SYSTEMS, METHODS, COMPOSITIONS AND SOLUTIONS FOR PERFUSING AN ORGAN

- TransMedics, Inc.

The disclosure, in various embodiments, provides systems, methods, and solutions for perfusing an organ. The present application describes illustrative methods for perfusing an ex-vivo organ in a functioning state under physiological or near physiological conditions, including receiving an ex-vivo organ in an organ care system, the ex-vivo organ coupled to a perfusion circuit, infusing a solution into the system, combining the solution with a perfusion fluid to create a mixed solution, pumping the mixed solution to the ex-vivo organ, and receiving the mixed solution from the ex-vivo organ.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 16/053,206, filed Aug. 2, 2018, which is a continuation of U.S. patent application Ser. No. 14/734,769, filed Jun. 9, 2015, now U.S. Pat. No. 10,039,276, which is a divisional of U.S. patent application Ser. No. 11/246,919, filed Oct. 7, 2005, now U.S. Pat. No. 9,078,428, and claims priority to U.S. Provisional Patent Application No. 60/725,168, filed Oct. 6, 2005, entitled “Systems and Methods for Ex-Vivo Organ Care,” and U.S. Provisional Patent Application No. 60/694,971, filed Jun. 28, 2005, entitled “Systems and Methods for Ex-Vivo Organ Care.” The entire contents of each of the above is incorporated herein by reference.

FIELD

The invention relates generally to organ perfusion. In various illustrated embodiments the invention provides for systems, methods, compositions and solutions for perfusing an organ.

BACKGROUND

Many patients need organ transplants but are unable to obtain suitable organs. One of the primary reasons is that organs that are otherwise viable when harvested from a donor are not preserved for long enough periods of time to allow them to be transported to appropriate recipients. With current preservation techniques, transplantable organs remain viable for about three to four hours and, beyond that time, suffer ischemia and tissue injury, which renders them unviable for transplant.

Current preservation techniques include cryopreservative methods, which involve the cooling of the transplantable organ to temperatures well below physiological temperatures (e.g., below 25° C.). Such techniques typically use preservation solutions that do not replenish energy sources within the organ during transplant or maintain the organ in a functioning state and therefore are largely ineffective in preventing ischemia and other injuries to the organ. The solutions also often rely on the use of high molecular weight impermeants to maintain the organ at these temperatures prior to transplantation, and such components are in many cases harmful to the organ.

Although improvements to solutions have been made over the last several years, particularly as disclosed in U.S. Pat. Nos. 6,100,082, 6,046,046, and PCT Application No. PCT/US98/19912, further improvements are still needed.

SUMMARY

The invention addresses deficiencies in the art by providing, in various embodiments, improved systems, methods, compositions and solutions for perfusing an organ prior to transplantation. According to one aspect, the solutions include components adapted to preserving and maintaining a harvested organ ex-vivo in its functioning state under physiologic or near physiologic conditions prior to its transplantation, while minimizing reperfusion injury and ischemia to the organ while awaiting transplantation.

According to another feature, the solutions include energy sources for the organ while it is being prepared for and undergoing transplantation. Energy sources include, for example, one or more of carbohydrate sources, components for synthesis of energy-rich molecules, such as phosphate sources, and other components. According to one embodiment, the solutions provide for amino acids for assisting the organ in cellular protein synthesis during perfusion. According to one embodiment, the solutions also provide components that help maintain the organ's normal functionality during perfusion, such as cardio stimulants, insulin and other hormones, electrolytes, etc. The solutions may also be adapted to include drugs or other therapeutics for preservation of the organ and/or patient care.

In another aspect, the invention provides for systems and methods for improved preservation. In certain embodiments the solutions are combined with blood products for perfusion. The solutions, systems and methods are also adaptable to drug delivery systems. In certain embodiments, the solutions are combined with additional additives at the point of use.

In one embodiment, a composition for use in a solution for perfusing an organ, such as a heart, is provided comprising one or more carbohydrates, one or more organ stimulants, and a plurality of amino acids that do not include asparagine, glutamine, or cysteine.

In another embodiment, a system for perfusing an organ, such as a heart, is provided comprising an organ and a substantially cell-free composition, where the composition comprises one or more carbohydrates, one or more organ stimulants, and a plurality of amino acids that do not include asparagine, glutamine, or cysteine.

BRIEF DESCRIPTION OF THE FIGURES

The foregoing and other objects and advantages of the invention will be appreciated more fully from the following further description thereof, with reference to the accompanying figures wherein:

FIG. 1 depicts an embodiment of a perfusion system using two solutions for maintaining a heart.

FIG. 2 depicts an embodiment of a perfusion system using two solutions and also a priming solution.

FIG. 3 depicts an embodiment of a perfusion system using two solutions, a priming solution, and a composition or solution having sensitive material added prior to perfusion.

FIG. 4 depicts a chart demonstrating electrolyte stability for an organ undergoing perfusion according to an embodiment of the invention.

FIG. 5 depicts a chart demonstrating electrolyte stability for an organ undergoing perfusion according to an embodiment of the invention.

FIG. 6 depicts a chart demonstrating the arterial blood gas profile for an organ undergoing perfusion according to an embodiment of the invention.

DETAILED DESCRIPTION

The invention addresses deficiencies in the prior art by providing in various illustrated embodiments systems, methods, compositions, and solutions for maintaining an organ in a functioning state ex vivo under physiological or near physiological conditions. In one embodiment, the organ is a heart and is maintained in a beating state at a physiologic or near physiologic temperature during perfusion of a perfusion fluid, which may include one or more of the solutions described herein. According to certain embodiments, solutions with particular solutes and concentrations are selected and proportioned to enable the organ to function at physiologic or near physiologic conditions. For example, such conditions include maintaining organ function at or near a physiological temperature and/or preserving an organ in a state that permits normal cellular metabolism, such as protein synthesis.

In certain embodiments, solutions are formed from compositions by combining components with a fluid, from more concentrated solutions by dilution, or from more dilute solutions by concentration. In exemplary embodiments, suitable solutions include one or more energy sources, one or more stimulants to assist the organ in continuing its normal physiologic function prior to and during transplantation, and one or more amino acids selected and proportioned so that the organ continues its cellular metabolism during perfusion. Cellular metabolism includes, for example, conducting protein synthesis while functioning during perfusion. Some illustrative solutions are aqueous based, while other illustrative solutions are non-aqueous, for example organic solvent-based, ionic-liquid-based, or fatty-acid-based.

The solutions may include one or more energy-rich components to assist the organ in conducting its normal physiologic function. These components may include energy rich materials that are metabolizable, and/or components of such materials that an organ can use to synthesize energy sources during perfusion. Exemplary sources of energy-rich molecules include, for example, one or more carbohydrates. Examples of carbohydrates include monosaccharides, disaccharides, oligosaccharides, polysaccharides, or combinations thereof, or precursors or metabolites thereof. While not meant to be limiting, examples of monosaccharides suitable for the solutions include octoses; heptoses; hexoses, such as fructose, allose, altrose, glucose, mannose, gulose, idose, galactose, and talose; pentoses such as ribose, arabinose, xylose, and lyxose; tetroses such as erythrose and threose; and trioses such as glyceraldehyde. While not meant to be limiting, examples of disaccharides suitable for the solutions include (+)-maltose (4-O-(α-D-glucopyranosyl)-α-D-glucopyranose), (+)-cellobiose (4-O-(β-D-glucopyranosyl)-D-glucopyranose), (+)-lactose (4-O-(β-D-galactopyranosyl-β-D-glucopyranose), sucrose (2-O-(α-D-glucopyranosyl-β-D-fructofuranoside). While not meant to be limiting, examples of polysaccharides suitable for the solutions include cellulose, starch, amylose, amylopectin, sulfomucopolysaccharides (such as dermatane sulfate, chondroitin sulfate, sulodexide, mesoglycans, heparan sulfates, idosanes, heparins and heparinoids), and glycogen. In some embodiments, monossacharides, disaccharides, and polysaccharides of both aldoses, ketoses, or a combination thereof are used. One or more isomers, including enantiomers, diastereomers, and/or tautomers of monosaccharides, disaccharides, and/or polysaccharides, including those described and not described herein, may be employed in the solutions described herein. In some embodiments, one or more monosaccharides, disaccharides, and/or polysaccharides may have been chemically modified, for example, by derivatization and/or protection (with protecting groups) of one or more functional groups. In certain embodiments, carbohydrates, such as dextrose or other forms of glucose, are preferred.

Other possible energy sources include adenosine triphosphate (ATP), co-enzyme A, pyruvate, flavin adenine dinucleotide (FAD), thiamine pyrophosphate chloride (co-carboxylase), β-nicotinamide adenine dinucleotide (NAD), β-nicotinamide adenine dinucleotide phosphate (NADPH), and phosphate derivatives of nucleosides, e.g., nucleotides, including mono-, di-, and tri-phosphates (e.g., UTP, GTP, GDP, and UDP), coenzymes, or other bio-molecules having similar cellular metabolic functions, and/or metabolites or precursors thereof. For example, phosphate derivatives of adenosine, guanosine, thymidine (5-Me-uridine), cytidine, and uridine, as well as other naturally and chemically modified nucleosides are contemplated.

In certain embodiments, one or more carbohydrates is provided along with a phosphate source, such as a nucleotide. The carbohydrate helps enable the organ to produce ATP or other energy sources during perfusion. The phosphate source may be provided directly through ATP, ADP, AMP, or other sources. In other illustrative embodiments, a phosphate is provided through a phosphate salt, such as glycerophosphate, sodium phosphate or other phosphate ions. A phosphate may include any form thereof in any ionic state, including protonated forms and forms with one or more counter ions.

The solutions may include one or more organ stimulants for assisting the organ's normal physiologic function during perfusion. In some illustrative embodiments, where the transplanted organ is a heart, cardio stimulants are provided to enable the heart to continue functioning (e.g., continue beating) during perfusion and transplantation. Such stimulants may include, for example, catecholamines, such as epinephrine and/or norepinephrine, which facilitate beating of the heart. Other cardio stimulants may be used, such as certain forms of peptides and/or polypeptides (e.g., vasopressin, Anthropleurin-A and Anthropleurin-B), and/or β1/β2 —adrenoreceptor blocking agents (such as CGP 12177), bupranolol, pindolol, alprenolol, and cardiac glycosides. One or more natural products may also be used, such as digitalis (digoxin), palustrin, and/or ferulic acid. Stimulants such as those mentioned above can be included with the solutions or added at the point of use by the user.

In some instances, additional components are provided to assist the organ in conducting its metabolism during perfusion. These components include, for example, forms or derivatives of adenine and/or adenosine, which may be used for ATP synthesis, for maintaining endothelial function, and/or for attenuating ischemia and/or reperfusion injury. According to certain implementations, a magnesium ion source is provided with a phosphate, and in certain embodiments, with adenosine to further enhance ATP synthesis within the cells of the perfused organ.

As noted, the solution may include one or more amino acids, preferably a plurality of amino acids, to support protein synthesis by the organ's cells. Suitable amino acids include, for example, any of the naturally-occurring amino acids. The amino acids may be in various enantiomeric or diastereomeric forms. For example, solutions may employ either D- or L-amino acids, or a combination thereof, e.g., solutions enantioenriched in more of the D- or L-isomer or racemic solutions. Suitable amino acids may also be non-naturally occurring or modified amino acids, such as citrulline, ornithine, homocysteine, homoserine, β-amino acids such as β-alanine, amino-caproic acid, or combinations thereof.

Certain exemplary solutions include some but not all naturally-occurring amino acids. In some embodiments, solutions include essential amino acids. For example, a solution may be prepared with one or more or all of the following amino acids:

Glycine Alanine Arginine Aspartic Acid Glutamic Acid Histidine Isoleucine Leucine Methionine Phenylalanine Proline Serine Threonine Tryptophan Tyrosine Valine Lysine acetate

In certain embodiments, non-essential and/or semi-essential amino acids are not included in the solutions. For example, in some embodiments, asparagine, glutamine, and/or cysteine are not included. In other embodiments, the solution contains one or more non-essential and/or semi-essential amino acids. Accordingly, in other embodiments, asparagine, glutamine, and/or cysteine are included.

The solutions may also contain electrolytes, particularly calcium ions for facilitating enzymatic reactions, cardiac contractility, and/or coagulation within the organ. Other electrolytes may be used, such as sodium, potassium, chloride, sulfate, magnesium and other inorganic and organic charged species or combinations thereof. It should be noted that any component provided hereunder may be provided, where valence and stability permit, in an ionic form, in a protonated or unprotonated form, in salt or free base form, or as ionic or covalent substituents in combination with other components that hydrolyze and make the component available in aqueous solutions, as suitable and appropriate.

In certain embodiments, the solutions contain buffering components. For example, suitable buffer systems include 2-morpholinoethanesulfonic acid monohydrate (IVIES), cacodylic acid, H2CO3/NaHCO3 (pKa1), citric acid (pKa3), bis(2-hydroxyethyl)-imino-tris-(hydroxymethyl)-methane (Bis-Tris), N-(carbamoylmethyl)iminodiacetic acid (ADA), 3-bis[tris(hydroxymethyl)methylamino]propane (Bis-Tris Propane) (pKa1), piperazine-1,4-bis(2-ethanesulfonic acid) (PIPES), N-(2-Acetamido)-2-aminoethanesulfonic acid (ACES), imidazole, N,N-bis(2-hydroxyethyl)-2-aminoethanesulfonic acid (BES), 3-(N-morpholino)propanesulphonic acid (MOPS), NaH2PO4/Na2HPO4 (pKa2), N-tris(hydroxymethyl)methyl-2-aminoethanesulfonic acid (TES), N-(2-hydroxyethyl)-piperazine-N′-2-ethanesulfonic acid (HEPES), N-(2-hydroxyethyl)piperazine-N′-(2-hydroxypropanesulfonic acid) (HEPPSO), triethanolamine, N-[tris(hydroxymethyl)methyl]glycine (Tricine), tris(hydroxymethyl)aminoethane (Tris), glycinamide, N,N-bis(2-hydroxyethyl) glycine (Bicine), glycylglycine (pKa2a2), N-tris(hydroxymethyl)methyl-3-aminopropanesulfonic acid (TAPS), or a combination thereof. In some embodiments, the solutions contain sodium bicarbonate, potassium phosphate, or TRIS buffer.

The solutions may include other components to help maintain the organ and protect it against ischemia, reperfusion injury and other ill effects during perfusion. In certain exemplary embodiments, these components may include hormones (e.g., insulin), vitamins (e.g., an adult multi-vitamin, such as multi-vitamin MVI-Adult™), and/or steroids (e.g., dexamethasone and SoluMedrol™).

In another aspect, a blood product is provided with the solution to support the organ during metabolism. Exemplary suitable blood products may include whole blood and/or one or more components thereof such as blood serum, plasma, albumin, and red blood cells. In embodiments where whole blood is used, the blood may be passed through a leukocyte and platelet depleting filter to reduce pyrogens, antibodies and/or other items that may cause inflammation in the organ. Thus, in some embodiments, the solution employs whole blood that has been at least partially depleted of leukocytes and/or whole blood that has been at least partially depleted of platelets.

The solutions are preferably provided at a physiological temperature and maintained thereabout throughout perfusion and recirculation. As used herein, “physiological temperature” is referred to as temperatures between about 25° C. and about 37° C., for example, between about 30° C. and about 37° C., such as between about 34° C. and about 37° C.

Table 1 sets forth components that may be used in a preservative solution for preserving an organ as described herein.

TABLE 1 Component of Exemplary for a Preservative Solution Exemplary Concentration Component Ranges in a preservative solution Alanine about 1 mg/L-about 10 g/L Arginine about 1 mg/L-about 10 g/L Asparagine about 1 mg/L-about 10 g/L Aspartic Acid about 1 mg/L-about 10 g/L Cysteine about 1 mg/L-about 10 g/L Cystine about 1 mg/L-about 10 g/L Glutamic Acid about 1 mg/L-about 10 g/L Glutamine about 1 mg/L-about 10 g/L Glycine about 1 mg/L-about 10 g/L Histidine about 1 mg/L-about 10 g/L Hydroxyproline about 1 mg/L-about 10 g/L Isoleucine about 1 mg/L-about 10 g/L Leucine about 1 mg/L-about 10 g/L Lysine about 1 mg/L-about 10 g/L Methionine about 1 mg/L-about 10 g/L Phenylalanine about 1 mg/L-about 10 g/L Proline about 1 mg/L-about 10 g/L Serine about 1 mg/L-about 10 g/L Threonine about 1 mg/L-about 10 g/L Tryptophan about 1 mg/L-about 10 g/L Tyrosine about 1 mg/L-about 10 g/L Valine about 1 mg/L-about 10 g/L Adenine about 1 mg/L-about 10 g/L ATP  about 10 ug/L-about 100 g/L Adenylic Acid  about 10 ug/L-about 100 g/L ADP  about 10 ug/L-about 100 g/L AMP  about 10 ug/L-about 100 g/L Ascorbic Acid  about 1 ug/L-about 10 g/L D-Biotin  about 1 ug/L-about 10 g/L Vitamin D-12  about 1 ug/L-about 10 g/L Cholesterol  about 1 ug/L-about 10 g/L Dextrose (Glucose)   about 1 g/L-about 150 g/L Multi-vitamin Adult        about 1 mg/L-about 20 mg/L or 1 unit vial Epinephrine about 1 ug/L-about 1 g/L  Folic Acid about 1 ug/L-about 10 g/L Glutathione about 1 ug/L-about 10 g/L Guanine about 1 ug/L-about 10 g/L Inositol  about 1 g/L-about 100 g/L Riboflavin about 1 ug/L-about 10 g/L Ribose about 1 ug/L-about 10 g/L Thiamine about 1 mg/L-about 10 g/L Uracil about 1 mg/L-about 10 g/L Calcium Chloride  about 1 mg/L-about 100 g/L NaHCO3  about 1 mg/L-about 100 g/L Magnesium sulfate  about 1 mg/L-about 100 g/L Potassium chloride  about 1 mg/L-about 100 g/L Sodium glycerophosphate  about 1 mg/L-about 100 g/L Sodium Chloride  about 1 mg/L-about 100 g/L Sodium Phosphate  about 1 mg/L-about 100 g/L Insulin   about 1 IU-about 150 IU Serum albumin   about 1 g/L-about 100 g/L Pyruvate  about 1 mg/L-about 100 g/L Coenzyme A  about 1 ug/L-about 10 g/L Serum   about 1 ml/L-about 100 ml/L Heparin   about 500 U/L-about 1500 U/L SoluMedrol ™  about 200 mg/L-about 500 mg/L Dexamethasone about 1 mg/L-about 1 g/L  FAD  about 1 ug/L-about 10 g/L NADP  about 1 ug/L-about 10 g/L adenosine  about 1 mg/L-about 10 g/L guanosine  about 1 mg/L-about 10 g/L GTP  about 10 ug/L-about 100 g/L GDP  about 10 ug/L-about 100 g/L GMP  about 10 ug/L-about 100 g/L

The solutions described herein may be prepared from compositions having one or more components such as those described above in concentrations such as those described above. An exemplary embodiment of a composition adaptable for use in organ perfusion includes one or more components selected from Table 2. The amounts provided describe preferred amounts relative to other components in the table and may be scaled to provide compositions of sufficient quantity. In some embodiments, the amounts listed in Table 2 can vary by±about 10% and still be used in the solutions described herein.

TABLE 2 Component of Exemplary Composition for a Preservative Solution Component Amount Adenosine about 675 mg-about 825 mg Calcium Chloride dihydrate about 2100 mg-about 2600 mg Glycine about 315 mg-about 385 mg L-Alanine about 150 mg-about 200 mg L-Arginine about 600 mg-about 800 mg L-Aspartic Acid about 220 mg-about 270 mg L-Glutamic Acid about 230 mg about 290 mg L-Histidine about 200 mg-about 250 mg L-Isoleucine about 100 mg about 130 mg L-Leucine about 300 mg-about 380 mg L-Methionine about 50 mg-about 65 mg L-Phenylalanine about 45 mg-about 60 mg L-Proline about 110 mg-about 140 mg L-Serine  about 80 mg-about 105 mg L-Threonine about 60 mg-about 80 mg L-Tryptophan about 30 mg-about 40 mg L-Tyrosine  about 80 mg-about 110 mg L-Valine about 150 mg-about 190 mg Lysine Acetate about 200 mg-about 250 mg Magnesium Sulfate about 350 mg-about 450 mg Heptahydrate Potassium Chloride about 15 mg-about 25 mg Sodium Chloride about 1500 mg-about 2000 mg Dextrose  about 25 g-about 120 g Epinephrine about 0.25 mg-about 1.0 mg  Insulin  about 75 Units-about 150 Units MVI-Adult ™ 1 unit vial SoluMedrol ™ about 200 mg-about 500 mg Sodium Bicarbonate    about 10-about 25 mEq

A solution may be prepared combining components selected by a user with an aqueous fluid to form a preservative solution. In an illustrative embodiment, one or more components from Table 2 are combined in the relative amounts listed therein per about 1000 mL of aqueous fluid to form a preservative solution for perfusing an organ, such as a heart. In some embodiments the quantity of aqueous fluid can vary±about 10%.

In another aspect, solutions may be administered in combinations as desired by a medical operator. FIG. 1 is a conceptual diagram of a system 10 for perfusing a heart 12 according to an illustrative embodiment of the invention. As shown, the illustrative system 10 includes two solutions, a first solution 13 and a second solution 15, which are joined through fluid line 14 and become part of a perfusion circuit that includes lines 16, 18 and 20. The fluid lines interconnect a perfusion operating system 22 and a mixing chamber 24, with solutions 13 and 15 to perfuse the heart 12. In some embodiments, the system 10 is sterilized prior to use. In operation, the solutions 13 and 15 feed to the mixing chamber 24 via the conduit 14. Solutions 13 and 15 mix with a perfusion fluid that circulates in the perfusion circuit. The perfusion fluid may be blood or synthetic blood based. Perfusion fluid recirculates from the heart 12 via the fluid conduit 18, the perfusion operating system 22, and the fluid conduit 20 to the mixing chamber 24. There the recirculated fluid is mixed with solutions 13 and 15 and fed to the heart via the conduit 16. Since the heart 12 is maintained in a beating state, it continues to receive fluid from the conduit 16 and feed it back via the conduit 18 for remixing in the mixing chamber 24.

FIG. 2 is another conceptual drawing depicting a further illustrative embodiment of the perfusion system 10. As shown, the illustrative embodiment of FIG. 2 includes a priming solution 17 applied to the system prior to addition of a perfusion fluid. As shown in FIG. 2, the priming solution 17 flows into line 26 to provide the priming solution 17 to the system prior to perfusing the heart 12 with a perfusion fluid containing solutions 13 and 15. The priming solution 17 may include, for example, any or all of the components included in the solutions 13 and 15.

In other illustrative embodiments, the solutions 15 and/or 17 may be provided systematically to optimize the effectiveness of the system. For example, as shown in FIG. 3, the solution 13 may include a composition or solution 19, which may include components that are sensitive to sterilization and other techniques used to prepare the solution 13 for perfusion. For example, the composition or solution 19 may include insulin or other bio-components, organic compounds, and/or biological molecules that would decompose or degrade when sterilized, such as via an autoclave. In this instance, as shown in FIG. 3, the composition or solution 19 may be added to the solution 13 prior to or during perfusion of the donor organ. As shown in FIG. 3, the composition or solution 19 may be applied to the solution 13 through a fluid conduit 28. The composition or solution 19 may also be applied to fluid conduit 14 (though not shown in FIG. 3), and then mixed with the perfusion fluid in chamber 24 just prior to application to the conduit 16.

In some illustrative embodiments, the solutions described herein are combined with a blood product, such as whole blood or components thereof, which may also be oxygenated, to give a perfusion fluid. Additionally, synthetic blood products may be used as a substitute or in combination with blood products. Such combined solutions may be included in a perfusion circuit, such as those shown in FIGS. 1-3.

In other illustrative embodiments, one of the composition or solutions 13, 15, 17, or 19 may include one or more organ stimulants, such as cardio stimulants, and/or other components. The other components may include, for example, adenosine, a magnesium ion source, one or more phosphates, calcium ions, etc. In embodiments utilizing two or more solutions, such as those in FIGS. 1-3, the solution 15 may include one or more carbohydrates and may also include a phosphate source. According to some illustrative embodiments, the perfusion fluid includes more than one composition or solution 13, 15, 17 or 19 to avoid precipitation of calcium phosphate in embodiments where calcium ions and phosphate ions are used.

In one embodiment, a maintenance solution is made from the combination of a first solution, substantially formed from one or more amino acids, and a second solution, substantially formed from one or more carbohydrates, such as dextrose or glucose. The maintenance solution may also have additives, such as those described herein, administered at the point of use just prior to infusion into the organ perfusion system. For example, additional additives that can be included with the solution or added at the point of use by the user include hormones and steroids, such as dexamethasone and insulin, as well as vitamins, such as an adult multi-vitamin, for example adult multivitamins for infusion, such as MVI-Adult™. Additional small molecules and large bio-molecules may also be included with the solution or added at the point of use by the user, for example, therapeutics and/or components typically associated with blood or blood plasma, such as albumin.

In some embodiments, therapeutics that may be included in the compositions, solutions, and systems described herein include hormones, such as thyroid hormones, for example T3 and/or T4 thyroid hormones. Further therapeutics that may be included include drugs such as anti-arrhythmic drugs, for example, for heart therapy, and beta blockers. For instance, in certain embodiments, one or more thyroid hormones, one or more anti-arrhythmic drugs, and one or more beta blockers are added to the first solution, the second solution, and/or the maintenance solution either before or during perfusion of the organ. The above therapeutics may also be added directly to the system, for example to the perfusion circuit before or during perfusion of the organ.

Table 3 sets forth an exemplary first solution, comprising a tissue culture media having the components identified in Table 3 and combined with an aqueous fluid, which may be used to perfuse an organ as described herein. The amounts of components listed in Table 3 are relative to each other and to the quantity of aqueous solution used. In some embodiments, about 500 mL of aqueous fluid is used. In other embodiments, about 1 L of aqueous fluid is used. For example, a combination of about 500 mL of first solution with 500 mL of second solution affords a maintenance solution of about 1 L. In some embodiments, the quantity of aqueous solution can vary±about 10%. The component amounts and the quantity of aqueous solution may be scaled as appropriate for use. The pH of the first solution, in this embodiment, may be adjusted to be about 7.0 to about 8.0, for example about 7.3 to about 7.6.

TABLE 3 Composition of Exemplary First Solution (about 500 mL aqueous Tissue Culture Component Amount Specification Adenosine 750 mg ±about 10% Calcium Chloride dihydrate 2400 mg  ±about 10% Glycine 350 mg ±about 10% L-Alanine 174 mg ±about 10% L-Arginine 700 mg ±about 10% L-Aspartic Acid 245 mg ±about 10% L-Glutamic Acid 258 mg ±about 10% L-Histidine 225 mg ±about 10% L-Isoleucine 115.5 mg   ±about 10% L-Leucine 343 mg ±about 10% L-Methionine  59 mg ±about 10% L-Phenylalanine  52 mg ±about 10% L-Proline 126 mg ±about 10% L-Serine  93 mg ±about 10% L-Threonine  70 mg ±about 10% L-Tryptophan  35 mg ±about 10% L-Tyrosine  92 mg ±about 10% L-Valine 171.5 mg   ±about 10% Lysine Acetate 225 mg ±about 10% Magnesium Sulfate Heptahydrate 400 mg ±about 10% Potassium Chloride  20 mg ±about 10% Sodium Chloride 1750 mg  ±about 10%

Since amino acids are the building blocks of proteins, the unique characteristics of each amino acid impart certain important properties on a protein such as the ability to provide structure and to catalyze biochemical reactions. The selection and concentrations of the amino acids provided in the first solution provide support of normal physiologic functions such as metabolism of sugars to provide energy, regulation of protein metabolism, transport of minerals, synthesis of nucleic acids (DNA and RNA), regulation of blood sugar and support of electrical activity, in addition to providing protein structure. Additionally, the concentrations of specific amino acids found in the first solution can be used to predictably stabilize the pH of the first solution and/or other solutions and fluids to which the first solution may be added, for example the maintenance solution and/or the perfusion fluid.

Table 4 provides an exemplary second solution 15, comprising a solution which may be used with the systems of FIGS. 1-3. The second solution 15 includes dextrose and an aqueous solvent. In some embodiments, the second solution 15 further includes sodium glycerol phosphate. The amount of component in Table 4 is relative to the amount of aqueous solvent (about 500 mL) and may be scaled as appropriate. In some embodiments, the quantity of aqueous solvent varies±about 10%.

TABLE 4 Components of Exemplary Second Solution (about 500 mL) Component Amount Specification Dextrose 40 g ±about 10%

Additional components may be included in the first or second solution, the maintenance solution, and/or the priming solution and may include one or more of those set forth in Table 5. These additional or supplemental components may be added individually, in various combinations, or all at once as a composition. For example, in certain embodiments, the epinephrine, insulin, and MVI-Adult™, listed in Table 5, are added to the maintenance solution. In another example, the SoluMedrol™ and the sodium bicarbonate, listed in Table 5, are added to the priming solution. The additional components may also be combined in one or more combinations or all together and placed in solution before being added to the first or second solution or to the maintenance solution. These additional components may be degraded or otherwise inactivated if subjected to sterilization, and, as such, may be appropriately applied directly to the perfusion circuit or to a first solution 13 or a second solution 15 after sterilization, such as described with regard to the composition or solution 19 of FIG. 3. In some embodiments, dexamethasone may also be added to solutions described herein. The component amounts listed in Table 5 are relative to each other and to the amounts of components in Table 3, Table 4 and/or Table 6 and/or to the amount of aqueous solution used in the first or second solution, the maintenance solution, or the priming solution and may be scaled as appropriate for the amount required.

TABLE 5 Exemplary Supplemental Components Added Prior to Use Component Amount Type Function Specification Epinephrine about 0.50 mg Catecholamine Maintains Vascular ±about 10% Hormone Tone, Basal Catecholamine Levels for Cardiac Function Insulin about 100 Units Hormone Facilitates Glucose ±about 10% Absorption MVI-Adult ™ 1 unit vial Vitamin Source of ±about 10% Antioxidants SoluMedrol ™ about 250 mg in Steroid ±about 10% about 4 mL water Sodium about 20 mEq Buffer Maintains pH ±about 10% Bicarbonate

As described above, in certain illustrative embodiments, solutions are used to perfuse an organ prior to transplantation. In certain embodiments, one or more solutions are applied to maintain the organ, and a separate solution is applied to prime the organ prior to applying one or more maintenance solutions. Certain exemplary components for priming solutions are identified in Table 6. In some embodiments, the priming solutions include one or more carbohydrates. The component amounts in Table 6 are relative to each other and to the amount of aqueous solvent (about 500 mL) and may be scaled as appropriate. In certain embodiments, the quantity of aqueous solvent varies±about 10%.

TABLE 6 Composition of Exemplary Priming Solution (about 500 mL aqueous solution) Component Amount Specification Mannitol 12.5 g  ±about 10% Sodium Chloride 4.8 g  ±about 10% Potassium Chloride 185 mg ±about 10% Magnesium Sulfate Heptahydrate 185 mg ±about 10% Sodium Glycerophosphate 900 mg ±about 10%

In some illustrative embodiments, the invention combines solutions as described herein in desired sequences to provide for improved physiological performance of the organ. Although the first and second solutions are shown above and referenced as solutions 13 and 15 in FIGS. 1-3, the solutions may be divided into any number of solutions, their positions interchanged in system 10, and applied in any manner necessary to maintain the functioning organ.

In some illustrative embodiments, the solutions contemplated herein may be adapted for use with an organ perfusion system such as that described in U.S. Pat. Nos. 6,100,082, 6,046,046, or PCT Application No. PCT/US98/19912, the specifications of which are hereby incorporated by reference in their entireties.

Certain embodiments of the first solution include epinephrine and a plurality of amino acids. In certain embodiments, the first solution includes electrolytes, such as calcium and magnesium. The first solution may also include other components such as adenosine and/or one or more components from Table 3. In certain embodiments a second solution includes one or more carbohydrates and may also include a phosphate source. The second solution is typically maintained at a pH of about 5.0 to about 6.5, for example, about 5.5 to about 6.0.

In certain embodiments, methods for perfusing an organ are provided through the use of solutions described herein, such as through the systems illustrated in FIGS. 1-3. As shown in the figures, perfusion may occur through a beating organ and may take place at a physiological temperature. In certain embodiments, the methods include providing a fluid for perfusing the organ, the fluid including a phosphate source, one or more organ stimulants, such as epinephrine, and one or more carbohydrates. In certain embodiments, the fluid may be perfused through the organ and the organ is maintained in a beating state without the use of an applied vasopressor to effect the circulation of the fluid. In some embodiments, the organ is a heart.

In one illustrative embodiment, the system described in FIGS. 1-3 is used to perfuse an organ. In this embodiment, the system has a port for a priming solution that includes the priming solution shown in Table 6, a first chamber that contains a first solution having the components described above in Table 3, a port for a supplemental composition or solution having the components described in Table 5, and a second chamber containing a second solution having the components shown in Table 4. The priming solution may also be contained in a third chamber. The first solution, second solution, and the priming solution are sterilized through an autoclave, for example, to a sterility assurance level of about 10−6, then the supplemental composition or solution is added to the first solution. The perfusion system is then primed with the priming solution for about 10 minutes. In some embodiments, the priming is through perfusion in the direction of normal flow of fluid through the organ. In other embodiments, the priming is through retrograde perfusion, e.g., having a flow direction that is counter directional to the normal flow of fluid through the organ. After priming, the remaining solutions are combined and infused into the perfusion system at a flow rate selected to replace necessary substrates as they are consumed by the organ. In some embodiments, the flow rate approximates physiologic fluid flow through the organ. Fluid flow rates during heart priming are about 1 L/min, while solution infusion rates during heart perfusion are about 10 mL/hr to about 40 mL/hr. Optionally, an additional solution containing a blood product is provided through which the blood product is combined with the circulating fluid and used to perfuse the organ.

In one embodiment, a composition for use in a solution for perfusing an organ, such as a heart, is provided comprising one or more carbohydrates, one or more organ stimulants, and a plurality of amino acids that do not include asparagine, glutamine, or cysteine. The composition may also include other substances, such as those used in solutions described herein.

In another embodiment, a system for perfusing an organ, such as a heart, is provided comprising an organ and a substantially cell-free composition, comprising one or more carbohydrates, one or more organ stimulants, and a plurality of amino acids that do not include asparagine, glutamine, or cysteine. Substantially cell-free includes systems that are substantially free from cellular matter; in particular, systems that are not derived from cells. For example, substantially cell-free includes compositions and solutions prepared from non-cellular sources.

In another aspect, the solutions and systems may be provided in the form of a kit that includes one or more organ maintenance solutions. An exemplary maintenance solution may include components identified above in one or more fluid solutions for organ perfusion. In certain embodiments, the maintenance solution may include multiple solutions, such as a first solution and a second solution and/or a supplemental composition or solution, or may include dry components that may be regenerated in a fluid to form one or more solutions for organ perfusion. The kit may also comprise one or more concentrated solutions which, on dilution, provide a first, second, and/or supplemental solution as described herein. The kit may also include a priming solution. In an exemplary embodiment, the maintenance solution includes a first solution and second solution such as those described above, and a priming solution such as that described above.

In certain embodiments, the kit is provided in a single package, wherein the kit includes one or more solutions (or components necessary to formulate the one or more solutions by mixing with an appropriate fluid), and instructions for sterilization, flow and temperature control during perfusion and use and other information necessary or appropriate to apply the kit to organ perfusion. In certain embodiments, a kit is provided with only a single solution (or set of dry components for use in a solution upon mixing with an appropriate fluid), and the single solution (or set of dry components) is provided along with a set of instructions and other information or materials necessary or useful to operate the solution in a perfusion apparatus or system.

In another aspect, the systems, solutions and methods may be used to deliver therapeutics to an organ during perfusion. For example, one or more of the solutions and/or systems described above may include one or more drugs, biologics, gene therapy vectors, or other therapeutics which are delivered to the organ during perfusion. Suitable exemplary therapeutics may include drugs, biologics, or both. Suitable drugs may include, for example, antifungals, anti-microbials or anti-biotics, anti-inflammatories, anti-proliferatives, anti-virals, steroids, retinoids, NSAIDs, vitamin D3 and vitamin D3 analogs, calcium channel blockers, complement neutralizers, ACE inhibitors, immunosuppressants, and other drugs. Suitable biologics may include proteins; suitable biologics may also include vectors loaded with one or more genes for gene therapy application.

For example, suitable steroids include but are not limited to androgenic and estrogenic steroid hormones, androgen receptor antagonists and 5-α-reductase inhibitors, and corticosteroids. Specific examples include but are not limited to alclometasone, clobetasol, fluocinolone, fluocortolone, diflucortolone, fluticasone, halcinonide, mometasone, prednisone, prednisolone, methylprednisolone, triamcinolone, betamethasone, and dexamethasone, and various esters and acetonides thereof.

Suitable retinoids include but are not limited to retinol, retinal, isotretinoin, acitretin, adapalene, tazarotene, and bexarotene.

Suitable NSAIDs include but are not limited to naproxen, suprofen, ketoprofen, ibuprofen, flurbiprofen, diclofenac, indomethacin, celecoxib, and rofecoxib.

Suitable vitamin D3 analogues include but are not limited to doxercalciferol, seocalcitol, calcipotriene, tacalcitol, calcitriol, ergocalciferol, and calcifediol.

Suitable anti-viral agents include but are not limited to trifluridine, cidofovir, acyclovir, penciclovir, famciclovir, valacyclovir, ganciclovir, and docosanol.

Suitable human carbonic anhydrase inhibitors include but are not limited to methazolamide, acetazolamide, and dorzolamide.

Suitable anti-proliferative agents include but are not limited to 5-FU, taxol, daunorubicin, and mitomycin.

Suitable antibiotic (antimicrobial) agents include but are not limited to bacitracin, chlorhexidine, chlorhexidine digluconate, ciprofloxacin, clindamycin, erythromycin, gentamicin, lomefloxacin, metronidazole, minocycline, moxifloxacin, mupirocin, neomycin, ofloxacin, polymyxin B, rifampicin, rufloxacin, tetracycline, tobramycin, triclosan, and vancomycin. The antiviral and antibacterial prodrugs described herein may be used to treat appropriately responsive systemic infections.

Certain experimental data are available to describe certain embodiments of the solutions described herein and their use in organ perfusion. Certain data are set for in FIGS. 4-6. FIG. 4 depicts a chart demonstrating electrolyte stability for an organ undergoing perfusion according to an embodiment of the invention. In the embodiment associated with FIG. 4, the organ is a heart wherein perfusion is conducted analogous to physiological conditions; e.g., a solution as described herein is perfused in the typical physiological direction out of the aorta. The rate of infusion is approximately 30 mL/hr. As can be seen from FIG. 4, the levels of various electrolytes: sodium, potassium, calcium, and chloride ions, as well as dissolved glucose, remain at stable levels throughout the course of perfusion, from before the organ is connected to the perfusion system (the pre-instrumentation (PI) period) to six hours after connection of the organ.

FIG. 5 depicts a chart demonstrating electrolyte stability for an organ undergoing perfusion according to another embodiment of the invention. In the embodiment associated with FIG. 5, the organ is a heart wherein perfusion is conducted, in one respect, in an atypical fashion from physiological conditions; that is, a solution as described herein is perfused in the reverse physiological direction into the aorta. Such a retrograde perfusion perfuses the coronary sinus. In some embodiments, the left side of the heart remains empty. The rate of infusion is approximately 30 mL/hr. As can be seen from FIG. 5, the levels of various electrolytes: sodium, potassium, calcium, and chloride ions, as well as dissolved glucose, remain at stable levels throughout the course of perfusion, from before the organ is connected to the perfusion system (the pre-instrumentation (PI) period) to six hours after connection of the organ. FIG. 5 also demonstrates that the levels of the electrolytes and glucose remain at levels similar to those for the base line (BL) normal physiological state for the organ.

FIG. 6 depicts a chart demonstrating the arterial blood gas profile for an organ undergoing perfusion according to another embodiment of the invention. As can be seen from FIG. 6, the levels of various blood gasses: carbon dioxide and oxygen, and pH remain at stable levels throughout the six-hour course of perfusion. FIG. 6 also demonstrates that the levels of carbon dioxide, oxygen, and pH remain at levels similar to those for two base line (BL) measurements for the normal physiological state for the organ. FIGS. 4-6 demonstrate the ability of the present systems and methods to maintain an organ under stable physiological or near physiological conditions.

This application incorporates by reference the specification from each of the following applications: U.S. application Ser. No. 09/534,092, filed on Mar. 23, 2000; PCT/US98/19912, filed on Sep. 23, 1998; U.S. application Ser. No. 09/054,698, filed on Apr. 3, 1998; and U.S. application Ser. No. 08/936,062, filed on Sep. 23, 1997.

It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. For example, a variety of systems and/or methods may be implemented based on the disclosure and still fall within the scope of the invention. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

1. A method for perfusing an ex-vivo heart, the method comprising:

receiving an ex-vivo heart in an organ care system, wherein the ex-vivo heart is coupled to a perfusion circuit;
receiving a first solution in a first chamber of the organ care system;
receiving a second solution in a second chamber of the organ care system;
infusing, into a first fluid line in fluid communication with the first chamber and the second chamber, the first solution and the second solution;
combining, in a second fluid line of the perfusion circuit, the first solution and the second solution with a perfusion fluid to create a mixed solution; and
circulating the mixed solution through the perfusion circuit, the circulating comprising: pumping, via the second fluid line of the perfusion circuit, the mixed solution to the ex-vivo heart; and receiving, in a third fluid line of the perfusion circuit, the mixed solution from the ex-vivo heart.

2. The method of claim 1, comprising:

receiving a priming solution in the perfusion circuit.

3. The method of claim 2, wherein the priming solution comprises mannitol, sodium chloride, potassium chloride, magnesium sulfate heptahydrate, and sodium glycerophosphate.

4. The method of claim 2, comprising:

combining the priming solution with the perfusion fluid.

5. The method of claim 4, wherein the perfusion fluid comprises whole blood or synthetic blood.

6. The method of claim 5, comprising:

at least partially depleting the whole blood of leukocytes or platelets before combining the perfusion fluid with the at least one of the first solution, the second solution, or the priming solution.

7. The method of claim 2, comprising:

circulating the priming solution through the perfusion circuit in a forward flow direction.

8. The method of claim 2, comprising:

circulating the priming solution through the perfusion circuit in a retrograde flow direction.

9. The method of claim 1, wherein one of at least the first solution or the second solution comprises:

one or more cardio stimulants comprising a catecholamine, a peptide, a polypeptide, a β1/β2-adrenoreceptor blocking agent, bupranolol, pindolol, alprenolol, a cardiac glycoside, digitalis, palustrin, or ferulic acid;
a plurality of amino acids selected from the group consisting of alanine, arginine, aspartic acid, glutamic acid, histidine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, and lysine that do not include asparagine, glutamine, and cysteine; and
one or more electrolytes.

10. The method of claim 1, comprising:

receiving a composition or a third solution comprising a component sensitive to sterilization in at least one of the first solution or the second solution after at least one of the first solution or the second solution has been sterilized.

11. The method of claim 10, wherein the component sensitive to sterilization comprises insulin.

12. The method of claim 10, wherein the at least one of the first solution or the second solution receives the composition or the third solution via a fourth fluid line.

13. The method of claim 1, comprising:

infusing the first solution or the second solution into the perfusion circuit at a rate of between about 10 mL/hr to about 40 mL/hr.

14. The method of claim 13, comprising:

infusing the first solution or the second solution into the perfusion circuit at a rate of about 30 mL/hr.

15. The method of claim 13, comprising:

infusing the first solution into the perfusion circuit at a first rate; and
infusing the second solution into the perfusion circuit at a second rate different from the first rate.

16. The method of claim 1, comprising:

circulating the mixed solution through the perfusion circuit at a rate of about 1 L/min.

17. A method comprising:

receiving an ex-vivo organ in an organ care system, wherein the ex-vivo organ is coupled to a perfusion circuit;
receiving a first solution in a first chamber of the organ care system;
receiving a second solution in a second chamber of the organ care system;
infusing, into the perfusion circuit, the first solution and the second solution at a rate of between about 10 mL/hr to about 40 mL/hour, wherein after the infusing, the first solution and the second solution are combined with the perfusion fluid to create a mixed solution; and
circulating a perfusion fluid through the perfusion circuit, wherein the circulating comprises: pumping, via a first fluid line of the perfusion circuit, the mixed solution to the ex-vivo organ; and
receiving, in a second fluid line of the perfusion circuit, the mixed solution from the ex-vivo organ.

18. The method of claim 17, comprising:

receiving a priming solution in the perfusion circuit.

19. The method of claim 18, wherein the priming solution comprises mannitol, sodium chloride, potassium chloride, magnesium sulfate heptahydrate, and sodium glycerophosphate.

20. The method of claim 18, wherein one of at least the first solution or the second solution comprises:

a plurality of amino acids comprising alanine, arginine, histidine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and lysine that do not include asparagine, glutamine, and cysteine; and
one or more electrolytes.

21. A method for perfusing an ex-vivo organ, the method comprising:

receiving an ex-vivo organ in a chamber of an organ care system, wherein the ex-vivo organ is coupled to a perfusion circuit;
circulating a priming solution through the perfusion circuit;
after circulating the priming solution through the perfusion circuit, infusing a first solution and a second solution into the perfusion circuit at a rate of between about 10 mL/hr to about 40 mL/hour, wherein after the infusing, the first solution and the second solution are combined with the perfusion fluid to create a mixed solution; and
circulating a perfusion fluid through the perfusion circuit at a rate of between about 1 L/min and about 5 L/min.

22. The method of claim 21, comprising:

circulating the priming solution through the perfusion circuit in a forward flow direction.

23. The method of claim 21, comprising:

circulating the priming solution through the perfusion circuit in a retrograde flow direction.

24. The method of claim 21, comprising:

infusing the first solution into the perfusion circuit at a first rate; and
infusing the second solution into the perfusion circuit at a second rate different from the first rate.

25. The method of claim 21, comprising:

infusing the first solution or the second solution into the perfusion circuit at a rate of about 30 mL/hr.

26. The method of claim 21, wherein the priming solution comprises mannitol, sodium chloride, potassium chloride, magnesium sulfate heptahydrate, and sodium glycerophosphate.

27. The method of claim 21, wherein one of at least the first solution or the second solution comprises:

one or more cardio stimulants comprising a catecholamine, a peptide, a polypeptide, a β1/β2-adrenoreceptor blocking agent, bupranolol, pindolol, alprenolol, a cardiac glycoside, digitalis, palustrin, and ferulic acid;
a plurality of amino acids selected from the group comprising alanine, arginine, aspartic acid, glutamic acid, histidine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, and lysine that do not include asparagine, glutamine, and cysteine; and
one or more electrolytes.
Patent History
Publication number: 20230413805
Type: Application
Filed: Sep 1, 2023
Publication Date: Dec 28, 2023
Applicant: TransMedics, Inc. (Andover, MA)
Inventors: Waleed H. HASSANEIN (North Andover, MA), Tamer I. KHAYAL (North Andover, MA), Paul LEZBERG (Westford, MA)
Application Number: 18/241,286
Classifications
International Classification: A01N 1/02 (20060101); C12M 3/00 (20060101);