METHODS AND SOLUTIONS FOR TISSUE PRESERVATION

Described herein are compositions and methods particularly useful in the medical arts. The compositions and methods may be used in connection with the preservation of a portion of a mammal, for example, tissues, organs, appendages, limbs, extremities, stem cells, myocytes, bone marrow, skeletal muscle as well as an array of other medical procedures, such as cardiac surgery, transplantation and/or preservation. In various embodiments, the inventive composition may be hyperoxygenated and be formulated to resemble the biochemistries of natural intracellular fluids. The inventive composition includes active ingredients to reduce ischemic, hypothermic and reperfusion injury during transplantation, thereby resulting in improved post-transplant graft function and quality, when used in connection with organ transplantation and storage procedures, for example cardiac transplantation.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
FIELD OF THE INVENTION

The invention relates to compositions and methods useful in the medical arts. In particular, the inventive composition may be used as an improved cardiac storage solution in connection with heart transplantation procedures, open-heart surgery, and preservation and/or transplantation of various other portions of a mammal, such as tissues, organs, appendages, limbs, extremities, bone marrow, skeletal muscle, stem cells and myoctyes.

BACKGROUND OF THE INVENTION

All publications herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The following description includes information that may be useful in understanding the present invention. It is not an admission that any of the information provided herein is prier art or relevant to the presently claimed invention, or that any publication specifically or implicitly reference is prior art.

There are approximately 2,500 heart transplants performed every year. A significant obstacle to successful heart transplantation is the limited tolerated ischemia time of the heart to about six hours with currently available preservation methods and solutions. This time frame significantly restricts the size of the geographic region from which a donor organ can transported before the viability of the donor organ for transplantation is compromised. Graft quality also decreases with prolonged storage time, and various studies have shown that suboptimal quality of a heart during transplantation is associated with increased risk of organ rejection in the postoperative period.

A number of transplantation preservation solutions are known in the art See, e.g., P. Michel et al., “A comparative study of the most widely used solutions for cardiac graft preservation during hypothermia,”J. Heart Lung Transplant,. 21(9);1030-39 (2002). More than ten different preservation solutions are already in clinical use. Id, Moreover, there are several different solutions in use that are so-called “Intracellular solutions.”Id. Examples of “intracellular solutions” include University of Wisconsin (e.g., “UW,” “Belzer™” or “Viaspan®”) solution, the most commonly-used formulation, histidine-tryptophan-ketoglutarate (e.g., “HTK,” “Bretschneider's” or “Custodiol®”) solution, Stanford (“STF”) solution, and Eurocollins (“EC”) solution. See, e.g., G. M. Collins, “What solutions are best? Overview of flush solutions,” Transplant, Proc., 29:3543-44 (1997), D. G. Stein et al., “Cardiac preservation in patients undergoing transplantation. A clinical trial comparing University of Wisconsin solution and Stanford solution,” J. Thorac. Cardiovasc. Surg., 102:657-665 (1991), and D. T. Hsu et al., “Quantitative effects of myocardial edema on the left ventricular pressure-volume relation. Influence of cardioplegia osmolarity over two hours of isohemia arrest,” J. Thorac. Cardiovasc. Surg., 106:651-657 (1993).

There is a need in the art for an improved cardiac transplantation solution. In particular, there is a need for a solt.ition that enables the prolonged storage of an organ, such as a heart, for transplantation. By prolonging the tolerable ischemia time of an organ graft, the geographic region from which a donor organ can transported before the viability of the donor organ for transplantation is compromised may be markedly expanded. This, in turn, may improve organ transplantation quality and increase the number of potential donors.

SUMMARY OF INVENTION

This invention relates to methods and compositions to preserve a portion of a mammal, for example, tissues, organs, appendages, limbs, extremities, stem cells, myocytes, bone marrow and skeletal muscle. In various embodiments of the present invention, the methods and compositions may be used to improve the outcome after organ transplantation procedures, such as cardiac transplantation probedures.

In various embodiments the inventive composition may include a hyper-oxygenated solution to preserve the tissues, organs, appendages, limbs, extremities, stem cells, myocytes, bone marrow and skeletal muscle, Further embodiments of the inventive composition may include active ingredients to reduce ischemic, hypothermic and reperfusion injuring during transplantation, thus resulting in improved post-transplant graft function and quality.

In alternate embodiments the inventive composition may be with various tissue types for the preservation and/or transplantation of organs.

Further embodiments include methods to perfuse organs during the process of harvesting the organ and/or transplanting the organ.

Additional embodiments include methods to preserve an appendage, limb and/or extremity after a loss or detachment of the appendage limb and/or extremity.

Other features and advantages of the invention will become apparent from the foliowing detailed description.

DESCRIPTION OF THE INVENTION

All references cited herein are incorporated by reference in their entirety as though fully set forth. Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Singleton et al., Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiiey & Sons (New York, N.Y. 1994); and March, Advanced Organic Chemistry Reactions, Mechanisms and Structure 4th ed., J. Wiley & Sons (New York, N.Y. 1992), provide one skilled in the art with a general guide to many of the terms used in the present application.

One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described. For purposes of the present invention, the following terms are defined below.

“Mammal” as used herein refers to any member of the class Mammalia. including, without limitation, humans and nonhuman primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs, and the like. The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be including within the scope of this term.

“Hyper-oxygenate” water as used herein refers to having an amount of oxygen dissolved in water that is greater than an amount of oxygen that is dissolved in water at standard temperature and atmospheric pressure (“STP”).

“Biochemistry of natural intracellular fluids” as used herein refers to the various electrolyte concentrations of the fluids that surround the target organ, for example, the natural intracellular fluid in cardiac tissue or the natural intracellular fluid in liver tissue, etc.

“Appendage” as used herein refers to a body part or organ that is joined to the trunk of the body. “Limb” as used herein refers to a body part that may be used for locomotion or grasping. “Extremity” as used herein refers to an outlying body part. Examples include but are not limited to an arm, leg, hand, foot, finger toe, ear and nose. It is to be understood that these portions of the body may be classified as any one or more of the aforementioned terms.

The invention relates to compositions and methods useful in the medical arts. In various embodiments, the composition may be used to improve outcome after organ transplantation procedures; in particular, cardiac transplantation procedures. In accordance with various embodiments, the inventive composition includes active ingredients to reduce ischemic, hypothermic and reperfusion injury during transplantation; thereby resulting in improved post-transplant graft function and quality. The inventive composition may also be useful in the storage of myocytes or stem cells for cardiac or other applications. Further application includes the administration of the preservation solution during open-heart surgery, for example, with cardiopulmonary arrest.

In additional embodiments, the inventive composition may be used in connection with medical procedures related to a loss or detachment of an appendage, a limb or an extremity, for example, due to a traumatic injury, cosmetic surgery or reconstructive surgery. In these instances the appendage, limb or extremity, for example, an arm, a leg, a hand, a foot, a finger or a toe, may be placed and preserved in the inventive composition until a surgical reattachment procedure.

In alternate embodiments, the inventive composition may be used with variant tissue types and in connection with different medical procedures. By way of example, the inventive composition may be used in connection with the preservation and/or transplantation of any number of organs, such as kidney, liver, pancreas, eye, cornea, large intestine and small bowel; and with the preservation and/or transplantation of connective tissue such as skin cartilage, and bone. It may also be used as a base solution for lung or pulmonary preservation and/or transplantation, particularly when the inventive composition is supplemented with a protectant for lung surfactant and alveolar surface. The inventive composition may also be used as a perfusate to perfuse an organ during the process of harvesting and/or transplanting.

Further embodiments contemplate the use of the inventive composition for storage of bone marrow and skeletal muscle.

The concept underlying the inventive composition of the present invention is believed to run contrary to current practices in storing hearts for transplantation. Current practices typically involve ischemic (i.e., low oxygen) storage solutions used at hypothermic temperature (i.e., about 4° C.) and formulated at or about physiological pH (i.e., from about 7.2 to about 7.4) for storage of the donor organ. Ischemia is generally intended to minimize injury to cellular structures of the donor organ caused by highly reactive radicals that form in the presence of oxygen, such as superoxide radicals, hydroxyl radicals, nitric oxide radicals, and peroxynitrate radicals. See, e.g., J. J. Lemasters et al., “Reperfusion injury after liver preservation for transplantation,” Ann. Rev. Pharmacol. Toxicol., 37:327-38 (1997).

The inventive composition, on the other hand, includes a significant quantity of dissolved oxygen; indeed, it may be hyperoxygenated. In various embodiments, the inventive composition may exhibit an oxygen tension of from about 490 mmHg to about 590 mmHg. An oxygen tension of about 540 mmHg may be advantageous in accordance with particular embodiments of the present invention. Furthermore, the osmolarity of the inventive composition may be from about 325 mOsm to about 335 mOsm. An osmolarity of about 330 mOsm may be advantageous in accordance with particular embodiments of the present invention. This is believed to be in sharp contrast to currently available transplantation preservation solutions; some are oxygenated, but none are hyperoxygenated. Standard atmospheric oxygen content is less than about 160 mmHg, and under air dissolved oxygen content in aqueous solutions is significantly less; particularly as compared with the range of oxygen content in the inventive composition. See, e.g., U.S. patent publication Nos. 2004/0058432 and 2004/0038192, and Lemasters at 327-38. Moreover, the mere oxygenation of a transplantation solution is not described in the art, except perhaps under very specific circumstances of normothermic (i.e., at about 37° C.) storage without cardioplegia (i.e., without artificially induced heart arrest). See, e.g., U.S. Pat. No. 6,046,046.

Furthermore, the inventive composition may include one or more energy sources capable of being metabolized through the tricarboxylic acid (TCA) cycle; for example, glucose (with or without added insulin), glutamate, glutamine and/or glycine, either alone or in any suitable combination. Furthermore, the inventive composition may have a pH of from about 6.5 to about 7.0, which is markedly lower than currently available solutions. A pH of about 7.0 may be advantageous in accordance with particular embodiments of the present invention. While not wishing to be bound by any theory, it is believed that the maintenance energy provided by the one or more energy sources along with the relatively low pH protect against the loss of cell viability and membrane degradation. As the temperature of the donor organ is decreased from normal body temperature (at or around 37° C.) to the hypothermic temperature at which the donor organ may be maintained in the inventive composition (at or around 4° C.), adenosine triphosphate (ATP) shuttles are slowing, and this the low pH is believed to inactivate proteolytic enzymes that would otherwise degrade membrane proteins at these lower temperatures (i.e., temperatures below about 20° C.).

The inventive composition may include glucose at a concentration of from about 19 mmol/L to about 21 mmol/L. A concentration of glucose of about 20 mmol/L may be advantageous in accordance with particular embodiments of the present invention. The inventive composition may include insulin a concentration of from about 45 U/L to about 60 U/L. A concentration of insulin of from about 49 U/L to about 51 U/L may be advantageous in accordance with particular embodiments of the present invention. The inventive composition may include glutamine at a concentration of from about 1.7 mmol/L to about 3.0 mmol/L. A concentration of glutamine of from about 1.9 mmol/L to about 2.1 mmol/L may be advantageous in accordance with particular embodiments of the present invention. The inventive composition may include glutamate at a concentration of from about 0.8 mmol/L to about 1.5 mmol/L. A concentration of glutamate of from about 0.9 mmol/L to about 1.1 mmol/L may be advantageous in accordance with particular embodiments of the present invention. The inventive composition may include glycine at a concentration of from about 9.5 mmol/L to about 11.0 mmol/L. A concentration of glycine of from about 9.8 mmol/L to about 10.2 mmol/L may be advantageous in accordance with particular embodiments of the present invention.

Additionally, the inventive composition may include one or more radical scavengers and/or reductants to protect cellular structures against oxidative damage from radicals. By way of example, the inventive composition may include allopurinol and/or glutathione, either alone or in combination. Again, while not wishing to be bound by any particular theory, it is believed that the presence of one or more energy sources and the maintenance of aerobe metabolic activity in the presence of the oxygen that is dissolved in the inventive composition preserves the metabolic integrity of the cells of the donor organ and minimizes the generation of, and injury caused by, oxygen radicals to the same.

The inventive composition may include allopurinol at a concentration of from about 0.8 mmol/L to about 1.2 mmol/L. A concentration of allopurinol of about 1.0 mmol/L may be advantageous in accordance with particular embodiments of the present invention. The inventive composition may include glutathione at a concentration of from about 2.7 mmol/L to about 3.6 mmol/L. A concentration of glutathione of from about 2.9 mmol/L to about 3.2 mmol/L may be advantageous in accordance with particular embodiments of the present invention.

In various embodiments, the inventive composition may include active ingredients at an essential or near essential minimum. The use of active ingredients at an essential or near essential minimum aids in reducing the cost and complexity of manufacturing, yet the composition demonstrates superior function as compared to currently available products, as illustrated by the Examples herein described. It also imparts a relatively lower viscosity to the composition as compared with commercially available compositions typically used in connection with cardiac transplantation procedures. The relative viscosity of the inventive composition may be less than about 1.5 (relative to water). Relative viscosities of from about 1.0 to about 1.2 (relative to water) may be advantageous in accordance with particular embodiments of the present invention. Furthermore, as compared with currently available products, the inventive composition need not include calcium channel blockers, steroids and/or vitamins; although in alternate embodiments of the present invention, such items may be included. Again, while not wishing to be bound by any particular theory, it is believed that the low viscosity of the inventive composition combined with the osmolarity of from about 325 mOsm to about 336 mOsm, prevents osmotic stress on the cells, keeps the oxygen tension in the solution relatively high and more uniform, and allows the quicker introduction and removal with faster onset of organ protection as compared with the use of currently available transplantation solutions.

The inventive composition may also include various electrolytes, which may be formulated so as to resemble the biochemistry of natural intracellular fluids. Representative electrolytes include sodium chloride (NaCl), potassium chloride (KCl), magnesium sulfate (MgSO4), and phosphate (KH2PO4/K2HPO4). The electrolytes may be present in the inventive composition at concentrations that approximate their respective concentrations in the natural intracellular fluid that the inventive composition is intended to mimic; for example, intracellular fluid in cardiac tissue in those embodiments of the instant invention wherein the inventive composition is for use in connection with cardiac transplantation or other procedures involving cardiac tissue (e.g., open heart surgery with cardiac arrest, cardiac arterial bypass graft surgery, etc.). Particularly for embodiments of the instant invention involving use of the inventive composition in cardiac procedures, the electrolytes may include sodium chloride (NaCl) at a concentration of from about 3.75 mmol/L to about 4.50 mmol/L; potassium chloride (KCl) at a concentration of from about 115 mmol/L to about 125 mmol/L; magnesium sulfate (MgSO4) at a concentration of from about 2.1 mmol/L to about 2.8 mmol/L; and phosphate (KH2PO4/K2HPO4) at a concentration of from about 3.5 mmol/L to about 4.0 mmol/L. Concentrations of about 4.00 mmol/L of sodium chloride (NaCl), about 120 mmol/L of potassium chloride (KCl), about 2.4 mmol/L of magnesium sulfate (MgSO4), and about 3.7 mmol/L of phosphate (KH2PO4/K2HPO4) may be advantageous in accordance with particular embodiments of the present invention. The respective concentrations of sodium chloride (NaCl) and potassium (KH2PO4/K2HPO4) cations, in particular, resemble intracellular concentrations, as opposed to extracellular concentrations, which are essentially the reverse. Therefore, the inventive composition falls within the category of so-called “intracellular solutions,” as opposed to so-called “extracellular solutions.” Once more, while not wishing to be bound by any particular theory, it is believed that this formulation prevents the physiological disruption of the cell membrane and allows the quicker introduction and removal with faster onset of organ protection.

The inventive composition may include adenosine, which may be present at a concentration of from about 2.5 mmol/L to about 4.0 mmol/L. Concentrations of adenosine of from about 2.9 mmol/L to about 3.1 mmol/L may be advantageous in accordance with particular embodiments of the present invention.

The inventive composition may include a buffer, such as HEPES (4-2-hydroxyethyl-1-piperazineethanesulfonic acid), an organic chemical buffer, which may be present at a concentration of from about 19 mmol/L to about 21 mmol/L. A concentration of HEPES of about 20 mmol/L may be advantageous in accordance with particular embodiments of the present invention.

EXAMPLES

The following examples demonstrate the preparation, use and efficacy of the inventive composition in connection with cardiac transplantation and preservation.

Example 1 Preparation of Cardiac Transplantation Preservation Solution

The cardiac transplantation solution is prepared by mixing each component thereof in distilled water. The media undergoes mechanical filter sterilization, oxygenation with 95% O2 and 5% CO2, pH and osmolarity adjustment

Example 2 Harvest of Donor Organs from Experimental Animals

Nine rats in each experimental group underwent a standard method of harvesting the heart for transplantation. Briefly, each animal was anesthetized and the chest was opened. The blood vessels leading to the heart were clamped and the heart was arrested (i.e., cardioplegia) with a perfusion of cold solution. The heart was removed and placed in cold (4° C.) solution for preservation. Death occurred in the anesthetized donor animal once the heart was removed.

Example 3 Comparative Study of Donor Organ Function in Transplantation Preservation Solution

The rat hearts were divided into groups based on different storage solutions and the length of cold preservation (i.e., 6, 8 or 10 hours of storage). Following the storage period, the hearts were attached to a Langendorff perfusion system, and physiological measurements of heart function were conducted over a period of 20 minutes. The results are illustrated in Table 1, below.

TABLE 1 Comparative Study of Inventive Composition and UW Solution Inventive UW Composition Measurements Mean ± SD Mean ± SD p < 0.05 Period 1 (0-5 minutes) HR1 170.7 ± 107.6 154.7 ± 72.3  Max-P1 14.5 ± 4.6  19.5 ± 9.0  EDP1 9.8 ± 3.1 8.2 ± 2.0 dP/dt1 241.9 ± 97.5  469.7 ± 297.3 X NdP/dt1 196.9 ± 57.7  398.0 ± 305.4 X PG1 4.8 ± 3.1 11.2 ± 8.5  X EF1 794.1 ± 681.5 1573.6 ± 1154.9 X Period 2 (5-10 minutes) HR2 270.0 ± 278.5 143.9 ± 61.2  Max-P2 14.5 ± 5.0  16.4 ± 5.2  EDP2 10.8 ± 3.9  8.1 ± 2.5 X dP/dt2 198.8 ± 56.9  337.7 ± 169.9 X NdP/dt2 166.4 ± 45.7  272.6 ± 115.8 X PG2 3.7 ± 2.5 8.3 ± 5.0 X EF2 720.7 ± 540.2 1170.2 ± 847.6  Period 3 (10-15 minutes) HR3 234.9 ± 208.6 164.9 ± 46.5  Max-P3 14.8 ± 5.7  14.6 ± 4.4  EDP3 10.3 ± 4.2  6.6 ± 3.2 X dP/dt3 205.0 ± 52.8  363.9 ± 180.4 X NdP/dt3 171.6 ± 36.7  318.6 ± 183.8 X PG3 4.6 ± 3.1 8.0 ± 4.7 X EF3 716.2 ± 361.5 1317.2 ± 881.7  X Period 4 (15-20 minutes) HR4 193.1 ± 132.1 179.3 ± 59.4  Max-P4 15.1 ± 6.5  15.9 ± 5.6  EDP4 9.1 ± 4.5 6.4 ± 3.4 X dP/dt4 237.4 ± 73.0  433.7 ± 201.2 X NdP/dt4 188.2 ± 50.8  383.1 ± 275.2 X PG4 6.0 ± 4.5 9.7 ± 5.7 EF4 830.9 ± 522.8 1783.0 ± 1345.3 X Period t (0-20 minutes) HR_t 868.7 ± 698.2 642.8 ± 203.8 Max-P_t 59.0 ± 19.5 66.3 ± 21.0 EDP_t 39.9 ± 14.4 29.2 ± 9.6  X dP/dt_t 883.1 ± 208.3 1605.0 ± 671.3  X NdP/dt_t 723.0 ± 150.8 1372.4 ± 802.9  X PG_t 19.1 ± 11.1 37.2 ± 20.0 X EF_t 3061.9 ± 1781.2 5843.9 ± 3509.1 X HR = heart rate (beats/min) Max-P = maximum pressure (mmHg) EDP = end diastolic pressure (mmHg) dP/dt = maximum rate of pressure rise (mmHg/sec) NdP/dt = maximum rate of pressure decline (mmHg/sec) PG = pressure gradient (Max-P − EDP) (mmHg) EF = ejection fraction (HR × PG)

As shown in Table 1, use of the inventive composition demonstrated significant (i.e., about 70% to about 130%) improvement over the UW solution; storage in the inventive composition resulted in greater ejection fraction (EF) from the left ventricle and greater pressure upslope (dP/dt) and pressure downslope (NdP/dt), compared to storage in University of Wisconsin (UW) solution. The improvements in EF, dP/dt and NdP/dt indicate greater strength and elasticity of the stored heart muscle using the inventive composition.

While the description above refers to particular embodiments of the present invention, it should be readily apparent to people of ordinary skill in the art that a number of modifications may be made without departing from the spirit thereof. The accompanying claims are intended to cover such modifications as would fall within the true spirit and scope of the invention. The presently disclosed embodiments are, therefore, to be considered in all respects as illustrative and not restrictive, the scope of the invention being indicated by the appended claims rather than the foregoing description. All changes that come within the meaning of and range of equivalency of the claims are intended to be embraced therein.

Claims

1. A method of preserving a portion of a mammal, comprising:

placing the portion of the mammal in contact with a preservation solution of claim 17, whereby the portion of the mammal is preserved, comprising: a quantity of water, and a quantity of dissolved oxygen,
wherein the quantity of dissolved oxygen is sufficient to hyper-oxygenate the water.

2-14. (canceled)

15. A method of preserving a myocyte and/or a stem cell, comprising:

placing the myocyte and/or stem cell in contact with a preservation solution of claim 17, whereby the myocyte and/or stem cell is preserved, comprising: a quantity of water, and a quantity of dissolved oxygen,
wherein the quantity of dissolved oxygen is sufficient to hyper-oxygenate the water.

16. (canceled)

17. A tissue preservation solution adapted to preserve a portion of a mammal, comprising:

a quantity of water;
a quantity of dissolved oxygen,
wherein the quantity of dissolved oxygen is sufficient to hyper-oxygenate the water; and
an energy source capable of being metabolized through the tricarboxylic acid cycle.

18. The solution of claim 17, wherein the quantity of dissolved oxygen produces an oxygen tension of from about 490 mmHg to about 590 mmHg.

19. (canceled)

20. The solution of claim 17, wherein the energy source is selected from the group consisting of a quantity of glucose, a quantity of glutamate, a quantity of glutamine, a quantity of glycine and combinations thereof.

21. The solution of claim 20, wherein the quantity of glucose is from about 19 mmol/L to about 21 mmol/L.

22. The solution of claim 20, wherein the quantity of glutamate is from about 0.8 mmol/L to about 1.5 mmol/L.

23. The solution of claim 20, where in the quantity of glutamine is from about 1.7 mmol/L to about 3.0 mmol/L.

24. The solution of claim 20, wherein the quantity of glycine is from about 9.5 mmol/L to about 11.0 mmol/L.

25. The solution of claim 17, wherein the energy source is a quantity of glucose, and the solution further comprises a quantity of insulin.

26. The solution of claim 25, wherein the quantity of glucose is from about 19 mmol/L to about 21 mmol/L and the quantity of insulin is from about 49 U/L to about 51 U/L.

27. The solution of claim 17, further comprising a reductant.

28. The solution of claim 27, wherein the reductant is selected from the group consisting of allopurinol, glutathione and combinations thereof.

29. The solution of claim 28, wherein the allopurinol is present in an amount of from about 0.8 mmol/L to about 1.2 mmol/L.

30. The solution of claim 28, wherein the glutathione is present in an amount of from about 2.7 mmol/L to about 3.6 mmol/L.

31. The solution of claim 17, further comprising an electrolyte.

32. The solution of claim 31, wherein the electrolyte is formulated to resemble the biochemistry of a natural intracellular fluid.

33. The solution of claim 31, wherein the electrolyte is selected from the group consisting of sodium chloride (NaCl), potassium chloride (KCl), magnesium sulfate (MgSO4), potassium phosphate (KH2PO4/K2HPO4), and combinations thereof.

34. The solution of claim 33, wherein the NaCl is present in an amount of from about 3.75 mmol/L to about 4.50 mmol/L.

35. The solution of claim 33, wherein the KCl is present in an amount of from about 115 mmol/L to about 125 mmol/L.

36. The solution of claim 33, wherein the MgSO4 is present in an amount of from about 2.1 mmol/L to about 2.8 mmol/L.

37. The solution of claim 33, wherein the KH2PO4/K2HPO4 is present in an amount of from about 3.5 mmol/L to about 4.0 mmol/L.

38. The solution of claim 17, wherein the solution has an osmolarity of from about 325 mOsm to about 335 mOsm.

39. The solution of claim 17, wherein the solution has a pH of from about 6.5 to about 7.0.

40. The solution of claim 17, wherein the solution has a viscosity relative to water that is less than 1.5.

41. The solution of claim 17, further comprising a quantity of adenosine.

42. The solution of claim 41, wherein the quantity of adenosine is from about 2.5 mmol/L to about 4.0 mmol/L.

43. The solution of claim 17, further comprising a quantity of a buffer.

44. The solution of claim 43, wherein the buffer is 4-2-hydroxyethyl-1-piperazineethanesulfonic acid, present in an amount from about 20 mmol/L to about 21 mmol/L.

45. A tissue preservation solution comprising:

a quantity of water;
a quantity of dissolved oxygen;
an energy source capable of being metabolized through the tricarboxylic acid cycle;
a reductant; and
an electrolyte formulated to resemble the biochemistry of a natural intracellular fluid,
wherein the quantity of dissolved oxygen produces an oxygen tension of about 490 mmHg to about 590 mmHg, the osmolarity is from about 325 mOsm to about 335 mOsm, and the pH is from about 6.5 to 7.0.
Patent History
Publication number: 20150111193
Type: Application
Filed: Oct 31, 2014
Publication Date: Apr 23, 2015
Applicant: CEDARS-SINAI MEDICAL CENTER (Los Angeles, CA)
Inventors: Andreas Kamlot (Northridge, CA), Alfredo Trento (Malibu, CA), Lawrence Czer (Santa Monica, CA), Jennifer Pass (Gaithersburg, MD)
Application Number: 14/530,006