments, the base and additives can be stored in separate containers in a single package or kit. Specific improved base compositions are also provided.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the viability index for MDCK cells after a storage interval;
Figure 2 shows the comparative viability of A10 cells after a one-day storage interval;
Figure 3 shows the comparative viability of CPAE cells after a one-day storage interval;
Figure 4 shows the comparative viability of A10 cells after a 3-day storage interval;
Figure 5 shows the comparative viability of CPAE cells after a 3-day storage interval;
Figure 6 shows the comparative viability of A10 cells during a 6 day period after a hypothermic storage interval of 3 days;
Figure 7 shows the comparative viability of CPAE cells during a 6 day period after a hypothermic storage interval of 3 days;
Figure 8 shows light microscopy histology of jugular vein segments after a period of cold ischemia in DMEM culture medium;
Figure 9 shows light microscopy histology of jugular vein segments after a period of cold ischemia in "I-Base-HK" preservation solution in accordance with this invention;
Figure 10 shows internal renal resistance measurements for canine kidneys perfused at ~9°C;
Figure 11 shows arterial flow rates for canine kidneys perfused at 9°C;
Figure 12 shows internal renal resistance measured in vivo for preserved kidneys transplanted ectopically after 20 hours machine perfusion preservation;
Figure 13 shows mean arterial flow measured ih vivo for preserved kidneys transplanted ectopically after 20 hours machine perfusion preservation; and Figure 14 shows biochemical indicators (creatinine and BUN) of kidney function for pig kidneys transplanted after a minimum of 20 hour cold storage preservation.
Figures 15 and 16 show comparative viability of cells cryopreserved with DMSO prepared in either the new phosphate-free, high potassium intracellular base vehicle solution (HK-CV), or EuroCollins solution. Data represents the mean (~SEM) of 4 replicate batches of cells.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
This invention provides a unified solution system for preparing multiple solutions designed and optimized for various stages of organ and/or tissue procurement, preservation and transplantation andlor bloodless surgery procedures.
In preferred embodiments, the present invention provides one or two base compositions and a number of different additives that can be added to the base compositions) to produce specific compositions useful for specific stages in the organ procurement, preservation and transplantation and/or bloodless surgery procedures. In embodiments, the base and additives can be stored in separate containers in a single package or kit.
A base formulation includes a design which takes into account the biophysical and minimal biochemical components that can be standardized for all or a desired subset of applications. This base unified solution can then be used as a vehicle for a range of additive "cocktails" to derive a system of solutions optimized for different needs.

In embodiments of the unified solution system series of solutions, solutions for warm ischemic time-organ preservation can include, for example, a hypothermic flush/purging solution, a hypothermic perfusate/maintenance solution, a "Normothermic" perfusate/rescue solution and/or a pre-reimplantation flush/rinse 5 solution.
Exemplary embodiments of the unified solution system series can include, for example, the following base solutions:
Unified solution system Intracellular Base Solution: minimum requirements for cold storage including cryopreservation solutions. An exemplary formulation of 10 such a solution as a preferred embodiment is given in Table 2.
Unified solution system Extracellular Base Solution: plasma-like electrolytes as base for oxygen carrying molecules and other substrates necessary for optimized "normothermic" perfusion.
Exemplary embodiments of the unified solution system series can include, for example, the following base plus additive solutions:
Purge = Extracellular Base plus purge additive ("cocktail") designed principally to purge the vasculature of blood in preparation for preservation.
Maintenance = Intracellular Base plus cytoprotection additive ("cocktail") designed to protect and maintain cellular stability during cold storage.
Ideally, this will apply to both static cold storage and cold machine perfusion.
Rescue = Extracellular Base plus rescue additive ("cocktail") for near nornothermic perfusion.
Rinse = Extracellular Base plus rinse additive ("cocktail") designed to flush out unwanted preservation molecules prior to reimplantation. This may fulfill a different role from the Purge solution designed to remove erythrocytes and other blood components prior to the preservation phase.
Cryo = Concentrated Intracellular Base plus permeating or non-permeating cryoprotective additives for sub-zero preservation of cells and tissues. For the Cryo embodiment, the intracellular base is preferably concentrated to a 3X to 4X
strength in comparison to its use alone and with most other additives. This facilitates its combination with additive cryoprotective compounds.
Examples of some illustration additives that can be utilized in accordance with this invention are listed in Table 3, although many other additives can be used.
Exemplary solutions for a clinical organ preservation program are summarized in Table 1.
Table 1. Solution Design Strategy for Clinical Organ Preservation Program A lication Tem erature Base Solution Additives Phase Ran a T a Organ Preparation10-37C Extracellular Purging cocktail in situlex viva ur a Organ Maintenance~-4C Intracellular Protection/Maintenance cold flush (Hi K cocktail Organ Maintenance$-1$C Intracellular Protection/Maintenance cold machine (High K or cocktail perfusion or Na) cardiopulmonary b ass Organ Rescue 30-37C Extracellular Rescue cocktail warm machine erfusion Organ 30-37C Extracellular Plasma-like Pre-reimplantation ex viva rinse Table 2. Formulation of unified solution system Intracellular Base (High Potassium) Na:'-62.5 C1- 30.1 HEPES 35 mM Dextran-40 6%
mM mM

K+ 70 mM H2P04 2.5 Lactobionate30 mM Glucose S mM
mM

CaZ+O.OS HC03- 5 mM Gluconate 70 mM Adenosine 2 mM
mM

Mg' 1 S mM Sucrose 15 mM reduced 3 mM
Glutathione Mannitol 25 mM
Table 3. Exemplary Biochemical and Pharmacological Additives for Preservation Media Classification Examples Anti-platelet aggregationlvasoactiveProstacyclin, Prostaglandin agents E-1 (PGE1), Mgz+

Calmodulin inhibitors Chlorpromazine (CPZ), trifluoperazine Calcium Channel Blockers Nicardipine, nifedipine, verapamil, CPZ

Protease and phospholipase CPZ, verapamil, calpain antagonists inhibitors Anti-oxidants/free radical Glutathione, catalase, super scavengers oxide dismutase (SOD), allopurinol, dimethylthiourea, vitamin-E (or Trolox), magnesium ascorbyl phosphate, Lazaroids Anti-apoptotic agents cycloheximide Iron chelators Desferroxamine Membrane Stabilizers CPZ, Dexamethosone, trehalose "Cytoprotective" agents PGE1, glycine Metabolic Substrates:

Sugars glucose, fructose, ribose Nucleotide precursors (HEP Adenine, Adenosine, Fructose enhancers) diphosophate, Glyceraldehyde-3-phosphate Oxygen-carriers Perfluorocarbons, PEG-hemoglobin Trophic Factors Growth factors, nucleic acid derivatives, ribonucleotides, glycosaminoglycans Cryoprotective Additives Dimethylsulfoxide (DMSO), (CPA) glycerol, propanediol, ethylene glycol, butanediol, polyvinylpyrrolidone (PVP), hydroxyethyl starch (HES), polyethylene glycol (PEG) A substantial number of improvements in the combinations of components and their respective concentrations have been incorporated in the design of the compositions and systems of this invention over that of the known art, including HypothermosolTM
Based upon the principles that have emerged from isolated organ preservation studies over the past few decades, a list of desirable properties of a hypothermic blood substitute solution has emerged, as discussed above. As outlined above, the strategic designs of solutions used for organ preservation have differed depending upon their ultimate use, either as flush solutions for static storage of the organ, or as perfusates for continuous, or intermittent, perfusion of the organ. As a unique approach, the unified solution system of this invention has been formulated with a view to developing universal solutions that may be used for both hypothermic static storage of tissues and organs, and also for machine perfusion preservation. An attempt has been made to combine the main characteristics of effective hypothermic solutions in the formulation of the base solution, and wherever possible, components that might fulfill multiple roles have been selected. For example, an extracellular base solution in accordance with this invention may be combined with various different additives to form purging solutions, organ rescue solutions, pre-implantation rinses and the like.
This strategy maximizes the intrinsic qualities of the solution that, by design as a universal tissue preservation solution, would inevitably be an improved hybrid of other hypothermic perfusates and storage media.
Salient design features for desirable base solutions are described below.
Reference may also be made to U.S. Patent No. 5,405,742, which is incorporated by reference herein in its entirety, for such features.
A fundamental biophysical property is to provide the optimum concentration of ions and colloids to maintain ionic and osmotic balance within the organ, or body tissues during hypothermia. In particular, one or more effective impermeant anions is or are included to partially replace chloride in the extracellular space and prevent osmotic cell swelling (i.e., to balance the fixed ions inside cells that are responsible for the oncotic pressure leading to osmotic cell swelling and eventual lysis during ischemia and hypothermia). A number of anions including citrate, glycerophosphate, gluconate and lactobionate, or the anionic forms of aminosulphonic acids, such as HEPES (N-2(hydroxyethyl-piperazine)N-2-ethanesulfonic acid), TES
(N-tris(hydroxymethyl)methyl-2-aminoethane sulfonic acid), MOPS
(3-(n-morpholino)propanesulfonic acid), TAPSO (3-3 N-tris(hydroxymethyl)methylaminohydroxypropane sulfonic acid) and DIPSO (2-3 N-bis(hydroxyethyl)amino 2-hydroxypropanesulfonic acid) could be suitable candidates. Lactobionate (FW=358) was used exclusively as the principal impermeant in many solutions developed in recent years; these include, for example, ViaspanTM, Hypothermosol, Celsior, Cardiosol and Churchill's solution.
Lactobionate is also known to be a strong chelator of calcium and iron and, may therefore contribute to minimizing cell injury due to calcium influx and free radical formation.
However, for organ perfusion, Belzer and Southard (Oman Preservation, Annual Review of Medicine 1994; 46:235-247) teach against using lactobionate in a perfusion solution, especially for kidneys. Moreover, Belzer and Southard have explained the importance of lactobionate as a crucial component of UW solution that could not be successfully replaced with a similar anion, such as gluconate.
Belzer and Southard therefore teach separate and exclusive roles for lactobionate and gluconate in organ preservation solutions using either the cold static storage, or perfusion approaches, respectively. Furthermore, Belzer teaches away from a combined approach that attempts to incorporate the best of simple flush storage and continuous perfusion because of "spectacularly poor results" (see "Or~L~ Preservation:
Basic and Applied Aspects", Ed D.E. Pegg et al., 1982, p339).
The osmotic components of this invention can be supplemented by the inclusion of sucrose and mannitol, the latter of which also possesses properties as a hydroxyl radical scavenger and reduces vascular resistance by inducing a prostaglandin-mediated vasodilation which may be of additional benefit.
A macromolecular oncotic agent is an important component of a blood substitute perfusate to help maintain oncotic pressure equivalent to that of blood plasma. Any oncotic agent that is sufficiently large to prevent or restrict its escape from the circulation by traversing the fenestration of the capillary bed may be considered. Examples of acceptable colloidal osmotic agents include blood plasma;
expanders, such as human serum albumin; hetastarch or hydroxyethyl starch (HES), an artificial colloid derived from a waxy starch and composed almost entirely of amylopectin with hydroxyethyl ether groups introduced into the alpha (1-4) linked glucose units, a gelatin polypeptide; polyethylene glycol; and polysaccharide polymers of D-glucose, such as the dextrans. A preferred oncotic agent is dextran-40 (average mol wt = 40,000 daltons) to balance the hydrostatic pressure of perfusion and help prevent interstitial edema. It has long been known that dextran can improve the efficiency of the removal of erythrocytes from the microvasculature of cooled organs by inhibiting red cell clumping and by increasing intravascular osmotic pressure and reducing vascular resistance. Dextran is widely used clinically as a plasma expander and is readily and rapidly excreted by the kidneys. Moreover, there is ample recent evidence that dextran-40 is an effective and well tolerated colloid in modern cold storage solutions for organ preservation.
Retention of the colloid in the vascular space is an important consideration for achieving optimal oncotic support and, in the context of isolated organ perfusion over 5 several days, other colloids might be preferred to dextran-40. However, for whole body perfusion for the order of 3 hours, the relative permeability of different colloids may be of less importance than other qualities, and non-antigenic clinical grade dextran-40 is preferred for the reasons outlined above. Any dextran that should permeate into the interstitial space during the hypothermic procedure will also be 10 readily eluted upon return to physiological conditions. Another possible advantage of the use of dextran is that the viscosity of the blood substitute will not be as high as with some other colloids, such as HES. This is also an important consideration for rheological aspects of whole-body, or even just cerebral perfusion. While the preferred embodiment described herein relates to kidney preservation in the first 15 instance, the universal design strategy of the unified solution system is intended for widespread hypothermic applications including total body washout.
Ionic balance, notably the Na+/K+ and Caa+/Mga+ ratios, is preferably adjusted to restrict passive diffusional exchange at low temperatures when ionic pumps are inactivated. In the preferred high potassium embodiment of the unified solution described above, the concentration of monovalent cations Na+ and K+ are approximately equimolar to restrict their passive transmembrane exchange. In an alternative low potassium embodiment of the unified solution system, the balance of Na/K is changed to 125/25 in consideration of concerns for a toxic effect of high concentrations of K+ in the heart. The concentration is kept sufficiently high to retain a cardioplegic effect of the solution. In the area of cardioplegia and myocardial preservation, there is good evidence for improved survival using elevated concentrations of magnesium and very low, but not zero, calcium to avoid the putative calcium paradox. Some glucose is included in these hypothermic solutions as a substrate, but the concentration is low to prevent exogenous overload during hypothermia. This can potentiate lactate production and intracellular acidosis by anaerobic glycolysis.
Acidosis is a particular hazard during hypothermia and attention has been given to the inclusion of a pH buffer that will be effective under non-physiological conditions that prevail at low temperatures. HEPES is preferred as one of the most -~~~ ed.'s~ ~, a~ ~~"'>~, ~ ~°~ '~ FAg 705 ' 2T87 ''":BRIDGE. ~ '~ ~~ w;
" ,.'t--'r ~. ~ . _~ ~ .
!b widely used biocompatible aminosulphonic acid buffers, which have been shown to possess superior buffering capacity at low temperatures, and have been included as a major component of other hypothermic tissue preservation media. Synthetic zwitterionic bugers, such as HEPES,~also contribute to osmotic support in the ~draeellular compartment by virtue of their molecular size (HEPES ~ 238 daltons).
Adenosine is a mufti-faceted molecule and may be included in the hypothermic blood substitutes not only as a substrate for the regeneration of ATP during rcwarming, but also as a vasoactive component to facilitate efficient vascular flushing by vasodilatation. Glutathionc~may be included as a cellular anti-oxidant aad hydroxyl radical scavenger, as well as a co-factor for glutathione pemxidase which enables metabolism of lipid peroxides and hydrogen peroxide.
Perfusion apparatus and methods in which embodiments of this unvention can be utilized ai~c described in copending U.S, patent application serial number 09!162,128, which is hereby incorporated by reference herein in its entirety.
Exemplary aqueous formulations of both intracellular and extracellular base solutions are illustrated below. The formulations can contain substantially about the amounts listed.
~~mnlary Intracellular Base Solutions onic 40-80 mM Na*;
50-90 mM K+;
0.01-0.1 mM Ca**;
5-25 mM Mgi'*;
20-40 mM Cl';
HB
1-5 mM HzP04;
3-? mM HC03;
25-50 mM HYPES;
25-50 mM Lactobionate;
1 OrxtM-1 M Sucrose;
15-30 Mannitol;
1-10 Glucose;
50-100 l,Iluconate;
~oUoids 6% Dextran 40;
SUHSTZTUTE SHEET

Gmnf~neoioif 9QI~n_ .~~~,~-~~:i' FAl 703~t3 Y787 ~.a~~..;,h.." .~11lbn. ~ x~..x .:_ ,.>
. CA 02398614 2002-07-30 Pharmacvlosics 0.1-2 mM Adcnosine; and 1-5 mM Glutathione.
High PotassiuntExemplary Intracellular Hasc Solution 62.5 mM Na'';
70.0 mM K~;
0.05 mM Ca'~'+
15.0 mM Mg'~"~;
30.1 mM Cl';
~~i Buffers 2.5 mM H2P0~;
5.0 mM HC03;
35.0 mM APES;
30.0 mM Lactobionate;
15.0 mM Sucxose;
25.0 mM Mannitol;
S.0 mM Glucose;
70.0 aiM Gluconate;
colloids 6% Dextran 40;
a.
Pharm~olosics 2.0 mM Adenosine; and 3.0 mM Glutathione.
'Ibis exemplary Intracellular Baac Solution has an osmolality (m0of 350, a pH of about 7.6, and a [K~[Cl~ of about 2100.
Low Potassium ExemIntracellular B,~c Solutions Ionic 100-I50 mM Na'';
15-40 mM K+;
0.01-0.1 mM Ca''"'';
5-25 mM Mg''"';
20-40 mM Cl';
p~ Buffers 1-5 mM H=P04; ' 3-7 mM HC03;
2s-so mM t~PES; , Imoermeants 25-50 mM Lactobionate;
10 mM-1 M Sucrose;
SUBSTITUTE SHEET
Fmnf~nEe~oit 9p. l~n. 9'l'.99 ~' ".~~ yms..,.i ~ ~ !~~;z~a ~.~r3' "=" 1'!1A I UJ.~ G I O 1 ~ -~~1~~<r Vw. ~
..
. dTVa ~ ~,< ~ -_- f ,f"> ._ P ~ ..
.1 .

' ' CA 02398614 2002-07-30 15-30 mM Mannitol;
1-10 mM Glucose; , 50-100 n~Ivi Gluconatc;
C I_o loids 6% Dextrar~ 40;
Pharmacoloaics 0.1-2 mM Adenosine; and 1-5 mM Glutatluone.
Excmolarv Low Potassium Intracellular Solutions . 125 mM Na ;
25.0 mM K'';
0.05 mM Cs''w ' 15.0 mM Mgt;
34.1 mM Cl-;
DH Buffe~~ ° .
. 2.5 mM H1P04;
5.0 mM HC03;
35.0 mM HEPES;
Imp' , ..
~ 30.0 mM Lactobionate; ' .
~5.o mM sucrose; ~ ~ . v 25.0 mM Mannitol; : . ' 5.0 mM Glucose; .
70.0 mM Gluconate;
' Colloids 69io Deac4an 40;
- 2.0 azM Adenosine; and 3.0 mM Glutathione. ' ~xemnlarY Extiaccllular~a~e Solutions 120-160 mM Na+;
3-9 mM ICr; ' ;
1-3 mM C ~;
1-10 mM Mg'";
too-150 ~M Cr;
1-10 mM (S04)~';
p~~, Buffers ~ .
1-3 mM H~P04;
20-30 mM HC03;
SUBSTITUTE SHEET
Fmnfonec~oit 7R..lan. 78:77 ~ ' n-y ~ ~ .1*~y' ~ z ~ ~°~~ ~~- ~' FAX 7 U 3 Y 7 !l ~f a ,,~ - tllt 1 L
bt. . V ~ ~ ; . ' ' .r~Q~
( t -,, ~~ r ' l s-is mM HEPES;
s-10 mM Glucose;
6% Dextran 40;
~Pharmacoloaics 0.1-2 mM Adenosine; and 1-5 mM Glutathionc.
~xemolarY Extracellular Base Solution Ionic 141.2 mM Na'';
1 s 6.0 mM K''; .
1.5 tnM Cs~'"~;
s.0 mM Mgi"~;
122.0 mM Cl' l .o mM (so4~~-;
ZO
I .2 mM HzPOo; ' zs.a ~nM Hco3;
2s.0 mM HEPES;
zs .~nt~
s.o mM Glucose;
~QuQid~
30 6% bextran 40; - j Pr 1.0 mM Adenosine; and 3.0 mM Glutathione.

3s ?his exemplary extracellular base solution has an osmolality (mOsmlKg) of 31 s, a pH of about 7.s, and a [K+][Cl ] of about 732.

EXA1V~LES
EXAMPLE 1: Hy,~Prcs~ation of Tissue Culture Cells Preliminary experiments were undertaken to examine the viability of cells after 40 hypothermic exposure in the high potassium exemplary embodiment of this invention and in other Storage media at 4°C. These experiments were carried out using a canine kndney cell lint (MDCK), since this provided a comparative reference with previous .
work in the development of hypothermic solutions. Cells were plated at 1 x 104 celis/well in Dulbecco's Modified Essential Medium (DMElvi) for culture at 37°C.
sussTITirrE sHEEr ~~'~ Froofan~s~ait 9R..lan. 93:79 The next day, the plate was placed on ice and the DMEM media was replaced with 100 w) of one of the four storage solutions evaluated in this experiment, as specified in Fig. 1. Each plate was kept at 4°C for either one or five days. After storage, the vehicle solution was removed and replaced with DMEM culture medium S in preparation for the assessment of viability using Alamar Blue, which is a non-toxic indicator of mitochondria) oxidative phosphorylation and as such directly measures the metabolic status of cells.
It can be seen from the data summarized in Fig. 1 that, after 24 hours exposure at 4°C, the viability index for cells stored in Dulbecco's Modified Essential Medium 10 (DMEM) was about as low as the "no-cell" baseline control, indicating that standard DMEM culture medium confers no protection during this period of hypothermic storage. The response of cells stored in EuroCollins (EC) was marginally better but significantly inferior compared with the indices for cells stored in the two "intracellular" solutions (the high potassium exemplary embodiment of this invention, 15 and ViaspanTM). We conclude that "intracellular" solutions, such as that of the invention and Viaspan~, offer superior cytoprotection during hypothermic exposure, compared with either standard culture media or EuroCollins organ preservation solution. Moreover, on the basis of this preliminary study, the new solution of this invention offers at least equivalent protection to other established hypothermic 20 solutions, including the industry standard for organs, ViaspanTM. These experiments have been expanded to include 24-hour storage of porcine kidneys, as described below.
These studies have been extended to inelude two additional cell types i~ vitro and examined cell viability using Alamar Blue at different time points following 2S hypothermic storage. The vascular smooth muscle cell Iine (A10) and a bovine pulmonary endothelial cell line (CPAE) were used in these experiments. Cells were plated at a density of 1 x 104 cells/well in standard cell culture media. The next day, the plates were placed on ice and the media was replaced with 100 p) of the various storage solutions (see Figs. 2-S).
The plates were stored in the refrigerator at 4°C for either one or three days.
Following storage, the plates were again placed on ice and the storage solutions were replaced with media for measurement of cell viability with Alamar Blue.
Viability was measured at two points, either immediately after exposure (Figs. 2-5), or for six consecutive days after exposure at 4°C (Fig. 6 and 7).
The comparative viability of cells at the end of each storage interval is shown in Figs. 2-5. Viability of both cell types after 24 hours of hypothermic storage in UHI~ was equivalent or better than in ViaspanTM (Figs. 2-5) After three days at 4°C, UHK demonstrated better viability than ViaspanTM for A10 cells, and similar viability for CPAE (Figs. 4 and 5). By comparison, the other solutions, DMEM, EC and Belzer's MPS (Machine Perfusion Solution), all demonstrated inferior protection after three days of storage. Viability indices were no greater than background levels (no cells) for A10 cells at either one or three days hypothermic storage. In EC
solution, CPAE cells demonstrated some viability after one day of hypothermic storage, but after three days, viability in EC was also down to background levels. DMEM and the Belzer solution were both at background levels after one and three days hypothermic storage for the CPAE cells.
Measurement of cell viability immediately after low temperature storage does not necessarily give a true indication of cell survival. Over time, cells may undergo further changes, including the repair of sub-lethal injury or cell death through the processes of apoptosis and necrosis. Whatever the eventual fate of hypothermically exposed cells may be, the processes required time for full manifestation and cell survival curves during the days following return to physiological temperatures are informative about the true state of viability. In light of these conditions, we further evaluated post-hypothermic viability during six consecutive days of culture at 37°C
(Figs. 6 and 7). Such evaluation was possible because Alamar Blue is non-toxic to cells and can be used repeatedly on the same batch of cells as a non-destructive viability assay.
Figs. 6 and 7 illustrate that, in general, the high potassium example ("HK") provided equivalent, or better, preservation, than the other solutions. Both "intracellular" solutions, ViaspanTM and the high potassium example, proved to be superior compared with the other solutions. After three days of hypothermic storage, only CPAE cells stored in ViaspanTM or the high potassium example, and only the A10 cells kept in the high potassium example, were able to proliferate in culture.
These and similar observations indicate that the high potassium example is at least equal to, or superior to, other commonly used solutions employed for hypothermic storage of tissues and organs.

EXAMPLE 2: Comparative Cr~preservation of Cells Using a Preferred Embodiment of the Exemplary I~-Ii.ah-K Intracellular-base Solution.
The same system as that described above for cells in microtiter plates was used to evaluate a preferred embodiment of the exemplary High-K Intracellular-base solution as a new carrier solution for cryoprotective additives (CPA).
Specifically, a phosphate-free formulation of the new High-K solution was prepared to avoid the known precipitation of divalent canon phosphates in the presence of the cryoprotectant DMSO at low temperatures. Cell survival was compared after freezing and thawing in the presence of a range of concentrations of DMSO prepared in either the new phosphate-free HK-cryoprotectant vehicle solution (HK-CV), or EuroCollins medium. EuroCollins is an organ preservation solution that has also been used in the field of cryobiology as a vehicle solution for cryoprotectants.
Cells were plated at 2x104 cells/well and after overnight incubation regular culture medium was replaced with the experimental CPA mixtures using the following protocol: Plates were cooled to -80°C at a controlled rate (1°C/min) and stored overnight at -135°C. The plates were thawed using a two-step warming protocol and the CPA was diluted using mannitol as an osmotic buffer. Cell viability was assessed using the Alamar Blue assay of cellular metabolic activity at 37°C.
Figures 15 and 16 show the comparative viability of two cell types after cryopreservation in the respective media. The data show that optimum survival of both types of cells was achieved using 1-2 molar DMSO, but the percentage viability, normalized to untreated cells, was markedly higher for cells preserved in the new HK
intracellular base vehicle solution compared with the EuroCollins medium. This was consistent for both the A10 smooth muscle cells and the bovine corneal endothelial (BCE) cells.
EXAMPLE 3: Structural Integrity of Hypothermically Stored Veins Hypothermic storage of whole organs flushed or perfused with a preservation solution is common practice in clinical transplantation. This procedure leaves the vascular endothelial cells in direct contact with the preservation medium during the cold ischemic period. The effect of storage conditions on the integrity of vascular endothelium is therefore of crucial importance for the quality of preservation of intact organs.
A pilot study was conducted to compare microscopic changes in tissue morphology when harvested vessels were immersed and transported in the high potassium example hypothermic preservation solution compared with the Dulbecco's Minimum Essential Medium (DMEM) which is a common culture medium used to incubate and transport tissues ex vivo. For three separate experiments, fresh jugular veins were harvested from rabbits (by the method we have used extensively) and immersed in either pre-cooled DMEM or the high potassium example. Both solutions were kept on ice. Tissue samples were then transported to the laboratory where the vessels were cut into segments for fixation and processing for histopathology.
The total cold ischemia time in these experiments was relatively short, 34.5 ~ 9.5 minutes for samples transported in DMEM, and 37 ~ 4 minutes for samples in the high potassium example.
Tissue immersed and transported in DMEM on ice exhibited microscopic changes within the tunica intima and tunica media, as shown in Fig. ~. The intima was intact, but there was extreme vacuolization (indicated by the arrows) of the underlying basal lamina causing, in turn, extrusion of the endothelial cells into the lumen and giving a "rounding-up" appearance. Apart from this vacuolization, the endothelial cells had a near normal appearance. The smooth muscle cells (SM) had a somewhat shrunken appearance with irregular contours. The tunica adventitia was essentially normal. The lumen L is also shown.
In contrast, jugular veins transported in the high potassium example demonstrated little if any histological changes compared with the DMEM group as shown in Fig. 9. The tunica intima was intact with little evidence of vacuolization of the underlying basement membrane. The smooth muscle cells (SM) did not appear shrunken and were in a normal, horizontal orientation.
These preliminary experiments demonstrate that in veins stored in culture medium, cold ischemia results in microscopic changes within less than one hour. In contrast, the high potassium example-stored veins did not demonstrate these microscopic changes. These are important observations since, historically, it has been concluded that saline is not the best preservation fluid even for short-term storage.
Moreover, several studies have demonstrated how sensitive the endothelial layer is to handling. For example, it has been shown that cold preservation resulted in severe endothelial cell changes and significant detachment after only 3 hours of cold storage.
Many other studies, however, have documented morphologic and functional damage to endothelial cells induced by cold storage in autologous whole blood, normal saline j '~ "a's ~ t yd'~r ~,. .x:4 ~.. Z' ~l1 1 V d~ ~ L I 0 1 ~, M 11i V1. s g.
", .. ' , solution, and Ringer's lactate. A recent study compared the "Gold Standard"
organ preservation solution, Viaspan~, with sutologuous whole blood and normal saline solution for canine vein storage. The study demonstrated that post-implantatyon intimal hyperplasia and vascular physiology were similar in Viaspan~-treated vein grafts and fresh, untreated controls. In contrast, both the other treatment groups demonstrated statisticahy significant changes. Nevertheless, others have reported that hyperkalemic-type solutions (of which Viaspann"i is an example) caa cause thrombophlebids and decreased fibrinolytic activity of venous endothelium. As discussed above, there were documented concerns for the detrimental effects on the heart of very high potassium levels (>I04mlvl) found in many organ preservation solutions.
F~KAMPLE 4: G~m~araative Organ Presexvation Studies Usul,g The H,~ah Potassium A. Comparison of new High-K tHK) solution with Belzer Machine Perfusion Solution A new intracellular base high potassium solution was compared with an established solution (Belzer machine perfusion solution (MPS]) during continuous 18-20 hour perfusion preservation of canine kidneys procured after one hour of warm ischemia. Following procurement from adult mongrel dogs (n-5), both kidneys were flushed with Viaspan solution and stored at 4°C for approximately two hours prior to initiation of perfusion. Perfusion was performed using a muhiple organ recovery perfusion prototype with two independent circuits. One kidney was pcrfuscd with the new intracellular base high potassium solution and the contralateral kidney with Bclzer WS, both at ~9°C and a controlled pressure of 35-40 mmHg. After houxs of perfusion, the kidneys were transplanted ectopieally using a canine carotid artery model.
Figures I O and 11, which show that mean arkerial flow was higher at each time point for kidneys perfused with the new solution compared with MPS and the vascular resistance curve was lower for the new solution group of kidneys. In general, these physiological parameters were similar in both groups during fn vfdo perfusion. At the end of one hour in vlvo, however, the new solution -perfused kidneys exhibited higher arterial flow rates (p<.OS) and lower internal renal resistances (p<.05) than the kidneys perfused with Belzer WS {Fig. 67.
SUBSTITUTE SKEET
'~ FmofanRC~ait ~A..lan. 93:79 The results suggest that the new solution may be a superior alternative to Belzer MPS for perfusion preservation of kidneys.
B. Comparison of new High K solution with Viaspan solution in a swine model 5 The efficacy of a new intracellular-base high potassium solution, which fulfills the minimum theoretical requirements for organ cold storage, was compared with commercial Viaspan in an established pig kidney transplant model. These ih vivo studies were performed using an autologous swine model in which a kidney was removed, flushed and stored with either the new high potassium solution (n=5) or 10 Viaspan (n=5) for at least 20 hours prior to reimplantation. The contralateral unoperated kidney was removed 5-7 days later and the performance of the preserved lcidney determined by assessments of BUN, creatinine and histopathology. Fig.
14, presents Biochemical indicators (Creatinine and BLTN) of kidney function for pig kidneys transplanted after a minimum of 20 hours cold storage preservation in either 15 new intracellular-base high potassium solution, or Viaspan, shows that no statistically significant differences was observed between the two groups of all assessment criteria.
In conclusion, these pre-clinical results indicate that the base formulation of the new high potassium solution provides hypothermic kidney cell preservation that is at least equivalent to that of the established standard, Viaspan, during clinically relevant 20 periods of kidney storage.

Claims

WHAT IS CLAIMED IS:

1. A method of procuring, preserving, transplanting and/or bloodless surgery involving an organ or tissue, comprising the steps of:
(a) providing a base electrolyte solution, comprising:
40-80 mM Na+;
50-90 mM K+;
Ca2+;
Mg2+, Cl2+;
H2POa;
HCO3;
HEPES;
Lactobionate;
Sucrose;
Mannitol;
Glucose;
Gluconate;
Dextran 40;
Adenosine; and optionally Glutathione;
(b) mixing a first portion of the base solution with a first additive to produce a first mixed solution;
c) contacting the first mixed solution with an organ or tissue during at least one stage of procurement, preservation, transplantation or bloodless surgery of that organ or tissue;

(d) mixing a second portion of the base solution with a second, different additive to product a second, different mixed solution; and (e) contacting the second mixed solution with an organ or tissue during at least one stage of procurement, preservation, transplantation or bloodless surgery of that organ or tissue.

2. The method of claim 1, wherein said base solution comprises:
40-80 mM Na+;
50-90 mM K+;
0.01-0.1 mM Ca++;
5-25 mM Mg++;
20-40 mM Cl-;
1-5 mM H2PO4;
3-7 mM HCO3;
25-50 mM HEPES;
25-50 mM Lactobionate;
mM -1M Sucrose;
15-30 mM Mannitol;
1-10 mM Glucose;
50-100 mM Gluconate;
6% Dextran 40;
0.1-2 mM Adenosine; and optionally 1-5 mM Glutathione.

3. A method of praocuring, preserving, transplanting and or bloodless surgery involving an organ or tissue, comprising the steps of:

(a) providing a base electrolyte solution, comprising:
120-160 mM Na+;
3-9 mM K+;
1-3 mM Ca++;
1-10 mM Mg++;
122-150 mM Cl-;
1-10 mM (SO4)2-;
1-3 mM H2PO4-;
20-30 mM HCO3;
5-15 mM HEPES;
5-10 mM Glucose;
6% Dextran 40;
0.1-2 mM Adenosine; and optionally 1-5 mM Glutathione;
(b) mixing a first portion of the base solution with a first additive to produce a first mixed solution;
(c) contacting the first mixed solution with an organ or tissue during at least one stage of procurement, preservation, transplantation or bloodless surgery of that organ or tissue;
(d) mixing a second portion of the base solution with a second, different additive to produce a second, different mixed solution; and (e) contacting the second mixed solution with an organ or tissue during at least one stage of procurement, preservarion, transplantation or bloodless surgery of that organ or tissue.

4. The method of claim 1 or 3, wherein said organ or tissue is isolated.

5. The method of claim 1 or 3, wherein said organ or tissue is in a cadaver.

b. The method of claim 1 or 3, wherein said organ or tissue is in a living patient.

7. The method of claim 6, wherein said patient is undergoing bloodless surgery.

8. The method of claim 1, 2 or 3, wherein the base solution does not contain Glutathione; and further comprising the step of adding said Glutathione to the bass solution or to the mixed solution.

9. The method of claim 1 or 3, wherein the organ or tissue of step (c) and the organ or tissue of step (e) are the same organ or tissue.

10. The method of claim 1 or 3, wherein the organ or tissue of step (c) and the organ or tissue of step (e) are different organs or tissue.

11. The method of claim 1 or 3, wherein at least one said contacting step comprises purging vasculature of said organ or tissue of blood in preparation fox preservation; and the additive of said at least one said contacting step forms a purge solution from said base solution.

12. The method of claim 1 or 3, wherein at least one said contacting step comprises protecting and maintaining cellular stability during cold storage of the organ or tissue; and the additive of said at least one said contacting step is a cytoprotcctive additive that forms a maintenance solution from said base solution.

13. The method of claim 1 or 3, wherein at least one said contacting step comprises restoring viability of said organ or tissue by near normothermie pc~usion;
and the addictive of said at least one said contacting step is a rescue additive.

14. The method of claim 1 or 3, wherein at least one said contacting step caxnprises flushurag unwanted molecules away from an organ or tissue; and the additive of said at least one said contacting step is a rinse additive.

15. The method of claim 14, wherein said unwanted molecules arc preservation solution molecules, and said at least one said contacting step is followed by reimplantation of the organ or tissue.

16. The method of claim 14, wherein said unwanted molecules are erythrocytes and other blood coonponents, and said at least one said contacting step is followed by a preservation process.

17. The method of claim 1 or 3, wherein at least one said stage of step (c) comprises cryopreservation of said organ ar tissue by sub-zero cooling; and said first additive is a cxyoprotective additive.

18. An organ and/or tissue procurement, preservation, transplantation and/or bloodless surgery solution, comprising:
40-80 mM Na+;
50-90 mM K+;
0.01-0.1 mM Ca++;
5-25 mM Mg++;
20-40 mM Cl-;
1-5 mM H2PO4;
3-7 mM HCO3;
25-50 mM HEPES;
25-50 mM Lactobionate;
mM -1M Sucrose;
15-30 mM Mannitol;

1-10 mM Glucose;
50-100 mM Gluconate;
6% Dextran 40;
0.1-2 mM Adenosine; and optionally 1-5 mM Glutathione.

19. An organ andlor tissue procurement, preservation, transplantation and/or bloodless surgery solution, comprising:
120-160 mM Na+;
3-9 mM K+;
1-3 mM Ca++;
1-10 mM Mg++;
122-150 mM Cl-;
1-10 mM (SO4)2-;
1-3 mM H2PO4;
20-30 mM HCO3;
5-15 mM HEPES;
5-10 mM Glucose;
6% Dextran 40;
0.1-2 mM Adenosine; and optionally 1-5 mM Glutathione.

20. The solution of claim 18 or 19, wherein said solution further comprises 1-5 mM Glutathione.

21. The solution of claim 18 of 19, wherein said solution further comprises about 3.0 mM Glutathione.

22. The solution of claim 19, wherein said solution has an osmolality (mOsm/Kg) of about 350, a pH of about 7.6, and a [K+[Cl] of about 2100.

23. The solution of claim 19, wherein said solution has an osmolality (mOsm/Kg) of about 315, a pH of about 7.5, and a [K+][Cl-] of about 732.

24. A solution kit comprising a saleable package containing:
1) at least one first container containing at least one base electrolyte solution, wherein the solution comprises 40-80 mM Na+;
50-90 mM K+;
Ca2+;
Nd2+;
Cl-;
H2PO4;
HCO3;
HEPES;
Lactobionate;
Sucrose;
Mannitol;
Glucose;
Gluconate;
Dextran 40; and Adenosine; and 2) at least two second containers respectively containing different additives for converting said base solution to different mixed solutions for use in different stages of procurement, presentation or transplantation of an organ or tissue.

25. The kit of claim 24, wherein said base solution comprises:
40-80 mM Na+;
50-90 mM K+;
0.01-0.1 mM Ca++;
5-25 mM Mg++;
20-40 mM Cl-;
1-5 mM H2PO4;
3-7 mM HCO3;
25-50 mM HEPES;
25-50 mM Lactobionate;
mM -1M Sucrose;
15-30 mM Mannitol;
1-10 mM Glucose;
50-100 mM Gluconate;
6% Dextran 40;
0.1-2 mM Adenosine; and optionally 1-5 mM Glutathione.
26. A solution kit, comprising a sealable package containing:
1) at least one first container containing at least one base electrolyte solution, wherein the solution comprises:
120-160 mM Na+;
3-9 mM K+;
1-3 mM Ca++;
1-10 mM Mg++;
122-150 mM Cl';

1-10 mM (SO4)2-;
1-3 mM H2PO4:
20-30 mM HCO3;
5-15 mM HEPES;
-5-10 mM Glucose;
6% Dextran 40; and 0.1-2 mM Adenosine; and 2) at least two second containers respectively containing different additives for converting said base solution to different mixed solutions for use in different stages of procurement, preservation or transplantation of an organ or tissue.
27. The kit of claim 24, 25 or 26, wherein said base solution further comprises 1-5 mM Glutathione or wherein said kit further comprises a container containing an amount of Glutathione that, when added to the base solution, produces a base solution that comprises 1-5 mM Glutathione.
28. The kit of claim 24, 25 or 26, wherein said base solution further comprises about 3.0 mM Glutathione or wherein said kit further comprises a container containing an amount of Glutathione that, when added to the base solution, produces a base solution that comprises about 3.0 mM Glutathione.
29. The kit of claim 27, wherein said base solution has an osmolality (mOsm/Kg) of about 350, a pH of about 7.6, and a [K+][Cl-] of about 2100.
30. The kit of claim 27, wherein said base solution has an osmolality (mOsm/Kg) of about 315, a pH of about 7.5, and s [K+][Cl-] of about 732.
31. The kit of claim 24 or 26, wherein at least one said additive is a purge additive for purging an organ or tissue of blood.

32. The kit of claim 24 or 26, wherein at least one said additive is a cryoprotection additive.
33. The kit of claim 24 or 26, wherein at least one said additive is a rescue additive.
34. The kit of claim 24 or 26, wherein at least one said additive is a rinse additive for flushing unwanted preservation molecules away from an organ or tissue.
CA002398614A 2000-01-31 2001-01-24 System for organ and tissue preservation and hypothermic blood substitution Abandoned CA2398614A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US17915300P 2000-01-31 2000-01-31
US60/179,153 2000-01-31
US09/628,311 US6492103B1 (en) 2000-01-31 2000-07-28 System for organ and tissue preservation and hypothermic blood substitution
US09/628,311 2000-07-28
PCT/US2001/002237 WO2001054495A1 (en) 2000-01-31 2001-01-24 System for organ and tissue preservation and hypothermic blood substitution

Publications (1)

Publication Number Publication Date
CA2398614A1 true CA2398614A1 (en) 2001-08-02

Family

ID=26875055

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002398614A Abandoned CA2398614A1 (en) 2000-01-31 2001-01-24 System for organ and tissue preservation and hypothermic blood substitution

Country Status (7)

Country Link
US (2) US6492103B1 (en)
EP (1) EP1257169B1 (en)
JP (1) JP2003520859A (en)
AU (2) AU2001231095B2 (en)
CA (1) CA2398614A1 (en)
DE (1) DE60102359T2 (en)
WO (1) WO2001054495A1 (en)

Families Citing this family (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8409846B2 (en) 1997-09-23 2013-04-02 The United States Of America As Represented By The Department Of Veteran Affairs Compositions, methods and devices for maintaining an organ
US7250292B2 (en) * 2000-01-26 2007-07-31 21St Century Medicine Hypertonic reduction of chilling injury
GB0028414D0 (en) 2000-11-22 2001-01-03 Univ Leeds Flush preservation solution
WO2002086134A2 (en) * 2001-04-23 2002-10-31 Amaxa Gmbh Buffer solution for electroporation and a method comprising the use of the same
US20040202993A1 (en) * 2003-04-10 2004-10-14 Poo Ramon E. Apparatus and method for organ preservation and transportation
US7897327B2 (en) * 2003-06-02 2011-03-01 Organ Recovery Systems, Inc. Method and apparatus for pressure control for maintaining viability of organs
US20050170019A1 (en) * 2003-10-22 2005-08-04 Fred Hutchinson Cancer Research Center Methods, compositions and devices for inducing stasis in cells
US20050136125A1 (en) * 2003-10-22 2005-06-23 Roth Mark B. Methods, compositions and devices for inducing stasis in cells, tissues, organs, and organisms
AU2004285468B2 (en) 2003-10-22 2011-02-24 Fred Hutchinson Cancer Research Center Methods, compositions and devices for inducing stasis in cells, tissues, organs, and organisms
US7504201B2 (en) 2004-04-05 2009-03-17 Organ Recovery Systems Method for perfusing an organ and for isolating cells from the organ
ES2348736T3 (en) 2004-10-07 2010-12-13 Transmedics, Inc. SYSTEMS AND METHODS FOR EX-LIVE ORGAN CARE.
US9301519B2 (en) 2004-10-07 2016-04-05 Transmedics, Inc. Systems and methods for ex-vivo organ care
US8304181B2 (en) 2004-10-07 2012-11-06 Transmedics, Inc. Method for ex-vivo organ care and for using lactate as an indication of donor organ status
US7713686B2 (en) * 2004-12-03 2010-05-11 Biorep Technologies, Inc. Organ preservation container and method
MX2007013196A (en) 2005-04-20 2008-01-18 Hutchinson Fred Cancer Res Methods, compositions and articles of manufacture for enhancing survivability of cells, tissues, organs, and organisms.
US9078428B2 (en) * 2005-06-28 2015-07-14 Transmedics, Inc. Systems, methods, compositions and solutions for perfusing an organ
US20070048725A1 (en) * 2005-08-25 2007-03-01 Arrington Ben O'mar Machine perfusion of tissue grafts for transplantation
US20070098695A1 (en) * 2005-10-31 2007-05-03 Deolden James Methods of islet separation during isolation
KR20090010176A (en) * 2006-03-31 2009-01-29 아데노바이오 엔.브이. Compositions, methods, and kits using adenosine and inosine in combination for diagnosis and treatment
EP3677118A1 (en) 2006-04-19 2020-07-08 Transmedics, Inc. Systems for ex vivo organ care
US7790437B2 (en) * 2006-12-14 2010-09-07 Biorep Technologies, Inc. Organ transportation device
BRPI0807918A2 (en) * 2007-02-17 2017-05-16 Harvard College compositions and method for preserving tissue
US8986978B2 (en) * 2007-03-01 2015-03-24 Lifeline Scientific, Inc. Perfusion regulation
US9457179B2 (en) 2007-03-20 2016-10-04 Transmedics, Inc. Systems for monitoring and applying electrical currents in an organ perfusion system
US7960098B2 (en) * 2007-07-12 2011-06-14 Warsaw Orthoperic, Inc. Methods and compositions for the preservation of cells and tissues
US7892724B2 (en) * 2007-07-12 2011-02-22 Warsaw Orthopedic, Inc Method for enhancing the viability of mammalian cells, tissues and organs using a solution comprising two low molecular weight PEGs
US8835104B2 (en) 2007-12-20 2014-09-16 Fenwal, Inc. Medium and methods for the storage of platelets
US8420380B2 (en) 2008-01-31 2013-04-16 Transmedics, Inc. Systems and methods for ex vivo lung care
JP2011512350A (en) 2008-02-15 2011-04-21 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Blood substitute solution
EP2271208A1 (en) 2008-03-27 2011-01-12 Biolife Solutions, Inc. Mehtod, system, and apparatus for hypothermic collection, storage, transport and banking of birth tissue
US20090263780A1 (en) * 2008-04-17 2009-10-22 Yanming Wang Organ preservation fluid
US20100081926A1 (en) * 2008-09-29 2010-04-01 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Histological facilitation systems and methods
US20100081924A1 (en) * 2008-09-29 2010-04-01 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Histological facilitation systems and methods
US20100081927A1 (en) * 2008-09-29 2010-04-01 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Histological facilitation systems and methods
US20100081916A1 (en) * 2008-09-29 2010-04-01 Searete Llc, A Limited Liability Corporation Of The State Of Delaware. Histological facilitation systems and methods
US20100081915A1 (en) * 2008-09-29 2010-04-01 Searete Llc, Alimited Liability Corporation Of The State Of Delaware Histological facilitation systems and methods
ES2743693T3 (en) 2009-04-29 2020-02-20 Biolife Solutions Inc Apparatus and compositions for cryopreservation of cell monolayers
US9357764B2 (en) 2009-06-18 2016-06-07 Giner, Inc. System for fluid perfusion of biological matter comprising tissue
US10091985B2 (en) * 2009-06-18 2018-10-09 Giner, Inc. Perfusing an organ with an in situ generated gas
KR101092411B1 (en) * 2009-07-22 2011-12-09 (주)시지바이오 Method for processing allograft skin and cryo-preserved allograft skin manufactured therefrom
US9943075B2 (en) * 2010-02-17 2018-04-17 Hememics Biotechnologies, Inc. Preservation solutions for biologics and methods related thereto
EP2593116B1 (en) * 2010-07-16 2016-06-29 Lifeline Scientific, Inc. Methods for increasing isolation yields of cellular products
CA2809514C (en) * 2010-09-01 2014-10-28 Organ Perfusion Pty Limited Perfusion composition
US9963679B2 (en) 2010-10-22 2018-05-08 Lifeline Scientific, Inc. Cultured pancreas islets
US20120148542A1 (en) 2010-12-10 2012-06-14 Lifeline Scientific, Inc. Machine perfusion with complement inhibitors
US9253976B2 (en) 2011-03-15 2016-02-09 Paragonix Technologies, Inc. Methods and devices for preserving tissues
US8828710B2 (en) 2011-03-15 2014-09-09 Paragonix Technologies, Inc. System for hypothermic transport of samples
US11178866B2 (en) 2011-03-15 2021-11-23 Paragonix Technologies, Inc. System for hypothermic transport of samples
US9426979B2 (en) 2011-03-15 2016-08-30 Paragonix Technologies, Inc. Apparatus for oxygenation and perfusion of tissue for organ preservation
WO2012125782A2 (en) 2011-03-15 2012-09-20 Paragonix Technologies, Inc. Apparatus for oxygenation and perfusion of tissue for organ preservation
US9867368B2 (en) 2011-03-15 2018-01-16 Paragonix Technologies, Inc. System for hypothermic transport of samples
JP2014518547A (en) 2011-04-07 2014-07-31 フェンウォール、インコーポレイテッド Automated method and system for providing platelet concentrates with reduced residual plasma volume and storage medium for such platelet concentrates
AU2012242578B2 (en) 2011-04-14 2016-07-21 Transmedics, Inc. Organ care solution for ex-vivo machine perfusion of donor lungs
US11490615B2 (en) 2011-06-09 2022-11-08 Lifeline Scientific, Inc. Assessing, maintaining and/or restoring viability of organs/tissues
US11021733B2 (en) 2011-09-26 2021-06-01 Qiagen Gmbh Stabilization and isolation of extracellular nucleic acids
WO2013045458A1 (en) 2011-09-26 2013-04-04 Preanalytix Gmbh Stabilisation and isolation of extracellular nucleic acids
WO2013114372A1 (en) 2012-02-01 2013-08-08 Nayacure Therapeutics Ltd. Method for inducing immune tolerance to organ transplants
US9560846B2 (en) 2012-08-10 2017-02-07 Paragonix Technologies, Inc. System for hypothermic transport of biological samples
US8785116B2 (en) 2012-08-10 2014-07-22 Paragonix Technologies, Inc. Methods for evaluating the suitability of an organ for transplant
CA2884915C (en) 2012-09-25 2022-05-17 Qiagen Gmbh Stabilisation of biological samples
US10433538B2 (en) 2012-10-12 2019-10-08 University of Pittsburgh—of the Commonwealth System of Higher Education Compositions and methods for organ preservation
US20140278468A1 (en) 2013-03-15 2014-09-18 I.D. Therapeutics Llc Apparatus and method for optimizing treatment using medication compliance patterns and glucose sensor
US10420337B2 (en) 2013-03-15 2019-09-24 Lifeline Scientific, Inc. Transporter with a glucose sensor for determining viability of an organ or tissue
US20160281133A1 (en) 2013-03-18 2016-09-29 Qiagen Gmbh Stabilization and isolation of extracellular nucleic acids
US11525155B2 (en) 2013-03-18 2022-12-13 Qiagen Gmbh Stabilisation of biological samples
JP2016520580A (en) * 2013-04-29 2016-07-14 ソマールション・エルエルシー Formulations containing poly (0-2-hydroxyethyl) starch for increasing the oxygen content, stability and shelf life of organ and tissue preservation solutions
BR112016006378A2 (en) 2013-09-24 2017-08-01 Giner Inc gas treatment system for a cell implant
EP3119197A1 (en) * 2014-03-18 2017-01-25 Qiagen GmbH Stabilization and isolation of extracellular nucleic acids
ES2839202T3 (en) 2014-06-02 2021-07-05 Transmedics Inc Ex vivo organ care system
EA035702B1 (en) 2014-07-11 2020-07-28 Акционерное Общество "Кардиосистемфарма" Universal cardioplegic solution (variants)
US11659835B2 (en) 2014-07-29 2023-05-30 21St Century Medicine, Inc. Method of introduction and removal of high concentrations of cryoprotectants by vascular perfusion
US20160095307A1 (en) * 2014-10-07 2016-04-07 NuTech Medical, Inc. Method and composition for hypothermic storage of placental tissue
USD765874S1 (en) 2014-10-10 2016-09-06 Paragonix Technologies, Inc. Transporter for a tissue transport system
CA2981588A1 (en) * 2015-04-03 2016-10-06 Tx Innovations B.V. Organ preservation composition
US10194655B2 (en) 2015-09-09 2019-02-05 Transmedics, Inc. Aortic cannula for ex vivo organ care system
ES2807897T3 (en) * 2015-10-23 2021-02-24 Rigshospitalet Stem cell therapy based on adipose stem cells
EP3377645B1 (en) 2015-11-20 2023-10-04 Qiagen GmbH Method of preparing sterilized compositions for stabilization of extracellular nucleic acids
CA3043468C (en) 2016-11-15 2021-06-22 Giner Life Sciences, Inc. Percutaneous gas diffusion device suitable for use with a subcutaneous implant
BR112019009761A2 (en) 2016-11-15 2019-08-13 Giner Life Sciences Inc self-regulating electrolytic gas generator and implant system comprising the same
CA3047360C (en) * 2017-01-17 2023-09-26 Xvivo Perfusion Ab Organ preservation and/or perfusion solutions
US11166452B2 (en) 2017-06-07 2021-11-09 Paragonix Technologies, Inc. Apparatus for tissue transport and preservation
CN107926933B (en) * 2018-01-14 2021-04-23 上海慧存医疗科技有限公司 Cell preservation solution
EP3793642A4 (en) 2018-05-17 2022-03-16 Giner Life Sciences, Inc. Electrolytic gas generator with combined lead and gas port terminals
US11452618B2 (en) 2019-09-23 2022-09-27 Dimicron, Inc Spinal artificial disc removal tool
US11632951B2 (en) 2020-01-31 2023-04-25 Paragonix Technologies, Inc. Apparatus for tissue transport and preservation
US20220145258A1 (en) 2020-11-10 2022-05-12 Vessl Therapeutics Ltd Methods of preconditioning vascular cells for transduction, methods of transduction and methods of preserving transduced cells

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2532183C3 (en) 1975-07-18 1982-03-04 Behringwerke Ag, 3550 Marburg Polyionic isotonic saline solution for the preservation of erythrocytes or for the perfusion and preservation of organs intended for transplantation
US4238482A (en) 1978-09-29 1980-12-09 Peyman Gholam A Intraocular infusion irrigation solution and method
US4403038A (en) 1979-04-11 1983-09-06 Toshio Asakura Buffered serum substitute for blood oxygen analyzer
US4271144A (en) 1979-06-06 1981-06-02 Holles Laboratories, Inc. Dextran composition for controlling corneal hydration
US4828976A (en) 1983-12-29 1989-05-09 Thomas Jefferson University Glucose free media for storing blood platelets
FR2555589B1 (en) 1983-11-30 1986-05-16 Choay Sa NOVEL DEXTRAN DERIVATIVES WITH ANTICOAGULANT ACTIVITIES IN ANTI-INFLAMMATORY, PROCESS FOR THEIR PREPARATION AND USE THEREOF AS ANTICOAGULANTS AND AS SUBSTITUTES OF BLOOD PLASMA
SE8403912D0 (en) 1984-07-30 1984-07-30 Pharmacia Ab PHARMACEUTICAL KIT OR COMPOSITION
US4696917A (en) 1985-08-01 1987-09-29 Lindstrom Richard L Irrigation solution
US4798824A (en) 1985-10-03 1989-01-17 Wisconsin Alumni Research Foundation Perfusate for the preservation of organs
US5248506A (en) 1986-03-19 1993-09-28 American National Red Cross Synthetic, plasma-free, transfusible storage medium for red blood cells and platelets
US4961928A (en) 1986-03-19 1990-10-09 American Red Cross Synthetic, plasma-free, transfusible storage medium for red blood cells and platelets
US4927806A (en) 1987-04-23 1990-05-22 The Regents Of The University Of California Saturated salt/concentrated dextran formulation to treat hemorrhage
DE3812525C1 (en) 1988-04-15 1989-06-22 Fresenius Ag, 6380 Bad Homburg, De
US4923442A (en) 1988-05-02 1990-05-08 Cryomedical Sciences Inc. Blood substitute
US5130230A (en) 1988-05-02 1992-07-14 Cryomedical Sciences, Inc. Blood substitute
US4920044A (en) 1988-11-08 1990-04-24 The Cleveland Clinic Foundation Intracellular flush solution for preserving organs
US4938961A (en) 1989-04-28 1990-07-03 Geoffrey Collins Organ preservation solution containing pokyethylene gycol and method of performing cardioplegia
US5082831A (en) 1989-12-05 1992-01-21 Cryovita Laboratories, Inc. Total body washout solution and method of use
US5112622A (en) 1990-01-19 1992-05-12 Kopp Klaus F Intravenous solutions for influencing renal function and for maintenance therapy
CA2041828A1 (en) 1990-03-05 1992-11-04 Richard L. Lindstrom Viscoelastic solution
US5145771A (en) 1990-04-12 1992-09-08 The University Of North Carolina At Chapel Hill Rinse solution for organs and tissues
US5498427A (en) 1990-11-20 1996-03-12 Pasteur Merieux Serums Et Vaccines Solutions for the perfusion, preservation and reperfusion of organs
US5290766A (en) 1991-02-18 1994-03-01 The National Heart Foundation Of New Zealand Cardioplegic compositions
CA2066374C (en) 1991-04-19 2002-01-29 Paul E. Segall Solution for perfusing primates
US5328821A (en) 1991-12-12 1994-07-12 Robyn Fisher Cold and cryo-preservation methods for human tissue slices
US5370989A (en) 1992-04-03 1994-12-06 The Trustees Of Columbia University In The City Of New York Solution for prolonged organ preservation
US5552267A (en) 1992-04-03 1996-09-03 The Trustees Of Columbia University In The City Of New York Solution for prolonged organ preservation
US5306711A (en) 1992-06-24 1994-04-26 Georgetown University Organ preservative solution
US5599659A (en) 1993-03-11 1997-02-04 Breonics, Inc. Preservation solution for ex vivo, warm preservation of tissues, explants,organs and vascular endothelial cells comprising retinal-derived fibroblast growth factor, cyclodextrin and chondroitin sulfate
AU6409594A (en) 1993-03-16 1994-10-11 Alliance Pharmaceutical Corporation Preservation solution and method for warm organ preservation
PT701455E (en) 1993-06-04 2001-09-27 Biotime Inc PLASMA SIMPLE SOLUTION
US5405742A (en) * 1993-07-16 1995-04-11 Cyromedical Sciences, Inc. Solutions for tissue preservation and bloodless surgery and methods using same
WO1995031897A1 (en) 1994-05-20 1995-11-30 Vec Tec, Inc. Method and apparatus monitoring viability of transplantable organs
US5643712A (en) 1994-05-20 1997-07-01 Brasile; Lauren Method for treating and rendering grafts nonthrombogenic and substantially nonimmunogenic using an extracellular matrix coating
AU690144B2 (en) 1994-10-17 1998-04-23 Argutus Intellectual Properties Limited Stabilising medium for alphaGST in urine for use in an enzyme immunoassay
US5554497A (en) 1994-12-12 1996-09-10 Charlotte-Mecklenburg Hospital Authority Cardioplegic solution for arresting an organ
WO1996031778A1 (en) 1995-04-03 1996-10-10 Syncor Intellectual Properties Limited Rapid assays for the detection of glutathione s-transferases
US5679565A (en) 1995-04-10 1997-10-21 The Regents Of The University Of California Method of preserving pancreatic islets
AU710668B2 (en) 1995-12-08 1999-09-23 Biotrin Intellectual Properties Limited Method of determining or detecting donor organ damage following xenotransplantation based on donor-organ derived analytes
ES2262153T3 (en) 1996-02-02 2006-11-16 Biotrin Intellectual Properties Limited METHOD FOR DETERMINING THE HEPATIC STATE OF A LIVER TRANSPLANT RECEPTOR.
US5843024A (en) 1996-05-17 1998-12-01 Breonics, Inc. Solution and process for resuscitation and preparation of ischemically damaged tissue
US5834178C1 (en) * 1997-07-09 2002-04-23 Univ Wayne State Flush-storage solution for donor organs

Also Published As

Publication number Publication date
WO2001054495A1 (en) 2001-08-02
WO2001054495A9 (en) 2002-10-24
EP1257169A1 (en) 2002-11-20
DE60102359D1 (en) 2004-04-22
AU2001231095B2 (en) 2004-07-08
US6492103B1 (en) 2002-12-10
EP1257169B1 (en) 2004-03-17
DE60102359T2 (en) 2005-03-10
US6994954B2 (en) 2006-02-07
AU3109501A (en) 2001-08-07
US20030118980A1 (en) 2003-06-26
JP2003520859A (en) 2003-07-08

Similar Documents

Publication Publication Date Title
EP1257169B1 (en) System for organ and tissue preservation and hypothermic blood substitution
US5145771A (en) Rinse solution for organs and tissues
RU2161405C2 (en) Solutions for transplants of organs and method of transplanting organs
AU680928B2 (en) Solutions for tissue preservation and bloodless surgery and methods using same
US5306711A (en) Organ preservative solution
US4879283A (en) Solution for the preservation of organs
US5702881A (en) Method and solution for organ preservation comprising retinal-derived growth factor, cyclodextrin, mucopolysaccharide and fluorocarbon
US6794124B2 (en) Preservation solution
KR101061293B1 (en) Protective solution for prevention of ischemia
US8802361B2 (en) Composition and method for the restoration and preservation of transplant organs procured from DCD donors
US20020042131A1 (en) Cryopreservation method using cryoprotective composition of propanediol and a vehicle solution
WO1996018293A1 (en) Organ transplant solutions and method for transplanting an organ
Toledo-Pereyra et al. Lung preservation techniques
Jeevanandam et al. University of Wisconsin solution for human donor heart preservation: initial clinical experience
Hachida et al. A new solution (UCLA formula) for lung preservation
JPH0822801B2 (en) Solution for organ preservation
KR100304594B1 (en) Composition for the Preservation of Organs and Blood Cells
De Loecker Hypothermia and preservation of organs in the liquid state
CA1282342C (en) Perfusate for the preservation of organs
Armitage METABOLISM AND PHYSIOLOGY OF CELLS AT LOW TEMPERATURES WJ Armitage
Palmes et al. Pancreas preservation
WO1999045769A2 (en) Preservation solution
JPH05271001A (en) Initial perfusate solution for transplanting organ

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued