Organ care solution for ex-vivo machine perfusion of donor lungs

Information

  • Patent Grant
  • 11856944
  • Patent Number
    11,856,944
  • Date Filed
    Wednesday, July 20, 2022
    a year ago
  • Date Issued
    Tuesday, January 2, 2024
    4 months ago
Abstract
An ex-vivo lung solution for machine perfusion of donor lungs on OCS. The solution may be mixed with whole blood or packed red blood cells to form the OCS lung perfusion solution.
Description
TECHNICAL FIELD

The disclosure generally relates a perfusion solution for ex-vivo organ care. More particularly, the disclosure relates to a solution for machine perfusion of donor lungs on an organ care system (“OCS”) at physiologic or near-physiologic conditions.


BACKGROUND

Current organ preservation techniques typically involve hypothermic storage of the organ in a chemical perfusion solution. In the case of the lung, it is typically flushed with a cold preservation solution such as Perfadex™ and then immersed in that same cold solution until it is transplanted. These techniques utilize a variety of cold preservation solutions, none of which sufficiently protect the lungs from tissue damage resulting from ischemia. Such injuries are particularly undesirable when an organ, such as a lung, is intended to be transplanted from a donor into a recipient.


Using conventional approaches, tissue injuries increase as a function of the length of time an organ is maintained ex-vivo. For example, in the case of a lung, typically it may be preserved ex-vivo for only about 6 to about 8 hours before it becomes unusable for transplantation. As a result, the number of recipients who can be reached from a given donor site is limited, thereby restricting the recipient pool for a harvested lung. Compounding the effects of cold ischemia, current cold preservation techniques preclude the ability to evaluate and assess an organ ex-vivo. Because of this, less-than-optimal organs may be transplanted, resulting in post-transplant organ dysfunction or other injuries, or resuscitatable organs may be turned down.


Prolonged and reliable ex-vivo organ care would also provide benefits outside the context of organ transplantation. For example, a patient's body, as a whole, can typically tolerate much lower levels of chemo-, bio- and radiation therapy than many particular organs. An ex-vivo organ care system would permit an organ to be removed from the body and treated in isolation, reducing the risk of damage to other parts of the body. Thus, there is a need to develop techniques and perfusion solutions that do not require hypothermic storage of the organ and extend the time during which an organ can be preserved in a healthy state ex-vivo. Such techniques would improve transplant outcomes and enlarge potential donor and recipient pools.


SUMMARY

The disclosure provides improved methods, solutions, and systems related to ex-vivo organ care. In general, in one aspect, the disclosure features a lung OCS solution for machine perfusion of donor lungs on OCS at near physiologic conditions. In another aspect, the disclosure includes a system and method for perfusing one or more lungs ex-vivo for an extended period of time in a functional and viable state maintenance mode at near physiologic conditions. In another aspect the disclosure includes a method of producing a solution for ex-vivo perfusion of a donor lung at near physiologic conditions.


The present disclosure describes an OCS lung perfusion solution that can be used for machine perfusion of donor lungs on OCS. The solution may include energy-rich perfusion nutrients, as well as a supply of therapeutics, vasodilators, endothelial stabilizers, and/or preservatives for reducing edema and providing endothelial support to the lungs. In a preferred embodiment, the solution comprises: dextran 40; sodium chloride; potassium chloride; magnesium sulfate anhydrate; disodium phosphate anhydrate; monopotassium phosphate; glucose monohydrate; milrinone; nitroglycerin; insulin; a multi-vitamin (M.V.I. Adult® or equivalent); sodium bicarbonate; methylprednisolone (SoluMedrol® or equivalent); cefazolin; Ciprofloxacin; voriconazole. The solution is mixed with whole blood or packed red blood cells to form the OCS lung perfusion solution. The solution provides the components for maintaining a functional (e.g., under respiration) and viable lung ex-vivo at near physiologic conditions.


According to certain embodiments, solutions with particular solutes and concentrations are selected and proportioned to provide for 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 and increasing colloid pressure, minimize lung edema and cell swelling.


In another embodiment, a method of perfusing a lung is featured. The method includes: positioning the lung in an ex-vivo perfusion circuit; circulating an OCS lung solution specifically for machine perfusion of donor lungs on OCS through the lung, the fluid entering the lung through a pulmonary artery interface and leaving the lung through a left atrial interface; ventilating the lung by flowing a ventilation gas through a tracheal interface; deoxygenating the perfusion solution until a predetermined first value of oxygen content in the perfusion solution is reached; reoxygenating the perfusion solution by ventilating the lung with an oxygenation gas until a predetermined second value of oxygen content in the perfusion solution is reached; and determining a condition of the lung based on a time taken for the lung to cause the oxygen content level in the perfusion solution to change from the first value of oxygen content to the second value of oxygen content.


In another embodiment, a method of producing a solution for perfusing a lung at near physiologic conditions is featured. This method includes combining pre-weighed raw materials including nutrients, colloids, hormones, steroids, buffers and vasodilators with water for injection (“WFI”) and mixed with heating until fully dissolved, monitoring the pH level of the resulting solution, allowing the solution to cool, filtering the cooled solution, dispensing the solution into a primary container and sterilizing the filled container.


In another aspect, a lung care system is featured. The lung system includes: a single use disposable module including an interface adapted to couple the single use disposable module with the multiple use module for electro-mechanical interoperation with the multiple use module; a lung chamber assembly optionally having a first interface for allowing a flow of a lung OCS perfusion solution into the lung, a second interface for allowing ventilation of the lung with a ventilation gas, and a third interface for allowing a flow of the perfusion solution away from the lung, the lung chamber assembly including a dual drain system for carrying the flow of the perfusion solution away from the lung, the dual drain system comprising a measurement drain for directing a part of the perfusion solution flow to a sensor of a perfusion solution gas content and a main drain for receiving a remaining part of perfusion solution flow; and an OCS lung perfusion solution specifically for machine perfusion of donor lungs on OCS.





BRIEF DESCRIPTION OF THE DRAWINGS

The following figures depict illustrative embodiments in which like reference numerals refer to like elements. These depicted embodiments may not be drawn to scale and are to be understood as being illustrative and not as limiting.



FIG. 1 is a schematic diagram of the lung perfusion circuit of the described embodiment.



FIG. 2 is an illustration of the organ care system drawn from a 45-degree angle from the front view, according to the described embodiment.



FIG. 3 is an illustration of the lung perfusion module, according to the described embodiment.



FIG. 4 is an illustration of the pulmonary artery cannula, according to the described embodiment.



FIG. 5 is an illustration of the tracheal cannula, according to the described embodiment.



FIG. 6 is an exploded illustration of the lung chamber, according to the described embodiment.



FIG. 7 is a schematic diagram of the described embodiment of a portable organ care system including shows the gas-related components of the lung perfusion module.





DETAILED DESCRIPTION

The following description and the drawings illustrate embodiments sufficiently to enable those skilled in the art to practice them. Other embodiments may incorporate structural, logical, electrical, process, and other changes. Examples merely typify possible variations. Individual components and functions are optional unless explicitly required, and the sequence of operations may vary. Portions and features of some embodiments may be included in or substituted for those of others. The scope of embodiments encompasses the full ambit of the claims and all available equivalents of those claims.


Improved approaches to ex-vivo organ care are provided. More particularly, various embodiments are directed to improved methods and solutions relating to maintaining a lung at or near normal physiologic conditions in an ex-vivo environment. As used herein, “physiological temperature” is referred to as temperatures between about 25 degrees C. and about 37 degrees C. A preferred embodiment comprises a lung OCS perfusion solution that may be administered in conjunction with an organ care system to maintain a lung in an equilibrium state by circulating a perfusion solution through the lung's vascular system, while causing the lung to rebreath a gas having an oxygen content sufficient to met the lung's metabolic needs.


The embodiments allow a lung to be maintained ex-vivo for extended periods of time, such as, for example, 3-24 or more hours. Such extended ex-vivo maintenance times expand the pool of potential recipients for donor lungs, making geographic distance between donors and recipients less important. Extended ex-vivo maintenance times also provide the time needed for better genetic and HLA matching between donor organs and organ recipients, increasing the likelihood of a favorable outcome. The ability to maintain the organ in a near physiologic functioning condition also allows a clinician to evaluate the organ's function ex-vivo, and identify organs that are damaged. This is especially valuable in the case of the lung, since lungs are often compromised as a direct or indirect result of the cause of the death of the donor. Thus even a newly harvested lung may be damaged. The ability to make a prompt assessment of a harvested organ allows a surgeon to determine the quality of a lung and, if there is damage, to make a determination of the nature of the problem. The surgeon can then make a decision as to whether to discard the lung, or to apply therapy to the lung. Therapies can include recruitment processes, removing or stapling off damaged areas of lung, suctioning secretions, cauterizing bleeding blood vessels, and giving radiation treatment. The ability to assess and, if necessary provide therapy to lungs at several stages from harvesting to implantation greatly improves the overall likelihood of lung transplant success and increases the number of organs available for transplant. In some instances, the improved assessment capability and extended maintenance time facilitates medical operators to perform physical repairs on donor organs with minor defects. Increased ex-vivo organ maintenance times can also provide for an organ to be removed from a patient, treated in isolation ex-vivo, and then put back into the body of a patient. Such treatment may include, without limitation, pharmaceutical treatments, gas therapies, surgical treatments, chemo-, bio-, gene and/or radiation therapies.


Overview of OCS Perfusion Solution


According to certain embodiments, a lung OCS perfusion solution with certain solutes provides for the lungs to function at physiologic or near physiologic conditions and temperature by supplying energy rich nutrients, oxygen delivery, optimal oncotic pressure, pH and organ metabolism. The perfusion solution may also include therapeutic components to help maintain the lungs and protect them against ischemia, reperfusion injury and other ill effects during perfusion. Therapeutics may also help mitigate edema, provide general endothelial tissue support for the lungs, and otherwise provide preventative or prophylactic treatment to the lungs.


The amounts of solutes provided describes preferred amounts relative to other components in the solution and may be scaled to provide compositions of sufficient quantity.


In one embodiment, the solution may include a phosphodiesterase inhibitor. To improve gas exchange and diminish leukocytosis, an adenosine-3′,5′-cyclic monophosphate (cAMP) selective phosphodiesterase type III (PDE III) inhibitor such as milrinone, amrinone, anagrelide, bucladesine, cilostamide, cilostazol, enoximone, KMUP-1, quazinone, RPL-554, siguazodan, trequinsin, vesnarinone, zardaverine may be added. In a preferred embodiment milrinone is added. Milrinone has the effects of vasorelaxation secondary to improved calcium uptake into the sarcoplasmic reticulum, inotropy (myocyte contraction) due to cAMP-mediated trans-sarcolemmal calcium flux, and lusitropy (myocyte relaxation) possibly due to improved actin-myosin complex dissociation. In a preferred embodiment milrinone is present in each 1 L of solution in an amount of about 3400 mcg to about 4600. In a particularly preferred embodiment, milrinone is present in each 1 L of solution in an amount of about 4000 mcg.


In certain embodiments the solution may include a nitrate which is useful in the nitrogen cycle. Nitroglycerin is a nitrate that may be added to the perfusion solution to promote stabilization of pulmonary hemodynamics and improve arterial oxygenation after transplantation. When a lung is removed from the body, nitric oxide levels fall quickly because it is quenched by superoxide generated during reperfusion, resulting in damage to the lung tissue. Nitroglycerin can act to promote nitric oxide levels in a lung ex-vivo by way of intracellular S-nitrosothiol intermediates to directly stimulate guanylate cyclase or to release nitric oxide locally in effector cells. To this end, Nitroglycerin improves vascular homeostasis and improves organ function by providing better arterial oxygenation after transplant. In a preferred embodiment nitroglycerin is present in each 1 L of solution in an amount of about 10 mg to about 50 mg.


In one other embodiment, magnesium sulfate anhydrate may be added to the solution. Pulmonary artery blood pressure is lower than blood pressure in the rest of the body and in the case of pulmonary hypertension, magnesium sulfate promotes vasodilatation in constricted muscles of the pulmonary arteries by modulating calcium uptake, binding and distribution in smooth muscle cells, thereby decreasing the frequency of depolarization of smooth muscle and thus promoting vasodilatation. Magnesium sulfate anhydrate is present in each 1 L of solution in an amount of about 0.083 g to about 0.1127 g. In a particularly preferred embodiment magnesium sulfate anhydrate is present in each 1 L of solution in an amount of about 0.098 g.


In a preferred embodiment, the addition of colloids offers numerous benefits including improving erythrocyte deformability, preventing erythrocyte aggregation, inducing disbanding of already aggregated cells and preserving endothelial-epithelial membrane. Colloids also have anti-thrombotic effects by being able to coat endothelial surfaces and platelets. In this embodiment dextran 40 is present in each 1 L of solution in an amount of about 42.5 g to about 57.5 g. In a particularly preferred embodiment, dextran 40 is present in each 1 L of solution in an amount of about 50 g.


The solution may also contain electrolytes, such as sodium, potassium, chloride, sulfate, magnesium and other inorganic and organic charged species, or combinations thereof. A suitable component may be those 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. In this embodiment, sodium chloride is present in each 1 L of solution in an amount of about 6.8 g to about 9.2 g. In a particularly preferred embodiment, sodium chloride is present in each 1 L of solution in an amount of about 8 g.


In a preferred embodiment the solution may have a low-potassium concentration. A low-level of potassium results in improved lung function. A low potassium level may also protect the lung during high flow reperfusion and lead to a lower PA pressure and PVR, lower percent decrease in dynamic airway compliance, and lower wet to dry ratio. In this embodiment potassium chloride is present in each 1 L of solution in an amount of about 0.34 g to about 0.46 g. In a particularly preferred embodiment potassium chloride is present in each 1 L of solution in an amount of about 0.4 g.


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 glucose monohydrate, 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. In a preferred embodiment glucose monohydrate is present in each 1 L of solution an amount of about 1.7 g to about 2.3 g. In a particularly preferred embodiment glucose monohydrate is present in each 1 L of solution an amount of about 2 g.


The solution 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 a hormone to promote and regulate carbohydrate and fat metabolism. Insulin acts to improve cell function by promoting optimum glucose and glycogen intake into the cells. In this preferred embodiment each 1 L of the solution may contain about 17 IU insulin to about 23 IU insulin. In a particularly preferred embodiment each 1 L of the solution may contain 20 IU insulin.


In addition, the solution may include a multi-vitamin that provides anti-oxidants and co-enzymes and helps maintain the body's normal resistance and repair processes. The multi-vitamin may include certain fat soluble vitamins such as Vitamins A, D, E, and K, and water soluble vitamins such as Vitamin C, Niacinamide, Vitamins B2, B1, B6, and Dexpanthenol, as well as stabilizers and preservatives. In a preferred embodiment, each 1 L of the solution contains one unit vial of M.V.I. Adult® multi-vitamin. M.V.I. Adult® includes fat soluble vitamins such as Vitamins A, D, E, and K, and water soluble vitamins such as Vitamin C, Niacinamide, Vitamins B2, B1, B6, and Dexpanthenol, as well as stabilizers and preservatives in an aqueous solution.


The solution may also include an anti-inflammatory agent such as a glucocorticoid steroid. Glucocorticoid steroids act as anti-inflamatory agents by activating to the cell's glucocorticoid receptors which in turn up-regulate the expression of anti-inflammatory proteins in the nucleus and reduce the expression of pro-inflammatory proteins. Glucocorticoid steroids include methylprednisolone, hydrocortisone, cortisone acetate, prednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone acetate and aldosterone. In this preferred embodiment, each 1 L of the solution may contain about 0.85 g mg to about 1.15 g methylprednisolone (SoluMedrol® or equivalent). In a particularly preferred embodiment, each 1 L of the solution may contain 1 g methylprednisolone (SoluMedrol® or equivalent)


In addition the solution may contain buffers to maintain the solution at an optimal pH. These may include disodium phosphate anhydrate, a physiologic balancing buffer or monopotassium phosphate to maintain the average pH of the solution during lung tissue perfusion. In this embodiment disodium phosphate anhydrate is present in each 1 L of solution in an amount of about 0.039 g to about 0.052 g, and/or monopotassium phosphate in an amount of about 0.053 g to about 0.072 g. In a particularly preferred embodiment, disodium phosphate anhydrate is present in an amount of 0.046 g, and/or monopotassium phosphate in an amount of 0.063 g. In some embodiments, the solution contains sodium bicarbonate, potassium phosphate, or TRIS buffer. In a preferred embodiment the sodium bicarbonate is present in each 1 L of solution in an amount of about 12.75 mEq to about 17.25 mEq. In a particularly preferred embodiment each 1 L of the solution may initially contain about 15 mEq sodium bicarbonate (5 mEq to each 500 mL bottle and 2-3 bottles are used), and additional amounts may be added throughout preservation based on clinical judgment. For example, 20-40 mEq can be added to the system as part of priming.


Other suitable buffers include 2-morpholinoethanesulfonic acid monohydrate (IVIES), cacodylic acid, H2CO3/NaHCO3 (pKa1), citric acid (pKa3), bis(2-hydroxyethyl)-imino-tris-(hydroxymethyl)-methane (Bis-Tris), N-carbamoylmethylimidino acetic 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), NaH.sub.2PO.sub.4/Na.sub.2HPO.sub.4 (pK.sub.a2), 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 hydroxymethylaminoethane (Tris), glycineamide, N,N-bis(2-hydroxyethyl) glycine (Bicine), glycylglycine (pKa2), N-tris(hydroxymethyl)methyl-3-aminopropanesulfonic acid (TAPS), or a combination thereof.


The solution may contain an antimicrobial or antifungal agent to prevent infection. These may include bacteria and fungal antimicrobial agents that provide protection against both gram negative and gram positive bacteria. Suitable antimicrobial or antifungal agents include cefazolin, ciprofloxacin, and voriconazole or equivalent. In a preferred embodiment, cefazolin is present in each 1 L of solution in an amount of about 0.85 g to about 1.15 g, ciprofloxacin is present in each 1 L of solution in an amount of about 0.17 g to about 2.3 g, and voriconazole is present in each 1 L of solution in an amount of about 0.17 g to about 2.3 g. In a particularly preferred embodiment, cefazolin is present in each 1 L of solution in an amount of about 1 g, ciprofloxacin is present in each 1 L of solution in an amount of about 0.2 g, and voriconazole is present in each 1 L of solution in an amount of about 0.2 g. Alternatively the solution may contain any effective antimicrobial or antifungal agent.


The solutions are preferably provided at a physiological temperature and maintained thereabout throughout perfusion and recirculation.


In a preferred embodiment the OCS lung perfusion solution comprises a nutrient, a colloid, a vasodilator, a hormone and a steroid.


In another preferred embodiment the solution comprises a nutrient including Glucose monohydrate, sodium chloride, potassium chloride, a multi-vitamin including fat-soluble and water-soluble vitamins; a colloid including dextran 40; a hormone including insulin; a steroid including methylprednisolone; buffering agents including disodium phosphate anhydrate, monopotassium phosphate and sodium bicarbonate; vasodilators including milrinone, nitroglycerin and magnesium sulfate anhydrate; antimicrobial or antifungal agents including cefazolin, ciprofloxacin, and voriconazole.


In another preferred embodiment the solution comprises an effective amount of dextran 40; sodium chloride; potassium chloride; magnesium sulfate anhydrate; disodium phosphate anhydrate; monopotassium phosphate; glucose monohydrate; milrinone; nitroglycerin; insulin; a multi-vitamin (M.V.I. Adult® or equivalent); sodium bicarbonate; methylprednisolone (SoluMedrol® or equivalent); cefazolin; ciprofloxacin; voriconazole.


In a preferred embodiment of the OCS lung perfusion solution, each 1 L of solution includes, milrinone in an amount of about 4000 mcg; nitroglycerin in an amount of about 10-50 mg; dextran 40 in an amount of about 50 g; sodium chloride in an amount of about 8 g; potassium chloride in an amount of about 0.4 g; magnesium sulfate anhydrate in an amount of about 0.098 g; disodium phosphate anhydrate in an amount of about 0.046 g; monopotassium phosphate in an amount of about 0.063 g; glucose monohydrate in an amount of about 2 g; insulin in an amount of about 20 IU; a multi-vitamin (M.V.I. Adult® or equivalent) in the amount of about 1 unit vial; sodium bicarbonate is initially present in an amount of about 15 mEq; methylprednisolone in an amount of about 1 g.


In a particularly preferred embodiment of the OCS lung perfusion solution, each 1 L of solution includes, milrinone in an amount of about 4000 mcg; nitroglycerin in an amount of about 10-50 mg; dextran 40 in an amount of about 50 g; sodium chloride in an amount of about 8 g; potassium chloride in an amount of about 0.4 g; magnesium sulfate anhydrate in an amount of about 0.098 g; disodium phosphate anhydrate in an amount of about 0.046 g; monopotassium phosphate in an amount of about 0.063 g; glucose monohydrate in an amount of about 2 g; insulin in an amount of about 20 IU; a multi-vitamin (M.V.I. Adult® or equivalent) in the amount of about 1 unit vial; sodium bicarbonate is initially present in an amount of about 15 mEq; methylprednisolone in an amount of about 1 g; cefazolin in an amount of about 1 g; ciprofloxacin in an amount of about 0.2 g; voriconazole in an amount of about 0.2 g.


In certain embodiments, the perfusion solution is maintained and provided to the lungs at a near physiologic temperature. According to one embodiment, the perfusion solution employs a blood product-based perfusion solution to more accurately mimic normal physiologic conditions. The perfusion solution may be supplemented with cellular media. The cellular media may include a blood product, such as whole blood, or packed red blood cells; allogenic packed red blood cells that are leukocyte depleted/reduced; donor's whole blood that is leukocyte and platelet depleted/reduced; and/or human plasma to achieve circulating hematocrit of 15-30%.


Overview of Method of Producing a Solution for Perfusing a Lung at Near Physiologic Temperature


In another aspect, a method of producing a solution for perfusing a lung at near physiologic temperature is provided. In a preferred method, the pre-weighed raw materials and WFI are added to a stainless steel mixing tank and mixed with heating until fully dissolved. The pH of the resulting solution is monitored and adjusted during the mixing process with 1M hydrochloric acid (HCl). The solution is allowed to cool and then filtered through a 0.2 μm filter and finally dispensed into a primary container. The filled container is terminally sterilized with heat using a sterilization cycle that has been validated to achieve a Sterility Assurance Level of 10−6. The raw materials in a preferred embodiment include a nutrient, a colloid, a vasodilator, a hormone and a steroid for perfusing a lung at near physiologic conditions.


In another preferred embodiment the raw materials include a nutrient including glucose monohydrate, sodium chloride, potassium chloride, a multi-vitamin including M.V.I. Adult® or equivalent; a colloid including dextran 40; a hormone including insulin; a steroid including methylprednisolone; buffering agents including disodium phosphate anhydrate, monopotassium phosphate and sodium bicarbonate; vasodilators including milrinone, nitroglycerin and magnesium sulfate anhydrate; an antimicrobial or antifungal agent.


In another preferred embodiment the raw materials include dextran 40; sodium chloride; potassium chloride; magnesium sulfate anhydrate; disodium phosphate anhydrate; monopotassium phosphate; glucose monohydrate; milrinone; nitroglycerin; insulin; a multi-vitamin (M.V.I. Adult® or equivalent); sodium bicarbonate; methylprednisolone (SoluMedrol® or equivalent); antimicrobial or antifungal agents including cefazolin, ciprofloxacin, and voriconazole for perfusing a lung at near physiologic conditions.


In a preferred embodiment, for each 1 L of solution, the raw materials include milrinone in an amount of about 4000 mcg; nitroglycerin in an amount of about 10-50 mg; dextran 40 in an amount of about 50 g; sodium chloride in an amount of about 8 g; potassium chloride in an amount of about 0.4 g; magnesium sulfate anhydrate in an amount of about 0.098 g; disodium phosphate anhydrate in an amount of about 0.046 g; monopotassium phosphate in an amount of about 0.063 g; glucose monohydrate in an amount of about 2 g; insulin in an amount of about 20 IU; a multi-vitamin (M.V.I. Adult® or equivalent) in the amount of about 1 unit vial; sodium bicarbonate is initially present in an amount of about 15 mEq; methylprednisolone in an amount of about 1 g; an antimicrobial or antifungal agent.


In another particularly preferred embodiment, for each 1 L of solution, the raw materials include milrinone in an amount of about 4000 mcg; nitroglycerin in an amount of about 10-50 mg; dextran 40 in an amount of about 50 g; sodium chloride in an amount of about 8 g; potassium chloride in an amount of about 0.4 g; magnesium sulfate anhydrate in an amount of about 0.098 g; disodium phosphate anhydrate in an amount of about 0.046 g; monopotassium phosphate in an amount of about 0.063 g; glucose monohydrate in an amount of about 2 g; insulin in an amount of about 20 IU; a multi-vitamin (M.V.I. Adult® or equivalent) in the amount of about 1 unit vial; sodium bicarbonate is initially present in an amount of about 15 mEq; methylprednisolone in an amount of about 1 g; cefazolin in an amount of about 1 g; ciprofloxacin in an amount of about 0.2 g; voriconazole in an amount of about 0.2 g.


Overview of Method of Flushing an Organ with a Solution Between Excise from the Donor and Instrumentation on OCS


In another aspect, there is provided a method of flushing an organ with a solution between excise from the body and instrumentation on OCS. In this embodiment, to prepare a donor lung for surgical removal from the donor's chest and to remove all old donor blood from the lung, the donor lung is flushed ante-grade using the pulmonary artery with the solution until the temperature of the donor lung is in the range of about 0 degrees C. to about 30 degrees C. Additionally, the solution may be used for retrograde flush of the lung using the pulmonary veins to remove any blood clots remaining in the donor lung prior to surgical removal of the lung from the donor's chest, and to ensure adequate homogenous distribution of flush solution to all lung segments. The lungs are ventilated using a ventilator during both ante-grade and retro-grade flushing to allow for homogenous distribution of the solution and to increase the oxygen concentration in the donor lung alveoli to minimize the impact of ischemia/reperfusion injury on the donor lung. Once the ante-grade and retrograde flushing of the donor lung is completed, the lung will be removed surgically while inflated to minimize collapsing of the alveoli. Once the donor lung is fully removed from the donor body, it is ready to the next phase of OCS perfusion.


In one embodiment, the solution comprises an energy-rich perfusion nutrient, a colloid, a hormone, a buffer, magnesium sulfate anhydrate, and a nitrate. In another embodiment, the solution comprises dextran 40; sodium chloride; potassium chloride; magnesium sulfate anhydrate; disodium phosphate anhydrate; monopotassium phosphate; glucose monohydrate; nitroglycerin.


In a particularly preferred embodiment each 1 L of solution for ante-grade flush comprises dextran 40 in an amount of about 50 g; sodium chloride in an amount of about 8 g; potassium chloride in an amount of about 0.4 g; magnesium sulfate anhydrate in an amount of about 0.098 g; disodium phosphate anhydrate in an amount of about 0.046 g; monopotassium phosphate in an amount of about 0.063 g; glucose monohydrate in an amount of about 2 g; nitroglycerin in an amount of about 50 mg.


In another particularly preferred embodiment each 1 L of solution for retrograde flush comprises dextran 40 in an amount of about 50 g; sodium chloride in an amount of about 8 g; potassium chloride in an amount of about 0.4 g; magnesium sulfate anhydrate in an amount of about 0.098 g; disodium phosphate anhydrate in an amount of about 0.046 g; monopotassium phosphate in an amount of about 0.063 g; glucose monohydrate in an amount of about 2 g; nitroglycerin in an amount of about 10 mg.


Overview of Method of Machine Perfusion Using Lung OCS Perfusion Solution


In another aspect, a method for machine perfusion of a donor lung is provided. The method includes perfusing the donor lung with a OCS lung perfusion solution comprising: dextran 40; sodium chloride; potassium chloride; magnesium sulfate anhydrate; disodium phosphate anhydrate; monopotassium phosphate; glucose monohydrate; milrinone; nitroglycerin; insulin; at least two vitamins; sodium bicarbonate; methylprednisolone (SoluMedrol® or equivalent); a microbial or antifungal agent.


In a further aspect, the method includes perfusing the donor lung with a particularly preferred OCS lung perfusion solution comprising for each 1 L of solution: milrinone in an amount of about 4000 mcg; nitroglycerin in an amount of about 10-50 mg; dextran 40 in an amount of about 50 g; sodium chloride in an amount of about 8 g; potassium chloride in an amount of about 0.4 g; magnesium sulfate anhydrate in an amount of about 0.098 g; disodium phosphate anhydrate in an amount of about 0.046 g; monopotassium phosphate in an amount of about 0.063 g; glucose monohydrate in an amount of about 2 g; insulin in an amount of about 20 IU; a multi-vitamin (M.V.I. Adult® or equivalent) in the amount of about 1 unit vial; sodium bicarbonate is initially present in an amount of about 15 mEq; methylprednisolone in an amount of about 1 g; cefazolin in an amount of about 1 g; ciprofloxacin in an amount of about 0.2 g; voriconazole in an amount of about 0.2 g.


Overview of the Lung Perfusion Circuit



FIG. 1 illustrates an exemplary lung perfusion circuit which can be used to circulate the perfusion solution noted above. The circuit is housed entirely within a lung perfusion module, and all its components may be disposable. The organ care system (OCS) disclosure, U.S. application Ser. No. 12/099,715, includes an exemplary embodiment of a lung perfusion circuit and is incorporated in its entirety by reference. Lung OCS perfusion solution 250 is placed in a reservoir and then circulates within the perfusion circuit, passing through various components of lung perfusion module before passing through the vascular system of lungs 404. Pump 226 causes perfusion solution 250 to flow around the lung perfusion circuit. It receives perfusion solution 250 from reservoir 224, and pumps the solution through compliance chamber 228 to heater 230. Compliance chamber 228 is a flexible portion of tubing that serves to refine the flow characteristics nature of pump 226. Heater 230 replaces heat lost by perfusion solution 250 to the environment during circulation of the fluid. In the described embodiment, the heater maintains perfusion solution 250 at or near the physiologic temperature of 30-37 degrees C., and preferably at about 34 degrees C. After passing through heater 230, perfusion solution 250 flows into gas exchanger 402. Gas exchanger 402 allows gases to be exchanged between gas and perfusion solution 250 via a gas-permeable, hollow fiber membrane. However, the gas exchanger has an effective gas exchange surface area of about 1 square meter, which is only a fraction of the 50-100 square meter effective exchange area of the lungs. Thus gas exchanger 402 has only a limited gas exchange capability compared to the lungs. Blood gas solenoid valve 204 regulates the supply of gas into gas exchanger 402. The composition of gas supplied to gas exchanger is determined by which mode the OCS is in. For example, when OCS 100 is in a sequential assessment mode, deoxygenation gas 500 from deoxygenation gas tank 501 is supplied to the gas exchanger. Sampling/injection port 236 facilitates the removal of a sample or the injection of a chemical just before perfusion solution 250 reaches the lungs. Perfusion solution then enters lungs 404 through cannulated pulmonary artery 232. Flow probe 114 measures the rate of flow of perfusion fluid 250 through the system. In the described embodiment, flow probe 114 is placed on the perfusate line as it leads towards the pulmonary artery. Pressure sensor 115 measures pulmonary arterial pressure at the point of entry of perfusion fluid 250 into the lungs. Oxygen probe 116 measures oxygen in perfusion fluid 250 just before it enters the lungs. In the described embodiment, perfusion solution 250 is the lung OCS solution described previously.



FIG. 2 is an overall view of OCS console 100 showing the single use, disposable lung perfusion module in a semi-installed position. As broadly indicated in FIG. 2, single use disposable lung perfusion module is sized and shaped to fit into OCS console 100, and to couple with it. Overall, the unit has a similar form to the organ care system described in U.S. patent application Ser. No. 11/788,865. Removable lung perfusion module 400, is insertable into OCS console 100 by means of a pivoting mechanism that allows module 400 to slide into the organ console module from the front, as shown in FIG. 2, and then pivot towards the rear of the unit. Clasp mechanism 2202 secures lung perfusion module 400 in place. In alternative embodiments, other structures and interfaces of lung perfusion module 400 are used to couple the module with OCS 100. When secured in place, electrical and optical connections (not shown) provide power and communication between OCS console 100 and lung perfusion module 400. Details of the electrical and optical connections are described in U.S. patent application Ser. No. 11/246,013, filed on Oct. 7, 2005, the specification of which is incorporated by reference herein in its entirety. A key component of lung perfusion module 400 is organ chamber 2204, which is described in detail below. Battery compartments 2206 and maintenance gas cylinder 220 (not shown) are located in the base of the OCS console 100. OCS console 100 is protected by removable panels, such as front panels 2208. Just below lung perfusion module are perfusion solution sampling ports 234 and 236. Mounted on top of OCS console 100 is OCS monitor 300.



FIG. 3 is a front view of lung perfusion module 400. Organ chamber 2204 includes a removable lid 2820 and housing 2802. Sampling ports, including LA sampling port 234 and PA sampling port 236 are visible below organ chamber 2802. Gas exchanger 402, bellows 418, and bellows plate 2502 are also visible in the figure.


The circulation path of the perfusion solution, which was first described in connection with FIG. 2, in terms of the components of lung perfusion module 400 is now addressed. Mounted below organ chamber 2204 are perfusion solution reservoir 224, which stores perfusion solution 250. The perfusion solution exits through one-way inflow valve 2306, line 2702, and pump dome 2704 to pump 226 (not shown). The perfusion solution is pumped through perfusion solution line 2404 through compliance chamber 228, and then to perfusion solution heater 230. After passing through heater 230, the perfusion solution passes through connecting line 2706 to gas exchanger 402.


The pulmonary artery (PA) cannula connects the perfusion circuit with the vascular system of lungs 404. An exemplary embodiment of a pulmonary artery (PA) cannula is shown in FIG. 4. Referring to FIG. 4, single PA cannula 802 has single insertion tube 804 for insertion into a single PA, and is used to cannulate the PA at a point before it branches to the two lungs. To connect the cannula to the pulmonary artery, insertion tube 804 is inserted into the PA, and the PA is secured onto the tube with sutures. The tracheal cannula 700 is inserted into the trachea to provide a means of connection between the lung perfusion module 400 gas circuit and the lungs. FIG. 5 illustrate an exemplary tracheal cannulae. Cannula 700 includes tracheal insertion portion 704 having an insertion portion tip diameter 702, to which the trachea is secured with a cable tie, or by other means. At the end of insertion portion 704 that is inserted into the trachea is rib 703; the rib helps secure insertion portion 704 at the inserted location within the trachea, and is secured with a cable tie placed around the trachea. At the opposite end of insertion portion 704, second rib 705, having a diameter about 0.2 inches greater than the base part diameter of insertion portion 704, acts as a stop for the silicone over-layer and as a stop for the trachea. The tracheal cannula may be clamped at flexible portion 706 prior to instrumentation to seal off air flow in and out of the lungs 404. Also illustrated is an optional locking nut 708.


The perfusion solution exits gas exchanger 402 through connecting line 2708 to the interface with the pulmonary artery. After flowing through the lung and exiting via the pulmonary vein and the left atrium, the perfusion solution drains through from the base of organ chamber 2204, as described below. These drains feed the perfusion solution to reservoir 224, where the cycle begins again.


Having described OCS console 100 and lung perfusion module 400, we now describe organ chamber 2204. FIG. 6 shows an exploded view of the components of organ chamber 2204. The top of organ chamber 2204 is covered with a sealable lid that includes front piece 2816, top piece 2820, inner lid with sterile drape (not shown), and sealing piece 2818 that seals front piece 2816 to top piece 2820. Base 2802 of chamber 2204 is shaped and positioned within lung perfusion module 400 to facilitate the drainage of the perfusion solution. Organ chamber 2204 has two drains, measurement drain 2804, and main drain 2806, which receives overflow from the measurement drain. Measurement drain 2804 drains perfusion solution at a rate of about 0.5 l/min, considerably less than perfusion solution 250 flow rate through lungs 404 of between 1.5 l/min and 4 l/min. Measurement drain leads to oxygen probe 118, which measures SaO2 values, and then leads on to reservoir 224. Main drain 2806 leads directly to reservoir 224 without oxygen measurement. Oxygen probe 118, which is a pulse oxymeter in the described embodiment, cannot obtain an accurate measurement of perfusion solution oxygen levels unless perfusion solution 250 is substantially free of air bubbles. In order to achieve a bubble-free column of perfusion solution, base 2802 is shaped to collect perfusion solution 250 draining from lungs 404 into a pool that collects above drain 2804. The perfusion solution pool allows air bubbles to dissipate before the perfusion solution enters drain 2804. The formation of a pool above drain 2804 is promoted by wall 2808, which partially blocks the flow of perfusion solution from measurement drain 2804 to main drain 2806 until the perfusion solution pool is large enough to ensure the dissipation of bubbles from the flow. Main drain 2806 is lower than measurement drain 2804, so once perfusion solution overflows the depression surrounding drain 2804, it flows around wall 2808, to drain from main drain 2806. In an alternate embodiment of the dual drain system, other systems are used to collect perfusion solution into a pool that feeds the measurement drain. In some embodiments, the flow from the lungs is directed to a vessel, such as a small cup, which feeds the measurement drain. The cup fills with perfusion solution, and excess blood overflows the cup and is directed to the main drain and thus to the reservoir pool. In this embodiment, the cup performs a function similar to that of wall 2808 in the embodiment described above by forming a small pool of perfusion solution from which bubbles can dissipate before the perfusion solution flows into the measurement drain on its way to the oxygen sensor.


Lungs 404 are supported by support surface 2810. The surface is designed to support lungs 404 without applying undue pressure, while angling lungs 404 slightly downwards towards the lower lobes to promote easy drainage of the perfusion solution. Support surface includes drainage channels 2812 to collect and channel perfusion solution issuing from lungs 404, and to guide the perfusion solution towards drain 2814, which feeds perfusion solution directly to the blood pool for measurement drain 2804. To provide additional support for the lungs, lungs 404 are wrapped with a polyurethane wrap (not shown) when placed on support surface 2810. The polyurethane wrap anchors lungs 404, helps keep the lungs in a physiologic configuration, and prevents the bronchi from being kinked and limiting the total volume of inflation. The wrap provides a smooth surface for the exterior of the lung to interface with organ chamber 2204, reducing the risk of the chamber applying excessive pressure on any part of lungs 404, which might cause undesirable hemorrhaging.



FIG. 7 is a schematic diagram of the described embodiment of a portable organ care system including the gas-related components of the lung perfusion module. The organ care system 1000 includes a permanent, multiple use, non-disposable section, OCS lung console 101, and a single use disposable section, lung perfusion module 400. Regulator 222/502 converts the gas tank pressure to 25 mm Hg for use in the system. Internal maintenance gas tank 221 contains a mixture that is designed to provide enough oxygen to maintain the lung tissue during maintenance mode. Pressure transducer 223 measures the pressure of internal maintenance gas in tank 221, so that the amount of gas remaining can be determined. Controller 202 manages the release of maintenance and assessment gases by controlling the valves, gas selector switch 216, and ventilator 214, thus implementing the preservation of the lungs in maintenance mode, or the assessment of the lungs in one of the assessment modes. Blood gas solenoid valve 204 controls the amount of gas flowing into blood gas exchanger 402. Airway pressure sensor 206 samples pressure in the airway of lungs 404, as sensed through isolation membrane 408. Relief valve actuator 207 is pneumatically controlled, and controls relief valve 412. The pneumatic control is carried out by inflating or deflating orifice restrictors that block or unblock the air pathway being controlled. This method of control allows complete isolation between the control systems in lung console module 200 and the ventilation gas loop in lung perfusion module 400. Pneumatic control 208 controls relief valve 207 and bellows valve actuator 210. Trickle valve 212 controls delivery of gas to the airway of lungs 404. Ventilator 214 is a mechanical device with an actuator arm that causes bellows 418 to contract and expand, which causes inhalation and exhalation of gas into and out of lungs 404. OCS monitor 300 provides user control of OCS 1000 via buttons, and displays data from the system's sensors that indicate the state of the lungs and of the various subsystems within OCS 1000. Monitor 300 is universal, i.e., it can be used for any organ. It includes monitor processor 302 that runs the software controlling monitor 300 and displays data on LCD 304. OCS monitor 300 includes four control buttons for the user: menu button 306 brings up the configuration menu; alarm button 308 silences the speaker; pump button 310 controls the circulatory pump; and action button 312 provides access to certain organ-specific actions, such as ventilator control, or to system actions, such as saving a session file to an external memory card. Other controls can also be included, such as a knob for controlling a value or selecting an item.


Use Models


An exemplary model for using the solution described above in the organ care system is described below.


The process of preparing the OCS perfusion module 400 for instrumentation begins by producing the solution by the method of producing a solution for perfusing a lung at near physiologic temperature as described previously. About 800 ml to about 2000 ml of the OCS lung perfusion solution is then added into the Organ Care System (OCS) sterile perfusion module 400. The solution is then supplemented with about 500 ml to about 1000 ml of cellular media. The cellular media may include one or combination of the following to achieve total circulating hematocrit concentration between 15-30%: typed allogenic packed red blood cells (pRBCs) that is leukocytes depleted/reduce; donor's whole blood that is leukocyte and platelet depleted/reduced; and/or human plasma to achieve circulating hematocrit of 15-30%. The OCS device operates to circulate and mix the solution and cellular media while warming and oxygenating the solution using a built in fluid warmer and gas exchanger 402. Once the solution is fully mixed, warmed and oxygenated, the pH of the solution will be adjusted using sodium bicarbonate or other available buffer solution as needed. Once the solution's hematocrit, temperature and pH levels reach an acceptable state, the donor lung will be instrumented on OCS.


Once the solution is fully mixed, pH is adjusted to 7.35-7.45 and hematocrit is adjusted to 15-30%, the donor lung will be instrumented on OCS. To begin instrumentation, first set the flow rate of the OCS Pump 226 to about 0.05 L/min. to ensure that perfusion solution does not exit the PA line 233 prior to connecting the trachea cannula 700. Place the lung in the OCS' organ chamber 224 and connect the trachea cannula 700 to the OCS trachea connector 710 and unclamp trachea cannula at section 706. Then connect a PA pressure monitoring line with pressure sensor 115, to the PA cannula 802, including pressure transducer connector 806. To connect the cannula to the pulmonary artery, insertion tube 804 is inserted into the PA, and the PA is secured onto the tube with sutures. Insertion tube 804 of cannula 802 connects to connector portion 805, which serves to position insertion tube 804 at an angle and location suitable for strain-free connection to the pulmonary artery of lungs 404. Connection portion 805 connects to main tube portion 808, which is attached to the perfusion fluid circuit. Trim the OCS' PA cannula 802 and prepare to connect to the OCS PA line connector 231. Next, increase the OCS' pump 226 flow to about 0.3 to about 0.4 L/min. so that a low-flow column of solution exits the PA line 233. Then remove any air from the lung by connecting the lung PA cannula 802 to the OCS PA line connector 231 and gradually filling the PA cannula 802 with perfusion solution. Once an air-free column of solution is reached inside the PA cannula 802, seal the connection between the PA cannula 802 and the OCS PA line connector 231.


Next, gradually raise the OCS fluid warmer 230 temperature to 37 degrees C., and bring the perfusion solution temperature from about 32 degrees C. to about 37 degrees C. Then begin increasing the pump flow gradually, ensuring that pulmonary arterial pressure (“PAP”) remains below 20 mmHg, until pulmonary flow rate reaches a target flow rate of at least 1.5 L/min. When the lung reaches a temperature of about 30 degrees C. to about 32 degrees C., begin OCS ventilation by turning the OCS ventilator 214 to “preservation” mode. The ventilator settings for instrumentation and preservation are specified in Table 1.









TABLE 1







Ventilator Settings (Instrumentation and Preservation)








Parameter
Requirement





Tidal Volume (TV)
= or <6 ml/kg


Respiratory Rate (RR)
10 breaths/min


Positive End Expiratory
7-8 cm H2O


Pressure (PEEP)
Note: decrease to 5 cmH2O after confirming



adequate inflation of lungs (within 2 hours)


I:E Ratio
1:2-1:3


Peak Airway Pressure
<25 cmH2O


(PAWP)









Next, gradually increase the perfusion and ventilation rate for up to about 30 minutes until reaching full ventilation and perfusion and allow ventilation parameters to stabilize. Once ventilation parameters of the donor lung on OCS have stabilized, wrap the lung to avoid over inflation injury to the donor lung ex-vivo. The lung may also be wrapped during “pause preservation” before beginning ventilation. During preservation of lung on OCS, ventilation settings are maintained as described in Table 1, the mean PAP is maintained under about 20 mmHg, and the pump flow is maintained at not less than about 1.5 L/min. Blood glucose, electrolytes and pH levels are monitored and adjusted within normal physiologic ranges by additional injections. Lung oxygenation function may be assessed using the OCS lung system in addition to lung compliance. In some instances it is desirable to provide therapy to the lung as described previously. Fiberoptic bronchoscopy may be performed for the donor lung ex-vivo on the OCS device. Once preservation and assessment of the donor lung on the OCS system is complete, the lung is cooled and removed from the OCS system to be transplanted into the recipient.


Donor lung cooling may be achieved by first shutting off the OCS pulsatile pump 226 and flush the donor lung with about 3 liters of perfusion solution at a temperature of about 0 degrees C. to about 15 degrees C. while continuing ventilation on the OCS system. Once the flush is complete the trachea 700 and pulmonary artery 802 cannulae may be disconnected from the OCS and the lung will be immersed in cold preservation solution until it is surgically attached to the recipient (transplanted). Alternatively, the entire system circulating OCS solution may be cooled down to 0 degrees C. to about 15 degrees C. using a heat-exchanger and cooling device while the lung is being ventilated on OCS. Once the target temperature of about 0 degrees C. to about 15 is achieved, the trachea 700 and pulmonary artery 802 cannulae will be disconnected from the OCS and the lung will be immersed in cold preservation solution until it is surgically attached to the recipient (transplanted).


The described system may utilize any embodiment of the lung OCS perfusion solution. In a preferred embodiment, the solution is mixed with red blood cells and placed into a system reservoir for use in the system.


It is to be understood that while the invention has been described in conjunction with the various illustrative embodiments, the forgoing 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. All references cited herein are incorporated by reference in their entirety and made part of this application.

Claims
  • 1. A method of perfusing a donor lung at or near physiologic conditions comprising: flushing the donor lung prior to excising the donor lung from a donor's body with at least one flushing solution including at least one nutrient, at least one colloid, at least one buffer, and magnesium sulfate anhydrate;excising the donor lung from the donor's body;placing the donor lung on an organ care system (OCS); andflowing a perfusion liquid through the donor lung, the perfusion liquid being at a physiologic temperature, the perfusion liquid comprising: a nutrient, a colloid, a hormone, a steroid, a buffer, magnesium sulfate anhydrate, an antimicrobial agent and at least one of a phosphodiesterase inhibitor or a nitrate.
  • 2. The method of claim 1, wherein in the perfusion liquid the nutrient includes glucose monohydrate, sodium chloride, potassium chloride, and a multi-vitamin; the colloid includes dextran 40; the hormone includes insulin; the steroid includes methylprednisolone; the buffer includes disodium phosphate anhydrate, monopotassium phosphate and sodium bicarbonate; phosphodiesterase inhibitor includes milrinone, and the nitrate includes nitroglycerin.
  • 3. The method of claim 2, wherein each liter of the perfusion liquid comprises milrinone in an amount of about 4000 mcg;nitroglycerin in an amount of about 10 mg to 50 mg;dextran 40 in the amount of about 50 g;sodium chloride in an amount of about 8 g;potassium chloride in an amount of about 0.4 g;magnesium sulfate anhydrate in an amount of about 0.098 g;disodium phosphate anhydrate in an amount of about 0.046 g;monopotassium phosphate in an amount of about 0.063 g;glucose monohydrate in an amount of about 2 g;insulin in an amount of about 20 IU;the multi-vitamin in an amount of about 1 unit vial;sodium bicarbonate in an amount of about 15 mEq; andmethylprednisolone in an amount of about 1 g.
  • 4. The method of claim 1 further comprising mixing the perfusion liquid with whole blood.
  • 5. The method of claim 1 further comprising mixing the perfusion liquid with red blood cells.
  • 6. The method of claim 1 further comprising mixing the perfusion liquid with leukocyte-depleted whole blood.
  • 7. The method of claim 1, further comprising ventilating the donor lung when flushing the donor lung prior to excising the donor lung to allow for homogenous distribution of the at least one flushing solution and to increase the oxygen concentration in the donor lung alveoli.
  • 8. A method of preserving a donor lung comprising: flushing the donor lung prior to excising the donor lung from a donor's body with at least one flushing solution including at least one nutrient, at least one colloid, at least one buffer, and magnesium sulfate anhydrate;excising the donor lung from the donor's body;placing the donor lung on an organ care system (OCS);perfusing the donor lung in a lung perfusion circuit of the OCS with a perfusion solution including at least one nutrient, at least one colloid, at least one buffer, and magnesium sulfate anhydrate to maintain the donor lung ex-vivo at a near physiological temperature and in a near physiologic functioning condition and to permit normal cellular metabolism in the donor lung, wherein the perfusion solution is supplemented with a circulating hematocrit of between 15% to 30%.
  • 9. The method of claim 8, wherein in the flushing solution: the at least one nutrient comprises glucose monohydrate, sodium chloride, and potassium chloride;the at least one colloid comprises dextran 40; andthe at least one buffer comprises disodium phosphate anhydrate and monopotassium phosphate.
  • 10. The method of claim 9, wherein in the flushing solution: the concentration of the glucose monohydrate is about 2 g/L;the concentration of the sodium chloride is about 8 g/L;the concentration of the potassium chloride is about 0.4 g/L;the concentration of the dextran 40 is about 50 g/L;the concentration of the disodium phosphate anhydrate is about 0.046 g/L;the concentration of the monopotassium phosphate is about 0.063 g/L; andthe concentration of the magnesium sulfate anhydrate is about 0.098 g/L.
  • 11. The method of claim 8, wherein the perfusion solution further comprises insulin, a multi-vitamin, sodium bicarbonate, methylprednisolone, milrinone, and nitroglycerin.
  • 12. The method of claim 11, wherein: the insulin is at a concentration of about 20 IU/L;the multi-vitamin is at a concentration of about 1 unit vial per liter;the sodium bicarbonate is at a concentration of about 15 mEq/L;the methylprednisolone is at a concentration of about 1 g/L;the milrinone is at a concentration of about 4000 mcg/L; andthe nitroglycerin is at a concentration of 10 mg/L to 50 mg/L.
  • 13. The method of claim 8, wherein the perfusion solution further comprises cefazolin, ciprofloxacin, and voriconazole.
  • 14. The method of claim 13, wherein: the cefazolin is at a concentration of about 1 g/L;the ciprofloxacin is at a concentration of about 0.2 g/L; andthe voriconazole is at a concentration of about 0.2 g/L.
  • 15. The method of claim 8, further comprising ventilating the donor lung when flushing the donor lung prior to excising the donor lung to allow for homogenous distribution of the at least one flushing solution and to increase the oxygen concentration in the donor lung alveoli.
  • 16. A method of preserving a donor lung comprising: flushing the donor lung antegrade prior to excising the donor lung from a donor's body with a first flushing solution;flushing the donor lung retrograde prior to excising the donor lung from a donor's body with a second flushing solution;excising the donor lung from the donor's body;placing the donor lung on an organ care system (OCS);perfusing the donor lung in a lung perfusion circuit of the OCS with a perfusion solution including at least one nutrient, at least one colloid, at least one buffer, and magnesium anhydrate to maintain the donor lung ex-vivo at a near physiological temperature and in a near physiologic functioning condition and to permit normal cellular metabolism in the donor lung, wherein the perfusion solution is supplemented with a circulating hematocrit of between 15% to 30%;wherein the first flushing solution and the second flushing solution each include at least one nutrient, at least one colloid, at least one buffer, and magnesium sulfate anhydrate; andwherein the first flushing solution comprises nitroglycerin at a concentration higher than a concentration of nitroglycerin in the second flushing solution.
  • 17. The method of claim 16, wherein the concentration of nitroglycerin in the first flushing solution is about 50 g/L.
  • 18. The method of claim 16, wherein the concentration of nitroglycerin in the second flushing solution is about 10 g/L.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional application of U.S. patent application Ser. No. 13/446,706 filed on Apr. 13, 2012, which claims the benefit under 35 U.S.C. § 119(e), of provisional application U.S. Ser. No. 61/475,524, filed on Apr. 14, 2011, entitled, “ORGAN CARE SOLUTION FOR EX-VIVO MACHINE PERFUSION OF DONOR LUNGS”, the entire subject matter of which is incorporated herein by reference. This application also incorporates by reference, the entirety of U.S. application Ser. No. 12/099,715, filed on Apr. 8, 2008, entitled, “SYSTEMS AND METHODS FOR EX VIVO LUNG CARE”.

US Referenced Citations (350)
Number Name Date Kind
3253595 Keller, Jr. et al. May 1966 A
3388803 Scott Jun 1968 A
3406531 Koski et al. Oct 1968 A
3468136 Koski et al. Sep 1969 A
3537956 Falcone Nov 1970 A
3545221 Koski et al. Dec 1970 A
3545605 Robins Dec 1970 A
3587567 Schiff Jun 1971 A
3607646 de Roissart Sep 1971 A
3632473 Belzer et al. Jan 1972 A
3639084 Goldhaber Feb 1972 A
3654085 Fritz et al. Apr 1972 A
3660241 Michielsen May 1972 A
3738914 Thorne et al. Jun 1973 A
3772153 De Roissart Nov 1973 A
3777507 Burton et al. Dec 1973 A
3843455 Bier et al. Oct 1974 A
3851646 Sarns Dec 1974 A
3881990 Burton et al. May 1975 A
3995444 Clark et al. Dec 1976 A
4004298 Freed Jan 1977 A
4069826 Sessions Jan 1978 A
4186253 Yokoyama et al. Jan 1980 A
4186565 Toledo-Pereyra Feb 1980 A
4231354 Kurtz et al. Nov 1980 A
4415556 Bretschneider Nov 1983 A
4598697 Numazawa et al. Jul 1986 A
4605644 Foker Aug 1986 A
4666425 Fleming May 1987 A
4719201 Foker Jan 1988 A
4723939 Anaise Feb 1988 A
4745759 Bauer et al. May 1988 A
4759371 Franetzki Jul 1988 A
4801299 Brendel et al. Jan 1989 A
4847470 Bakke Jul 1989 A
4920044 Bretan, Jr. Apr 1990 A
5051352 Martindale et al. Sep 1991 A
5066578 Wikman-Coffelt Nov 1991 A
5141847 Sugimachi et al. Aug 1992 A
5145771 Lemasters et al. Sep 1992 A
5157930 McGhee et al. Oct 1992 A
5200398 Strasberg et al. Apr 1993 A
5217860 Fahy et al. Jun 1993 A
5285657 Bacchi et al. Feb 1994 A
5306711 Andrews Apr 1994 A
5326706 Yland et al. Jul 1994 A
5338662 Sadri Aug 1994 A
5354268 Peterson et al. Oct 1994 A
5356593 Heiberger et al. Oct 1994 A
5356771 O'Dell Oct 1994 A
5358931 Rubinsky et al. Oct 1994 A
5362622 O'Dell et al. Nov 1994 A
5370989 Stern et al. Dec 1994 A
5381510 Ford et al. Jan 1995 A
5385821 O'Dell et al. Jan 1995 A
5395314 Klatz et al. Mar 1995 A
5405742 Taylor Apr 1995 A
5407669 Lindstrom et al. Apr 1995 A
5407793 Del Nido et al. Apr 1995 A
5472876 Fahy Dec 1995 A
5473791 Holcomb et al. Dec 1995 A
5494822 Sadri Feb 1996 A
5498427 Menasche Mar 1996 A
5505709 Funderburk et al. Apr 1996 A
5514536 Taylor May 1996 A
5552267 Stern et al. Sep 1996 A
5554123 Herskowitz Sep 1996 A
5554497 Raymond Sep 1996 A
5571801 Segall et al. Nov 1996 A
5584804 Klatz et al. Dec 1996 A
5586438 Fahy Dec 1996 A
5588816 Abbott et al. Dec 1996 A
5599173 Chen et al. Feb 1997 A
5599659 Brasile et al. Feb 1997 A
5613944 Segall et al. Mar 1997 A
5643712 Brasile Jul 1997 A
5654266 Chen et al. Aug 1997 A
5656420 Chien Aug 1997 A
5679565 Mullen et al. Oct 1997 A
5693462 Raymond Dec 1997 A
5698536 Segall et al. Dec 1997 A
5699793 Brasile Dec 1997 A
5702881 Brasile et al. Dec 1997 A
5716378 Minten Feb 1998 A
5723281 Segall et al. Mar 1998 A
5733894 Segall et al. Mar 1998 A
5747071 Segall et al. May 1998 A
5752929 Klatz et al. May 1998 A
5759148 Sipin Jun 1998 A
5770149 Raible Jun 1998 A
5776063 Dittrich et al. Jul 1998 A
5786136 Mayer Jul 1998 A
5787544 Meade Aug 1998 A
5807737 Schill et al. Sep 1998 A
5823799 Tor et al. Oct 1998 A
5843024 Brasile Dec 1998 A
5856081 Fahy Jan 1999 A
5882328 Levy et al. Mar 1999 A
5965433 Gardetto et al. Oct 1999 A
5998240 Hamilton et al. Dec 1999 A
6024698 Brasile Feb 2000 A
6034109 Ramasamy et al. Mar 2000 A
6042550 Haryadi et al. Mar 2000 A
6046046 Hassanein Apr 2000 A
6050987 Rosenbaum Apr 2000 A
6090776 Kuberasampath et al. Jul 2000 A
6100082 Hassanein Aug 2000 A
6110139 Loubser Aug 2000 A
6110504 Segall et al. Aug 2000 A
6144444 Haworth et al. Nov 2000 A
6168877 Pedicini et al. Jan 2001 B1
6217546 Hinchliffe et al. Apr 2001 B1
6365338 Bull et al. Apr 2002 B1
6375611 Voss et al. Apr 2002 B1
6375613 Brasile Apr 2002 B1
6389308 Shusterman May 2002 B1
6402461 Tebby Jun 2002 B1
6475716 Seki Nov 2002 B1
6490880 Walsh Dec 2002 B1
6492103 Taylor Dec 2002 B1
6492745 Colley, III et al. Dec 2002 B1
6524785 Cozzone et al. Feb 2003 B1
6526974 Brydon et al. Mar 2003 B1
6569615 Thatte et al. May 2003 B1
6582375 Melvin et al. Jun 2003 B2
6582953 Brasile Jun 2003 B2
6600941 Khuri Jul 2003 B1
6609987 Beardmore Aug 2003 B1
6631830 Ma et al. Oct 2003 B2
6642045 Brasile Nov 2003 B1
6673594 Owen et al. Jan 2004 B1
6696238 Murphy et al. Feb 2004 B2
6740484 Khirabadi et al. May 2004 B1
6764462 Risk, Jr. et al. Jul 2004 B2
6783328 Lucke et al. Aug 2004 B2
6792309 Noren Sep 2004 B1
6794124 Steen Sep 2004 B2
6811965 Vodovotz et al. Nov 2004 B2
6837851 Coroneo Jan 2005 B1
6878339 Akiyama et al. Apr 2005 B2
6894690 Capers May 2005 B2
6906325 Quek Jun 2005 B2
6925324 Shusterman Aug 2005 B2
6953655 Hassanein et al. Oct 2005 B1
6974436 Aboul-Hosn et al. Dec 2005 B1
7001354 Suzuki et al. Feb 2006 B2
7008380 Rees et al. Mar 2006 B1
7045279 Laske et al. May 2006 B1
7122371 Ma Oct 2006 B1
7238165 Vincent et al. Jul 2007 B2
7316666 Entenman et al. Jan 2008 B1
7410474 Friend et al. Aug 2008 B1
7431727 Cole et al. Oct 2008 B2
7452711 Daykin Nov 2008 B2
7572622 Hassanein et al. Aug 2009 B2
7651835 Hassanein et al. Jan 2010 B2
7811808 Van Der Plaats et al. Oct 2010 B2
8167869 Wudyka May 2012 B2
8304181 Hassanein et al. Nov 2012 B2
8323954 Kravitz et al. Dec 2012 B2
8409846 Hassanein et al. Apr 2013 B2
8420380 Fishman et al. Apr 2013 B2
8465970 Hassanein et al. Jun 2013 B2
8535934 Hassanein et al. Sep 2013 B2
8585380 Hassanein et al. Nov 2013 B2
8715305 Pate et al. May 2014 B2
8822203 Hassanein et al. Sep 2014 B2
9055740 Hassanein et al. Jun 2015 B2
9215867 Hassanein et al. Dec 2015 B2
9457179 Hassanein et al. Oct 2016 B2
9462802 Fishman et al. Oct 2016 B2
9706768 Freed et al. Jul 2017 B2
9894894 Hassanein et al. Feb 2018 B2
10076112 Hassanein et al. Sep 2018 B2
10124093 Francis et al. Nov 2018 B1
10321676 Hassanein et al. Jun 2019 B2
10327441 Freed et al. Jun 2019 B2
10362780 Kay et al. Jul 2019 B2
10433539 White et al. Oct 2019 B2
10736314 Hassanian et al. Aug 2020 B2
10750738 Hassanian et al. Aug 2020 B2
11122795 Hassanian et al. Sep 2021 B2
11154050 Hassanein et al. Oct 2021 B2
11191263 Hassanein et al. Dec 2021 B2
11570985 Hassanein et al. Feb 2023 B2
11723357 Hassanein et al. Aug 2023 B2
11785939 Freed Oct 2023 B2
20010003652 Freeman Jun 2001 A1
20010018569 Erbel et al. Aug 2001 A1
20010025191 Montgomery Sep 2001 A1
20020012988 Brasile Jan 2002 A1
20020102720 Steen Aug 2002 A1
20020132220 Berens et al. Sep 2002 A1
20020151950 Okuzumi Oct 2002 A1
20020164795 Gen Nov 2002 A1
20020177117 Wolf Nov 2002 A1
20020187132 Mcgregor et al. Dec 2002 A1
20020198504 Risk et al. Dec 2002 A1
20030040665 Khuri et al. Feb 2003 A1
20030050689 Matson Mar 2003 A1
20030053998 Daemen et al. Mar 2003 A1
20030073227 Hull et al. Apr 2003 A1
20030073912 Melvin et al. Apr 2003 A1
20030074760 Keller Apr 2003 A1
20030086830 Haywood et al. May 2003 A1
20030111604 Quek Jun 2003 A1
20030124503 Olivencia-Yurvati et al. Jul 2003 A1
20030135152 Kollar et al. Jul 2003 A1
20030147466 Liang Aug 2003 A1
20030168064 Daly et al. Sep 2003 A1
20040015042 Vincent et al. Jan 2004 A1
20040017658 Lo et al. Jan 2004 A1
20040018966 Segall et al. Jan 2004 A1
20040029096 Steen Feb 2004 A1
20040038192 Brasile Feb 2004 A1
20040058432 Owen et al. Mar 2004 A1
20040082057 Alford et al. Apr 2004 A1
20040086578 Segall et al. May 2004 A1
20040102415 Thatte et al. May 2004 A1
20040102678 Haindl May 2004 A1
20040106958 Mathis et al. Jun 2004 A1
20040110800 Bril et al. Jun 2004 A1
20040115689 Augello et al. Jun 2004 A1
20040138542 Khuri et al. Jul 2004 A1
20040168341 Petersen et al. Sep 2004 A1
20040170950 Prien Sep 2004 A1
20040171138 Hassanein et al. Sep 2004 A1
20040193096 Cooper Sep 2004 A1
20040202993 Poo et al. Oct 2004 A1
20040221719 Wright et al. Nov 2004 A1
20040224298 Brassil et al. Nov 2004 A1
20040235142 Schein et al. Nov 2004 A1
20040236170 Kim Nov 2004 A1
20040248281 Wright et al. Dec 2004 A1
20040258745 Kai et al. Dec 2004 A1
20050010118 Toyoda et al. Jan 2005 A1
20050019917 Toledo-Pereyra et al. Jan 2005 A1
20050027237 Weiner Feb 2005 A1
20050037330 Fischer et al. Feb 2005 A1
20050063860 Carpenter et al. Mar 2005 A1
20050085762 Vijay et al. Apr 2005 A1
20050142532 Poo et al. Jun 2005 A1
20050147958 Hassanein et al. Jul 2005 A1
20050153271 Wenrich Jul 2005 A1
20050170019 Roth Aug 2005 A1
20050182349 Linde et al. Aug 2005 A1
20050187469 Phillips Aug 2005 A1
20050202394 Dobson Sep 2005 A1
20050253390 Blazek Nov 2005 A1
20050255442 Brassil et al. Nov 2005 A1
20060034941 Dobson Feb 2006 A1
20060039870 Turner Feb 2006 A1
20060074470 Bartels et al. Apr 2006 A1
20060121438 Toledo-Pereyra et al. Jun 2006 A1
20060124130 Bonassa Jun 2006 A1
20060134073 Naka et al. Jun 2006 A1
20060148062 Hassanein et al. Jul 2006 A1
20060154357 Hassanein et al. Jul 2006 A1
20060154358 Hassanein et al. Jul 2006 A1
20060154359 Hassanein et al. Jul 2006 A1
20060160204 Hassanein et al. Jul 2006 A1
20060166360 Berthiaume et al. Jul 2006 A1
20060182722 Hering et al. Aug 2006 A1
20060292544 Hassanein et al. Dec 2006 A1
20070009881 Arzt et al. Jan 2007 A1
20070098694 Khuri et al. May 2007 A1
20070135752 Domash et al. Jun 2007 A1
20070135760 Williams Jun 2007 A1
20070196461 Weers Aug 2007 A1
20070275364 Hassanein et al. Nov 2007 A1
20080009815 Grabenkort et al. Jan 2008 A1
20080017191 Davies et al. Jan 2008 A1
20080017194 Hassanein Jan 2008 A1
20080057488 Steen Mar 2008 A1
20080234768 Hassanein et al. Sep 2008 A1
20080286746 Poo et al. Nov 2008 A1
20080295839 Habashi Dec 2008 A1
20090142830 Yamashiro Jun 2009 A1
20090143417 Smith et al. Jun 2009 A1
20090182302 Garabet Jul 2009 A1
20090191614 Miyahara Jul 2009 A1
20090197240 Fishman et al. Aug 2009 A1
20090197241 Fishman et al. Aug 2009 A1
20090197292 Fishman et al. Aug 2009 A1
20090197324 Fishman et al. Aug 2009 A1
20090197325 Fishman et al. Aug 2009 A1
20090215022 Page et al. Aug 2009 A1
20090312724 Pipkin et al. Dec 2009 A1
20100028850 Brassil Feb 2010 A1
20100056966 Toth Mar 2010 A1
20100092939 Belous et al. Apr 2010 A1
20100119554 Dobson May 2010 A1
20100204663 Wudyka Aug 2010 A1
20100322826 Locascio et al. Dec 2010 A1
20100322862 Ruoslahti et al. Dec 2010 A1
20110002926 Matthews et al. Jan 2011 A1
20110076666 Brassil Mar 2011 A1
20110129810 Owen et al. Jun 2011 A1
20110136096 Hassanein et al. Jun 2011 A1
20110177487 Simsir et al. Jul 2011 A1
20110190572 Brophy et al. Aug 2011 A1
20110212431 Bunegin et al. Sep 2011 A1
20110294108 Argoudelis et al. Dec 2011 A1
20120064050 Calle et al. Mar 2012 A1
20120077771 Fallouh et al. Mar 2012 A1
20120183945 Steen et al. Jul 2012 A1
20120277681 Kravitz et al. Nov 2012 A1
20120282591 Thatte et al. Nov 2012 A1
20120330438 Keshavjee et al. Dec 2012 A1
20130011823 Hassanein et al. Jan 2013 A1
20130078710 Hassanein et al. Mar 2013 A1
20130102917 Colbaugh et al. Apr 2013 A1
20130144227 Locke et al. Jun 2013 A1
20130157248 Fishman et al. Jun 2013 A1
20130220325 Davis et al. Aug 2013 A1
20130295552 Hassanein et al. Nov 2013 A1
20140001745 Lehmann et al. Jan 2014 A1
20140007961 Steen et al. Jan 2014 A1
20140017658 Steinman et al. Jan 2014 A1
20140017660 Steinman et al. Jan 2014 A1
20140135738 Panian May 2014 A1
20140220550 Van Der Plaats et al. Aug 2014 A1
20140283828 Acker et al. Sep 2014 A1
20140308654 Kay et al. Oct 2014 A1
20140315175 Nguyen et al. Oct 2014 A1
20140377849 Kay et al. Dec 2014 A1
20150017710 Freed et al. Jan 2015 A1
20150079580 Hassanein et al. Mar 2015 A1
20150093738 Potenziano et al. Apr 2015 A1
20150230453 Fontes et al. Aug 2015 A1
20150246164 Heaton et al. Sep 2015 A1
20150275176 Kobayashi et al. Oct 2015 A1
20150342177 Hassanein et al. Dec 2015 A1
20160113269 Woodard et al. Apr 2016 A1
20160262634 Steen et al. Sep 2016 A1
20160361476 Huang Dec 2016 A1
20170000110 Korkut et al. Jan 2017 A1
20170015963 Ott Jan 2017 A1
20170042141 Kay et al. Feb 2017 A1
20170049096 Kay et al. Feb 2017 A1
20190021308 Hassanein et al. Jan 2019 A1
20200128813 Kay et al. Apr 2020 A1
20200337298 Hassanein et al. Oct 2020 A1
20200352155 Fishman et al. Nov 2020 A1
20210244017 Ritchie et al. Aug 2021 A1
20220039373 Hassanein et al. Feb 2022 A1
20220071197 Hassanein et al. Mar 2022 A1
20220232823 Hassanein et al. Jul 2022 A1
20230210104 Hassanein Jul 2023 A1
20230263156 Hassanein Aug 2023 A1
Foreign Referenced Citations (101)
Number Date Country
2144952 Mar 1994 CA
2881613 Nov 2007 CA
2861545 Jul 2013 CA
2521324 Dec 2014 CA
1232723 Oct 1999 CN
1269471 Oct 2000 CN
2418882 Feb 2001 CN
1452863 Nov 2003 CN
2616058 May 2004 CN
1849102 Oct 2006 CN
103893205 Jul 2014 CN
203724222 Jul 2014 CN
104770361 Jul 2015 CN
114375945 Apr 2022 CN
4201259 Jul 1993 DE
10121159 Nov 2002 DE
102005048625 Apr 2007 DE
0347923 Dec 1989 EP
0376763 Jul 1990 EP
1062870 Jan 2003 EP
1488743 Dec 2004 EP
1017271 Jan 2006 EP
3347084 Nov 2020 EP
4032401 Jul 2022 EP
2830183 Apr 2003 FR
S57-010695 Jan 1982 JP
63270601 Nov 1988 JP
H02-282301 Nov 1990 JP
02-306901 Dec 1990 JP
H03-74302 Mar 1991 JP
04-099701 Mar 1992 JP
H04-128201 Apr 1992 JP
06-056601 Mar 1994 JP
06-305901 Nov 1994 JP
H07-196401 Aug 1995 JP
H08-89518 Apr 1996 JP
08-511012 Nov 1996 JP
2001516768 Oct 2001 JP
2003-206201 Jul 2003 JP
2003-315220 Nov 2003 JP
2004513889 May 2004 JP
2004525290 Aug 2004 JP
2004529938 Sep 2004 JP
2008-515914 May 2008 JP
2009-521931 Jun 2009 JP
2010-525076 Jul 2010 JP
2011-511000 Apr 2011 JP
2016-053030 Apr 2016 JP
6144238 Jun 2017 JP
6625384 Dec 2019 JP
6756775 Sep 2020 JP
WO-8805261 Jul 1988 WO
WO-9502326 Jan 1995 WO
WO-9503680 Feb 1995 WO
WO-9531897 Nov 1995 WO
WO-9618293 Jun 1996 WO
WO-9629865 Oct 1996 WO
WO-9722244 Jun 1997 WO
WO-9746091 Dec 1997 WO
WO-9915011 Apr 1999 WO
WO-0018226 Apr 2000 WO
WO-0022927 Apr 2000 WO
WO-200027189 May 2000 WO
WO-0035340 Jun 2000 WO
WO-0060936 Oct 2000 WO
WO-200101774 Jan 2001 WO
WO-0226034 Apr 2002 WO
WO-0235929 May 2002 WO
WO-02089571 Nov 2002 WO
WO-2003026419 Apr 2003 WO
WO-2004017838 Mar 2004 WO
WO-2004026031 Apr 2004 WO
WO-2006042138 Apr 2006 WO
WO-2006060309 Jun 2006 WO
WO-2006076590 Jul 2006 WO
WO-2006124820 Nov 2006 WO
WO-2007079185 Jul 2007 WO
WO-2007124044 Nov 2007 WO
WO-2008106724 Sep 2008 WO
WO-2008108996 Sep 2008 WO
WO-08150587 Dec 2008 WO
WO-2009099939 Aug 2009 WO
WO-2011002926 Jan 2011 WO
WO-2011072012 Jun 2011 WO
WO-2012142487 Oct 2012 WO
WO-2012148685 Nov 2012 WO
WO-2013068751 May 2013 WO
WO-2013068752 May 2013 WO
WO-2013068753 May 2013 WO
WO-2013106908 Jul 2013 WO
WO-2014011547 Jan 2014 WO
WO-2014059316 Apr 2014 WO
WO-2014194349 Dec 2014 WO
WO-2015126853 Aug 2015 WO
WO-2015143552 Oct 2015 WO
WO-2015154170 Oct 2015 WO
WO-2015154193 Oct 2015 WO
WO-2015187737 Dec 2015 WO
WO-2016090498 Jun 2016 WO
WO-2017044465 Mar 2017 WO
WO-2017205967 Dec 2017 WO
Non-Patent Literature Citations (285)
Entry
US 11,758,904 B2, 09/2023, Freed (withdrawn)
Gever et al. “Technique to Repair Damaged Donor Lungs for Graft Passes Clinical Test” Medpage Today (Year: 2008).
Wei et al. “Protective Effect of Specific Phosphodiesterease Inhibitor Milrinone for Donor Lung” Chinese Journal of New Drugs, vol. 16, Nov. 21, 2007.
Keshavjee et al. “The role of dextran 40 and potassium in extended hypothermic lung preservation for transplantation.” J. Thorac Cardiovasc Surg. Feb. 1992; 103(2): 314-25, (Year: 1992).
“2002 Design & Engineering Awards, Portable Organ Preservation System”, Science (2002) (1 page).
“Celsior™ Cold Storage Solution”, Sangstat Medical Corporation (internet reference) (1999) (5 pages).
“Heart Kept Beating Outside Body,” Associated Press, CNN.com (2001).
“History of Transplantation and Organ Preservation,” Barr Laboratories, Inc. (internet reference) (2004).
“Human heart beats on its own outside body”, USA Today (2001) (1 page).
“Human Heart Kept Alive Outside Body for First Time in Study of Portable Organ Preservation System™ at University of Pittsburgh Medical Center”, UPMC, McGowan Institute for Regenerative Medicine (2001) (2 pages).
“Machine Keeps Human Kidney Alive for 24-Hours,” www.worldhealth.net, Aug. 25, 2001.
“Machine May Be Organ Transplant Breakthrough,” USA Today (2001).
“New Discovery in Organ Transplantation,” MSNBC, 2001 (1 pages).
“The Nation: Warm-Storage Device May Aid Organ Transplants”, Dow Jones Publications Library (2001) (1 page).
“ViaSpan (Belzer UW) Cold Storage Solution,” Barr Laboratiories, Inc. (2002) (2 pages).
“Warm storage for donor organs”, University of Chicago Magazine (2001) (1 page).
Ahmad, N. et al., “A pathophysiologic study of the kidney tubule to optimize organ preservation solutions”, Kidney International 66(1):77-90 (2004), 14 pages.
Aitchison, J.D. et al., “Functional assessment of non-heart-beating donor lungs: prediction of post-transplant function”, European Journal of Cardio-thoracic Surgery, 20:187-194 (2001) (8 pages).
Aitchison, J.D. et al., “Nitric Oxide During Perfusion Improves Posttransplantation Function of Non-Heart-Beating Donor Lungs”, Transplantation, 75(12):1960-1964, Jun. 27, 2003, 5 pages.
Ananthaswamy “Machine Keeps Organs Alive for Longer,” New Scientist.com (2001).
Andreasson, et al., “Ex vivo lung perfusion in clinical lung transplantation—State of the art”, European Journal of Cardio-Thoracic Surgery, 46:779-788, 2014 (10 pages).
Aoki, M. et al., Anti-CD18 Attenuates Deleterious Effects of Cardiopulmonary Bypass and Hypothermic Circulatory Arrest in Piglets, J. Card. Surg. 10(Suppl):407-17 (1995) (11 pages).
Asparagine, Encyclopedia.com, https://www.encyclopedia.com/science-and-technology/biochemistry/biochemistry/asparagine, accessed May 7, 2022 (3 pages).
Baker, L.E. et al., “Artificial Maintenance Media for Cell and Organ Cultivation”, Journal of Experimental Medicine, 70:29-38, Jul. 1, 1939 (15 pages).
Bando et al. Oxygenated Perfluorocarbon, Recombinant Human Superoxide Dismutase, and Catalase Ameliorate Free Radical Induced Myocardial Injucy During Heart Preservation and Transplantation. J Thorac Cardiovasc Surg. 96:930-8(Dec. 1988).
Barinov, E.F., “Hormonal-metabolic disturbances during biological preservation of the heart”, Fiziologicheskii Zhurnal (Kiev), 29(3):293-299 (1983) (8 pages)—Russian Language with English Abstract.
Becker, et al., “Evaluating acellular versus cellular perfusate composition during prolonged ex vivo lung perfusion after initial cold ischaemia for 24 hours”, Transplant International, 29:88-97, 2016, published online Aug. 27, 2015 (10 pages).
Belzer, F.O., “Formula for Belzer MPS Solution”, University of Wisconsin-Madison Organ Preservation, (<http://www.surgery.wisc.edu/transplat/research/southard/BelzerMPS.shtml>) (Oct. 3, 2003) (2 pages).
Benichou et al. Canine and Human Liver Preservation for 6 to 18 Hr by Cold Infusion. Transplantation. 24(6):407-411(Dec. 1977).
Birkett, D. et al., “The Fatty Acid Content and Drug Binding Characteristics of Commercial Albumin Preparations”, Clinica Chimica Acta 85:253-258 (1978), 6 pages.
Blanchard et al. Techniques for Perfusion and Storage of Heterotopic Heart Transplants in Mice. Microsurgery. 6:169-174(1985).
Boggi, U. et al., “Pancreas Preservation with University of Wisconsin and Celsior Solutions”, Transplant Proceedings 36(3):563-565 (2004), 3 pages.
Boggi, U. et al., “Pancreas Preservation With University of Wisconsin and Celsior Solutions: A Single-Center, Prospective, Randomized Pilot Study”, Transplantation 27:77(8):1186-1190 (2004), 5 pages.
Botha, P., “Extended Donor Criteria in Lung Transplantation”, Current Opinion in Organ Transplantation, 14:206-210, 2009 (5 pages).
Boyle, Jr. et al. Ischemia-Reperfusion Injury. Ann. Thorac. Surg. 64:524-30 (1997).
Brandes, H. et al. “Influence of High Molecular Dextrans on Lung Function in an ex Vivo Porcine Lung Model,” Journal of Surgical Research, 101:2, 225-231 (2001) (7 pages).
Brasile, L. et al., “Organ Preservation Without Extreme Hypothermia Using an Oxygent™ Supplemented Perfusate”, Art. Cells, Blood Subs., and Immob. Biotech., 22(4):1463-68 (1994), 6 pages.
Burt et al., “Myocardial Function After Preservation for 24 Hours,” Jour. Thorac. and Cardiovascular Surg. 92(2):238-46 (1986).
Calhoon et al. Twelve-Hour Canine Heart Preservation With a Simple, Portable Hypothermic Organ Perfusion Device. Ann. Thorac. Surg. 62:91-3 (1996).
Canelo R, et al.; “Experience with Hystidine Tryptophan Ketoglutarate Versus University Wisconsin Preservation Solutions in Transplantation,” Int Surg. 88(3):145-51 (2003).
Carrier, B., “Chapter 4: Hypoxia and Oxygenation”, Alaska Air Medical Escort Training Manual, Fourth Edition, pp. 71-82, 2006, 12 pages.
Chambers, D.J. et al., “Long-Term Preservation of the Heart: The Effect of Infusion Pressure During Continuous Hypothermic Cardioplegia”, The Journal of Heart and Lung Transplantation, 11(4):665-75 (1992), 11 pages.
Chen, E. P. et al., “Milrinone Improves Pulmonary Hemodynamics and Right Ventricular Function in Chronic Pulmonary Hypertension”, Ann Thorac Surg, 63:814-821, 1997 (8 pages).
Chen, F. et al., “Development of New Organ Preservation Solutions in Kyoto University”, Yonsei Medical Journal, 46(6):1107-40 (2004), 8 pages.
Chien et al. Canine Lung Transplantation After More Than Twenty-four Hours of Normothermic Preservation. J. Heart Lung Transplant. 16:3340-51 (1997).
Chien, S. et al., “A simple technique for multiorgan preservation”, The Journal of Thoracic and Cardiovascular Surgery, 95(1):55-61 (1988), 7 pages.
Chien, S. et al., “Functional Studies of the Heart During a 24-Hour Preservation Using a New Autoperfusion Preparation”, The Journal of Heart and Lung Transplantation, 10(3):401-8 (1991), 8 pages.
Chinchoy, Edward Cheng-wey; “The Development, Refinement, and Uses of a Physiologically Working Isolated Ex Vivo Swine Heart Model”, A thesis submitted to the Faculty of the Graduate School of the University of Minnesota, Dec. 1999 (136 pages).
Christophi, C. et al., “A Comparison of Standard and Rapid Infusion Methods of Liver Preservation During Multi-Organ Procurement”, Aust. N.Z.J. Surg., 61(9):692-694 (1991), 3 pages.
Cimino, Adria, “Doctor develops device to preserve donated organs”, Mass High Tech (2001), 2 pages.
Collins, BH “Organ Transplantation: What Is the State of the Art?,” Ann Surg. 238(6 Suppl):S72-89 (2003).
Cronin et al., “Liver Transplantation at the University of Chicago,” Clin Transpl. 231-8 (1999).
Cysteine, Encyclopedia.com, https://www.encyclopedia.com/science-and-technology/biochemistry/biochemistry/cysteine, accessed May 7, 2022 (4 pages).
Daemen, J.H.C. et al., “Short-term outcome of kidney transplants from non-heart-beating donors after preservation by machine perfusion”, Transpl. Int. 9(Supp 1):S76-S80 (1996), 5 pages.
Definition of Examine, Merriam-Webster Dictionary on-line. www.merriam-webster.com/dictionary/examine, Printed Feb. 9, 2011, (1 page).
Demertzis, S. et al., “University of Wisconsin Versus St. Thomas′ Hospital Solution for Human Donor Heart Preservation”, Ann Thorac Surg 55:1131-7 (1993), 7 pages.
Den Butter, G. et al., “Comparison of solutions for preservation of the rabbit liver as tested by isolated perfusion”, Transpl. Int. 8(6):466-471 (1995), 6 pages.
Denham, B.S. et al., “Twenty-Four Hour Canine Renal Preservation By Pulsatile Perfusion, Hypothermic Storage, and Combinations of the Two Methods”, Transplantation Proceedings, 9(3):1553-1556 (1977), 4 pages.
Dobrian et al., “In vitro formation of oxidatively-modified and reassembled human low-density lipoproteins,” Biochimica et Biophysica Acta (BBA) 1169:12-24 (1993).
Drexler, H. et al., “Effect of L-arginine on coronary endothelial function in cardiac transplant recipients. Relation to vessel wall morphology,” Circulation 89(4):1615-1623 (1994) (10 pages).
Duarte, J.D. et al., “Pharmacologic treatments for pulmonary hypertension; exploring pharmacogenomics”, Future Cardiol., 9(3):335-349, 2013 (15 pages).
Egan, T. M. et al., “Ex Vivo Evaluation of Human Lungs for Transplant Suitability”, Ann Thorac Surg, vol. 81, No. 4, pp. 1205-1213 (Apr. 2006) (9 pages).
Eiseman et al., “A Disposable Liver Perfusion Chamber,” Surgery 6:1163-66 (1966).
Engelman,R.M. et al., “Influence of Steroids on Complement and Cytokine Generation After Cardiopulmonary Bypass”, Ann Thorac Surg 60(3):801-04 (1995) (4 pages).
European Commission, Scientific Committee on Food, “Opinion on Substances for Nutritional Purposes Which Have Been Proposed for Use in the Manufacture of Foods for Particular Nutritional Purposes (‘Parnuts’)”, SCF/CS/ADD/NUT/20/Final, http://www.europa.eu.int/comm/dg24/health/sc/scf/index_en.html, Dec. 5, 1999 (19 pages).
European Extended Search Report issued in EP 16844964.3, dated Apr. 26, 2019 (7 pages).
European Extended Search Report issued in EP20206681.7, dated Apr. 26, 2021 (8 pages).
European Extended Search Report issued in European Patent Application No. 22158928.6. dated Jun. 29, 2022 (13 pages).
European Search Report for European Patent Application No. 08795820.3 dated Apr. 17, 2014 (6 pages).
European Search Report for European Patent Application No. 09707471.0 dated May 27, 2014. 7 pages.
European Search Report issued for European Application No. EP19204566.4, dated May 25, 2020 (7 pages).
European Search Report issued in EP12770852.7, dated Sep. 23, 2014, 8 pages.
Extended European Search Report issued in EP15803127.8, dated May 22, 2018 (14 pages).
Extended European Search Report issued in European Application No. 17172411.5, dated Nov. 8, 2017 (7 pages).
Fabregas, Luis, “UPMC tests machine to aid heart transplants”, Pittsburg Tribune-Review (Feb. 24, 2002), (<http://www.pittsburghlive.com/x/pittsburghrib/print_19181.html>), 2 pages.
Faggian, G. et al., “Donor Organ Preservation in High-Risk Cardiac Transplantation”, Transplantation Proceedings 36:617-619 (2004), 3 pages.
FDA Premarket Approval 510k (extracts), “Perfadex Solution for Lung Perfusion”, dated Mar. 8, 2001 (61 pages).
FDA Premarket Approval 510k, “Perfadex with THAM”, dated Oct. 9, 2008 (5 pages).
FDA Summary of Safety and Probable Benefit, “XVIVO Perfusion System (XPS) with STEEN Solution Perfusate”, HUD Designation No. 08-0194, Notice of Approval dated Aug. 12, 2014 (52 pages).
Featherstone et al. “Comparison of Phosphodiesterase Inhibitors of Differing Isoenzyme Selectivity Addes to St. Thomas Hospital Cardioplegic Solution Used for Hypothermic Preservation of Rat Lungs.” Am. J. Respir. Crit. Care Med. Mar. 2000. 162(3):850-856.
Fehrenberg, C. et al., “Protective Effects of B2 Preservation Solution in Comparison to a Standard Solution (Histidine-Tryptophan-Ketoglutarate/Bretschneider) in a Model of Isolated Autologous Hemoperfused Porcine Kidney”, Nephron Physiol 96:52-58 (2004) (7 pages).
Ferrera et al. Comparison of Different Techniques of Hypothermic Pig Heart Preservation. Ann. Thorac. Surg. 57(5):1233-39 (1994).
File History for U.S. Appl. No. 60/616,835, filed Oct. 7, 2004 (82 pages).
File History for U.S. Appl. No. 60/694,971, filed Jun. 28, 2005 (280 pages).
File History for U.S. Appl. No. 60/725,168, filed Oct. 6, 2005 (699 pages).
Finn, A. et al., “Effects of Inhibition of Complement Activation Using Recombinant Soluble Complement Receptor 1 On Neutrophil CD11B/CD18 and L-Selectin Expression and Release of Interleukin-8 and Elastase in Simulated Cardiopulmonary Bypass”, J Thorac Cardiovasc Surg 111(2):451-459 (1996), 9 pages.
Fisher, et al., “An observational study of Donor Ex Vivo Lung Perfusion in UK lung transplantation: DEVELOP-UK”, Health Technology Assessment, vol. 20, No. 85, Nov. 2016 (310 pages).
Fourcade et al., “A New Method of Kidney Preservation with Collins′ Solution,” Biomed. 21(7):308-11 (1974).
Fraser, C.D. Jr. et al., “Evaluation of Current Organ Preservation Methods for Heart-Lung Transplantation”, Transplantation Proceedings, 20(1 Suppl. 1):987-990 (1988), 4 pages.
Gever, J., “Technique to Repair Damaged Donor Lungs for Graft Passes Clinical Test”, MedPage Today, https://www.medpagetoday.org/surgery/transplantation/12245, Accessed Jul. 11, 2020, dated Dec. 19, 2008 (4 pages).
Givertz, M.M. et al., “Effect of Bolus Milrinone on Hemodynamic Variables and Pulmonary Vascular Resistance in Patients With Severe Left Ventricular Dysfunction: A Rapid Test for Reversibility of Pulmonary Hypertension”, JACC, 28(7):1775-1780, Dec. 1996 (6 pages).
Glucose, The Merck Index, 11th ed. Entry 4353 (pp. 699-700) (1989), 3 pages.
Glutamine, Encyclopedia.com, https://www.encyclopedia.com/science-and-technology/biochemistry/biochemistry/glutamine, accessed May 7, 2022 (7 pages).
Gohrbandt, B., et al., “Glycine intravenous donor preconditioning is superior to glycine supplementation to low-potassium dextran flush preservation and improves graft function in a large animal lung transplantation model after 24 hours of cold ischemia”, The Journal of Thoracic and Cardiovascular Surgery, 131(3):724-729, Mar. 2006 (6 pages).
Grynberg, A. et al., “Fatty Acid Oxidation in the Heart”, Journal of Cardiovascular Pharmacology, 28(Suppl. 1):S11-S17 (1996) (8 pages).
Guarrera, J.V. et al., “Pulsatile Machine Perfusion With Vasosol Solution Improves Early Graft Function After Cadaveric Renal Transplantation”, Transplantation 77(8):1264-1268 (2004), 5 pages.
Gundry, S.R. et al., “Successful Transplantation of Hearts Harvested 30 Minutes After Death From Exsanguination”, Ann Thorac Surg 53(5):772-775 (1992), 4 pages.
Habazetti, H. et al., “Improvement in Functional Recovery of the Isolated Guinea IG Heart After Hyperkalemic Reperfusion With Adenosine”, J Thorac Cardiovasc Surg 111(1):74-84 (1996) (11 pages).
Hachida, M. et al., Abstract “Efficacy of myocardial preservation using HTK solution in continuous 120 min cross-clamping method—a comparative study with GIK method”, Nippon Kyobu Geka Gakkai Zasshi. 41(9):1495-1501 (1993), 1 page.
Hai, Human Body Atlas, First Edition, Liaoning Science and Technology Publishing House, p. 120, Oct. 31, 2011 (3 pages)—with English Translation.
Han, B. et al., “Study on the clinical efficacy of specific phosphodiesterase inhibitor in patients with pulmonary hypertension due to left heart disease”, Experimental and Therapeutic Medicine, 16:1175-1186, 2018 (12 pages).
Hardesty, R.L. et al., Original Communications, “Autoperfusion of the heart and lungs for preservation during distant procurement”, J Thorac Cardiovasc Surg, 93:11-18 (1987) (8 pages).
Hartman, J. C. The Role of Bradykinin and Nitric Oxide in the Cardioprotective Action of ACE Inhibitors. Ann. Thor. Surg. 60:789-92 (1995).
Hassanein, W.H. et al., “A Novel Approach for 12 Hour Donor Heart Preservation, Presented at the 70th Scientific Sessions of The American Heart Association”, Abstract was published in Circulation (1997), 1 page.
Hassanein, W.H. et al., “Continuous Perfusion of Donor Hearts in the Beating State Extends Preservation Time and Improves Recovery of Function”, The Journal of Thoracic and Cardiovascular Surgery, pp. 821-830 (1998), 10 pages.
Heil, J.E. et al., “A Controlled Comparison of Kidney Preservation by Two Methods: Machine Perfusion and Cold Storage”, Transplantation Proceedings 19(1):2046 (1987), 1 page.
Hoeper, M.M. et al., “Intensive Care Unit Management of Patients with Severe Pulmonary Hypertension and Right Heart Failure”, Am J Respir Crit Care Med, 184:1114-1124, 2011 (11 pages).
Howarth, F.C. et al., “Effects of extracellular magnesium and beta adrenergic stimulation on contractile force and magnesium mobilization in the isolated rat heart”, Magnesium Research, 7:187-197, 1994 (13 pages).
Hui-Li, G. “The Management of Acute Pulmonary Arterial Hypertension”, Cardiovascular Therapeutics, 29:153-175, 2011 (23 pages).
Hülsmann et al. “Loss of cardiac contractility and severe morphologic changes by acutely lowering the pH of the perfusion medium: protection by fatty acids,” BBAGEN 20256, Biochimica et Biophysica Acta., 1033:214-218 (1990) (5 pages).
Ida, K. “Titanium for Medical and Dental Use”, Japanese journal of medical electronics and biological engineering, 24(1):47-54, 1986 (12 pages)—with English Summary.
Imber et al.; “Advantages of Normothermic Perfusion Over Cold Storage in Liver Preservation,” Transplantation 73(5):701-09 (2002).
Ingemansson, et al., “Importance of Calcium in Long-Term Preservation of the Vasculature”, Ann Thorac Surg, 61:1158-1162, 1996 (5 pages).
International Search Report and Written Opinion for International Application No. PCT/US2012/033626 dated Sep. 20, 2012 (12 pages).
International Search Report and Written Opinion issued by the U.S. Patent and Trademark Office as Searching Authority, in International Application No. PCT/US16/50512, dated Dec. 12, 2016 (9 pages).
International Search Report, issued by the European Patent Office as Searching Authority, in PCT/US07/009652 International Search Report, dated Apr. 18, 2008, 5 pages.
International Search Report, issued by the European Patent Office as Searching Authority, issued in PCT/US98/19912, dated May 3, 1999 (4 pages).
International Search Report, issued by the U.S. Patent Office as Searching Authority, issued in PCT/US08/61454 International search report dated Dec. 5, 2008 (3 pages).
International Search Report, issued by the U.S. Patent Office as Searching Authority, issued in PCT/US09/032619, dated Jun. 4, 2009 (4 pages).
Janßen, H. et al., “UW is Superior to Celsior and HTK in the Protection of Human Liver Endothelial Cells Against Preservation Injury”, Liver Transplantation, 10(12):1514-1523 (2004), 10 pages.
Jaski, B.E. et al., “Positive inotropic and vasodilator actions of milrinone in patients with severe congestive heart failure. Dose-response relationships and comparison to nitroprusside”, J. Clin Invest., 75(2):643-649, 1985 (8 pages).
Jirsch, D.W. et al., “Ex Vivo Evaluation of Stored Lungs”, The Annals of Thoracic Surgery, 10(2):163-168, Aug. 1970 (6 pages).
Johnson, Kerry et al: “POPS: Portable Organ Preservation System.” UPMC Health System and TransMedics, Inc. Tribune Review (No date) 1 page.
Johnston, R., “What's Normal About DLCO?”, PFT Blog, Jan. 1, 2014 (17 pages).
Kawakami, et al., “Successful Preservation of the Isolated Canine Heart for 24 Hours by Low Pressure-Low Temperature Continuous Perfusion”, Japanese Annals of Thoracic Surgery, Japan, 7(6):543-547, Dec. 25, 1987 (13 pages)—English Translation.
Kawamura, T. et al., “Long-Term Preservation of Canine Pancreas By a New Simple Cold Storage Method Using Perfluorochemical—The Two-Layer Cold Storage Method (Euro-Collins′ Solution/Perfluorochemical)-”, Kobe J. Med. Sci., 38(2):135-145 (1992), 11 pages.
Kelly “Current Strategies in Lung Preservation,” J. Lab Clin. Med. 136:427-40 (2000).
Keshavjee, S.H. et al., “A method for safe twelve-hour pulmonary preservation”, J Thorac Cardiovasc Surg, 98:529-534 (1989), 6 pages.
Kioka, Y. et al., “Twenty-Four-Hour Isolated Heart Preservation by Perfusion Method With Oxygenated Solution Containing Perfluorochemicals and Albumin”, The Journal of Heart Transplantation, 5:437-443 (1986), 7 pages.
Koike, et al., “An Experimental Study on the Hypothermic Preservation of the Rabbit Heart Using Glucose-Insulin-Potassium Solution—Intermittent Perfusion Method Versus Simple Immersion Method”, Japanese Annals of Thoracic Surgery, 7(6):527-532, Dec. 25, 1987 (16 pages)—English Translation.
Kozaki, K. et al., “Usefulness of a Combination of Machine Perfusion and Pentoxifylline for Porcine Liver Transplantation From Non-Heart-Beating Donors With Prolonged Hypotension”, Transplantation Proceedings, 29:3476-3477 (1997), 2 pages.
Kubono, K. et al., “Examination of Plasma and Corpuscle Adenosine Concentration in Normal Subject by Radioimmunoassay, Rinshou Kagaku (Clinical Chemistry”, 20(2):72-77, Jun. 1991 (6 pages)—Japanese Language.
Kuroda, Y. et al., “A New, Simple Method for Cold Storage of the Pancreas Using Perfluorochemical”, Transplantation, 46(3):457-460 (1988), 4 pages.
Lasley et al. Protective Effects of Adenosine in the Reversibly Injured Heart. Ann. Thorac. Surg. 60(3):843-46 (1995).
Lawrence, C., “Machine preserves organs outside body,” Chicago Sun Times (2001), 1 page.
Lefer, A.M. Attenuation of Myocardial Ischemia-Reperfusion Injury With Nitric Oxide Replacement Therapy. Ann. Thorac. Surg. 60(3):847-51 (1995).
Li, G. et al., “Functional Recovery in Rabbit Heart after Preservation with a Blood Cardioplegic Solution and Perfusion,” J Heart Lung Transplant, 12(2)263-270 (1993) (8 pages).
Li, X. et al., “Insulin in University of Wisconsin Solution Exacerbates the Ischemic Injury and Decreases the Graft Survival Rate in Rat Liver Transplantation”, Transplantation, 15:76(1):44-49 (2003), 6 pages.
Liu,J. et al., “Annexin V Assay-proven Anti-apopototic Effect of Ascorbic Acid 2-glucoside after Cold Ischemia/Reperfusion Injury in Rat Liver Transplantation”, Acta Med. Okayama, 57(5):209-216 (2003), 8 pages.
Lobato, E.B. et al., “Treatment with phosphodiesterase inhibitors type III and V: milrinone and sildenafil is an effective combination during thromboxane-induced acute pulmonary hypertension”, British Journal of Anaesthesia, 96(3):317-322, 2006 (6 pages).
Loor, et al., “Prolonged EVLP Using OCS Lung: Cellular and Acellular Perfusates”, Author Manuscript published in final edited form as Transplantation, 101(10):2303-2311, Oct. 2017 (20 pages).
Macchiarini, P. et al. “Ex Vivo Lung Model of Pig-To-Human Hyperacute Xenograft Rejection”, The Journal of Thoracic and Cardiovascular Surgery, 114:3, 315-325 (1997) (11 pages).
Mankad et al. Endothelial dysfunction caused by University of Wisconsin preservation solution in the rat heart. J. Thorac. Cardiovasc. Surg. 104(6):1618-24 (1992).
Matsuno, N. et al., “Effectiveness of Machine Perfusion Preservation as a Viability Determination Method for Kidneys Procured from Non-Heart-Beating Donors,” Transplantation Proceedings, 26(4):2421-2422 (1994) (2 pages).
Matsuno, N. et al., “The Effect of Machine Perfusion Preservation Versus Cold Storage On the Function of Kidneys From Non-Heart-Beating Donors”, Transplantation, 57(2):293-294 (1994) (2 pages).
Menasche et al. Experimental evaluation of Celsior.RTM., a new heart preservation solution. Eur. J. Cardiothor. Surg. 8:207-13 (1994).
Menasche et al. Improved Recovery of Heart Transplants With a Specific Kit of Preservation Solutions. J. Thorac. Cardiovasc. Surg. 105(2):353-63 (1993).
Menasché, P., “The inflammatory response to cardiopulmonary bypass and its impact on postoperative myocardial function”, Current Opinion in Cardiology, 10:597-604 (1995) (8 pages).
Moisiuk, Y. et al., “Histidine-Tryptophan-Ketoglutarate Versus Euro-Collins for Preservation of Kidneys From Non-Heart-Beating Donors”, Transplantation Proceedings, 28(1):202 (1996) (1 page).
Moller-Pedersen et al.; “Evaluation of Potential Organ Culture Media for Eye Banking Using Human Donor Corneas,” Br J Ophthamol. 85(9):1075-79 (2001).
Morimoto et al., A Simple Method for Extended Heart-Lung Preservation by Autoperfusion. Trans Am Soc Artif Intern Organs. 30:320-24 (1984).
Munshi, et al., “Donor management and lung preservation for lung transplantation”, Lancet Respir Med, 1:318-328, published online Feb. 20, 2013 (11 pages).
Nicholson, M.L. et al., “A Comparison of Renal Preservation by Cold Storage and Machine Perfusion Using a Porcine Autotransplant Model”, Transplantation 78(3):333-337 (2004), 5 pages.
No Author Listed, “CUSTODIOL® HTK Solution for Multi-Organ Protection”, Saudi Center for Organ Transplantation, Date Unknown, originally cited to U.S. Patent Office Jun. 30, 2014, in U.S. Appl. No. 12/892,451 (2 pages).
No Author Listed, “SOLTRAN Kidney perfusion fluid”, Baxter, No Month Listed—2001-2004 (1 page).
No Author Listed, “The comprehensive resource for physicians, drug and illness information”, VIASPAN™ DuPont Pharma Cold Storage Solution, Date Unknown (3 pages).
No Author Listed, “UW Solution Composition”, DuPont Pharmaceutical, Date Unknown (1 page).
No Author Listed. “Custodiol HTK” Physicians′ Desk Reference, 57th Edition, Thomson PDR. ISBN:1-56363-445-7. No Month Listed—2003 (3 pages).
Odagiri, S. et al., “Pusatile Assist Device: New Pulsatile Pump Using Pulsatile Assist Device-Hemodynamic Comparison of Pulsatile V-A Bypass (VABP), Pulsatile Left Heart Bypass (LHBP) and Constant Flow Left Heart Bypass (LHB)”, Journal of Japan Surgical Society, 83(6):515-523, Jun. 1982, 12 pages—English Abstract.
Opelz et al., “Comparative Analysis of Kidney Preservation Methods. Collaborative Transplant Study,” Transplant Proc. 28(1):87-90 (1996).
Opelz, G. et al., “Advantage of Cold Storage Over Machine Perfusion for Preservation of Cadaver Kidneys”, Transplantation, 33(1):64-68 (1982), 5 pages.
Open Anesthesia—Milrinone: pharmacology, https://www.openanesthesia.org/milrinone_pharmacology/, accessed 2019 (3 pages).
Ota, K. et al., “Artificial Organ”, Current State and Future of Substitution of Functions, pp. 150-151, 1983 (7 pages)—English Translation.
Pearl, J.M. et al., Loss of endothelium-dependent vasodilatation and nitric oxide release after myocardial protection with University of Wisconsin solution, Cardiovascular Surgery 107(1):257-264 (1994) (8 pages).
Pego-Fernandes, et al., “Ex vivo lung perfusion: initial Brazilian experience”, J. Bras. Pneumol., 35(11):1107-1112, 2009 (6 pages).
Perfadex Guidelines, NHS Cardiothoracic Advisory Group (CTAG), Mar. 2016 (2 pages).
Petrovsky et al., Justification and Application of a New Method for Transorganic Oxygen Preservation of the Kidneys. Vestn Akad Med Nauk SSSR. (2):69-82(1989)—English Abstract—15 pages.
Pinsky, D. et al., “Restoration of the cAMP Second Messenger Pathway Enhances Cardiac Preservation for Transplantation in a Heterotopic Rat Model”, J. Clin. Invest. 92(6):2944-3002 (1993) (9 pages).
Ploeg et al. Successful 72-Hour Cold Storage of Dog Kidneys With UW Solution. Transplantation 46(2):191-96 (1988).
Pokorny, H. et al., “Histidine-tryptophan-ketoglutarate solution for organ preservation in human liver transplantation—a prospective multi-centre observation study”, Transpl Int 17(5):256-260 (2004), (5 pages).
Poston, R.S. et al., “Optimizing Donor Heart Outcome After Prolonged Storage With Endothelial Function Analysis and Continuous Perfusion”, Ann Thorac Surg, 78:1362-1370, 2004 (9 pages).
Potdar et al.; “Initial Experience Using Histidine-Tryptophan-Ketoglutarate Solution in Clinical Pancreas Transplantation,” Clin. Transplant. 18(6):661-65 (2004).
Pozniak, A., “Keeping Hearts Alive Doctors Develop a High-Tech System to Salvage Donated Organs”, ABC News.com, (Dec. 7, 2001) (<http://abcnews.go.com/print?id=117085>), (2 pages).
Probst, R. et al. “Carbohydrate and fatty acid metabolism of cultured adult cardiac myocytes”, Am. J. Physiol. 250 (Heart, Circ. Physiol. 19):H853-H860 (1986) (8 pages).
Pruitt, “Pharmacological Treatment of Respiratory Disorders”, RT Magazine, http://www.rtmagazine.com/2007/05/pharmacological-treatment-of-respiratory-disorders, May 3, 2007, accessed Jan. 1, 2019 (6 pages).
Rao, M.V. et al., “Magnesium Sulfate: Chemical and Technical Assessment”, MgSO4 (CTA), 2007 (5 pages).
Rao, V. et al., “Donor Blood Perfusion Improves Myocardial Recovery After Heart Transplantation”, J. Heart Lung Transplant. 16(6):667-673 (1997) (7 pages).
Reddy, S.P. et al., “Preservation of Porcine Non-Heart-Beating Donor Livers By Sequential Cold Storage and Warm Perfusion”, Transplantation, 77(9):1328-1332 (2004), 5 pages.
Rega, et al., “Long-term Preservation With Interim Evaluation of Lungs From a Non-Heart-Beating Donor After a Warm Ischemic Interval of 90 Minutes”, Annals of Surgery, 238(6):782-793, Dec. 2003 (12 pages).
Richens et al. Clinical Study of Crystalloid Cardioplegia vs Aspartate-Enriched Cardioplegia Plus Warm Reperfusion for Donor Heart Preservation. Transplant. Proc. 24(1):1608-10 (1993).
Rinder, C. et al., “Blockade of C5a and C5b-9 Generation Inhibits Leukocyte and Platelet Activation during Extracorporeal Circulation”, J. Clin. Invest. 96:3(1564-1572) 1995 (9 pages).
Rosenkranz, E.R., “Substrate Enhancement of Cardioplegic Solution: Experimental Studies and Clinical Evaluation”, Ann Thorac Surg 60:797-800 (1995) (4 pages).
Rossi, L. et al., “Innovations—report: New organ preservation solution easier to use”, (<http://www.innovations-report.com/html/reports/medicine_report-18854.html>), Feb. 6, 2003 (2 pages).
Rossi, L., “Portable Organ Preservation System™ Keeps Human Heart Alive Outside Body”, PITT Campaign Chronicle (Oct. 7, 2001), 2 pages.
Russell, H.E., Jr. et al., “An Evaluation of Infusion Therapy (Including Dextran) for Venous Thrombosis”, Circulation, 33:839-846, Jun. 1966 (8 pages).
Sato, H. et al., “Supplemental L-Arginine During Cardioplegic Arrest and Reperfusion Avoids Regional Postischemic Injury”, J Thorac Cardiovasc Surg 110(2):302-314 (1995), 13 pages.
Schmid, T. et al., “The Use of Myocytes as a Model for Developing Successful Heart Preservation Solutions”, Transplantation 52(1):20-26 (Jul. 1991) (7 pages).
Schon et al.; “Liver Transplantation After Organ Preservation with Normothermic Extracorporeal Perfusion,” Ann Surg. 233(1):114-23 (2001).
Schwalb et al. New Solution for Prolonged Myocardial Preservation for Transplantation. J. Heart Lung Transplant. 17(2):222-29 (1998).
Seccombe et al. Coronary Artery Endothelial Function After Myocardial Ischemia and Reperfusion. Ann. Thorac. Surg. 60(3):778-88 (1995).
Segel et al. Posttransplantation Function of Hearts Preserved with Fluorochemical Emulsion. J. Heart Lung Transplant. 13(4):669-80 (1994).
Segel,L.D. et al., “Recovery of Sheep Hearts After Perfusin Preservation or Static Storage with Crystalloid Media”, The Journal of Heart and Lung Transplantation, 17:211-221 (1998) (11 pages).
Sekine, M. et al., “Effect of Obese and Aging on Blood Fatty Acid Consumption in Japanese”, Bulletin of the Graduate School of Human Life Science, Showa Women's University, 4:63-70, 1995 (8 pages)—English Abstract.
Semat, H. and Katz, R., “Physics, Chapter 9: Hydrodynamics (Fluids in Motion)”, Hydrodynamics. University of Nebraska—Lincoln. Pap143. No Month Listed 1958 (18 pages).
Shimokawa, S. et al., “A New Lung Preservation Method of Topical Cooling by Ambient Cold Air: An Experimental Study”, Transplantation Proceedings, 23 (1):653-654 (1991) (2 pages).
Shirakura, R. et al., “Multiorgan Procurement from Non-Heart-Beating Donors by use of Osaka University Cocktail, Osaka Rinse Solution, and the Portable Cardiopulmonary Bypass Machine”, Transplantation Proceedings, 25(6):3093-3094 (1993) (2 pages).
Siobal, M.S. “Pulmonary Vasodilators”, Respir Care, 52(7):885-899, Jul. 2007 (15 pages).
Southard, J., “The Right Solution for Organ Preservation”, Business Briefings: Global Surgery 79-84 (2004) (6 pages).
Steen Solution, Consultation Procedure Public Assessment Report (CPAR), European Medicines Agency, EMEA/CHMP/329441/2005, Aug. 8, 2012 (20 pages).
Steen, S. et al., “Transplantation of lungs from non-heart-beating donors after functional assessment ex vivo”, Ann Thorac Surg, 76:244-252, 2003, 11 pages.
Stubenitsky et al., “Kidney Preservation in the Next Millennium,” Transpl. Int. 12:83-91 (1999).
Sunamori et al. Relative Advantages of Nondepolarizing Solution to Depolarizing University of Wisconsin Solution in Donor Heart Preservation. Transplant. Proc. 25(1):1613-17 (1993).
Synchrony Definition, http://dictionary.reference.com/browse/synchrony, Random House Unabridged Dictionary, 2006 (1 page).
Tang et al., “Warm Ischemia Lung Protection with Pinacidil: an ATP Regulated Potassium Channel Opener,” Ann Thorac Surg. 76:385-9 (2003).
Tesi, R.J. et al., Pulsatile Kidney Perfusion for Preservation and Evaluation: Use of High-Risk Kidney Donors to Expand the Donor Pool, Transplantation Proceedings, 25(6):3099-3100 (1993) (2 pages).
Turpin, B.P. et al., “Perfusion of Isolated Rat Adipose Cells”, The Journal of Clinical Investigation, 60:442-448 (1977), 7 pages.
U.S. Food and Drug Administration, Center for Drug Evaluation and Research, “Drugs@FDA—Solu-Medrol: Label and Approval History”, (Available online at http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.Label_ApprovalHistory#apphist . . . ), accessed Feb. 9, 2010 (3 pages).
U.S. Food and Drug Administration, Center for Drug Evaluation and Research, “Drugs@FDA—Solu-Medrol: Drug Details”, (Accessible online at http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.DrugDetails . . . ), accessed Feb. 9, 2010 (1 page).
Venuta, F. et al., “History of lung transplantation”, Journal of Thoracic Disease, 9(12):5458-5471, Dec. 2017 (14 pages).
Vinten-Johansen et al. Reduction in Surgical Ischemic-Reperfusion Injury With Adenosine and Nitric Oxide Therapy. Ann. Thorac. Surg. 60(3):852-57 (1995).
Voiglio, E. et al. “Rat Multiple Organ Blocks: Microsurgical Technique of Removal for Ex Vivo Aerobic Organ Preservation Using a Fluorocarbon Emulsion”, Microsurgery 20:3, 109-115 (2000) (7 pages).
Wallinder, et al., “Transplantation of initially rejected donor lungs after ex vivo lung perfusion”, Cardiothoracic Transplantation, 144(5):1222-1228, Nov. 2012 (7 pages).
Watanabe, S. et al., “Effects of free fatty acids on the binding of bovine and human serum albumin with steroid hormones”, Biochimica et Biophysica Acta (BBA), 1289:385-396 (1996), 12 pages.
Wei, Y. et al., “Protective Effect of Specific Phosphodiesterase Inhibitor Milrinone for Donor Lungs”, Chinese Journal of New Drugs, 16(21):1762-1765, 2007—English Translation issued by U.S. Patent and Trademark Office, Aug. 2020 (17 pages).
Wei, Z., et al., “A Study on the Preservation of Rat Kidney with HX-III Solution”, J WCUMS, 31(3):347-349 (2000)—English Abstract, 4 pages.
Wicomb, W. et al., “Orthotopic transplantation of the baboon heart after 20 to 24 hours′ preservation by continuous hypothermic perfusion with an oxygenated hyperosmolar solution”, J. Thorac Cardiovasc Surg, 83(1):133-140 (1982), 8 pages.
Wicomb, W.N. et al., “24-Hour Rabbit Heart Storage With UW Solution”, Transplantation, 48(1):6-9 (1989), 4 pages.
Wicomb, W.N. et al., “Cardiac Transplantation Following Storage of the Donor Heart by a Portable Hypothermic Perfusion System”, The Annals of Thoracic Surgery, 37(3):243-248 (1984), 6 pages.
Wright, N. et al. “A porcine ex vivo paracorporeal model of lung transplantation”, Laboratory Animals Ltd. Laboratory Animals, 34:1, 56-62 (2000) (7 pages).
XVIVO Perfusion, RedEye Equity Research, May 29, 2020 (3 pages).
Yang, W. et al., “Effect of Hypoxia and Reoxygenation on the Formation and Release of Reactive Oxygen Species by Porcine Pulmonary Artery Endothelial Cells”, Journal of Cellular Physiology, 164:414-423 (1995) (10 pages).
Yeung, J., et al., “Physiologic assessment of the ex vivo donor lung for transplantation”, Journal of Heart and Lung Transplantation, 31(10):1120-1126, Oct. 2012 (7 pages).
Yland, M.J. et al., “New Pulsatile Perfusion Method for Non-Heart-Beating Cadaveric Donor Organs: A Preliminary Report”, Transplantation Proceedings, 25(6):3087-3090 (1993), 4 pages.
Yokoyama, H. et al., “Isolated Dog Hearts Prepared in Cold Tyrode Solution and Reperfused with Arterial Blood Are Functionally and Ultrastructurally Normal”, The Tohoku Journal of Experimental Medicine, 156:121-134, 1988 (14 pages).
Zalewska, et al., National Standards for Organ Retrieval from Deceased Donors (extracts), NHS Blood and Transplant, UK National Health Service, MPD1043/8, effective date Oct. 15, 2018 (50 pages).
Zhang et al., “Research Progress on Preservation of Severed Limbs,” Chinese Journal of Reparative and Reconstructive Surgery 14(3):189-192 (2000).
Keshavjee, S.H. et al., “The role of dextran 40 and potassium in extended hypothermic lung preservation for transplantation”, Journal of Thoracic and Cardiovascular Surgery, 103(2):314-325, Feb. 1992 (12 pages).
Saez, D.G. et al., “Evaluation of the Organ Care System in Heart Transplantation With an Adverse Donor/Recipient Profile”, Ann. Thorac. Surg., 98:2099-2106, 2014 (8 pages).
Shimokawa, S. et al., “A New Lung Preservation Method of Topical Cooling by Ambient Cold Air Combined with High-Frequency Oscillation: An Experimental Study”, Transplantation Proceedings, 26(4):2364-2366, Aug. 1994 (3 pages).
Wittwer, et al., “Experimental Lung Transplantation: Impact of Preservation Solution and Route of Delivery”, The Journal of Heart and Lung Transplantation, 24(8):1081-1090, Aug. 2005 (10 pages).
Albes, et al., “Influence of the Perfusate Temperature on Lung Preservation: Is There an Optimum?”, European Surgical Research, 29:5-11, 1997 (7 pages).
Besterman, et al., “Regulation of protein synthesis in lung by amino acids and insulin”, American Journal of Physiology: Endocrinology and Metabolism, 245(5):E508-E514, Nov. 1, 1983 (7 pages).
Erasmus, et al., “Normothermic ex vivo lung perfusion of non-heart-beating donor lungs in pigs: from pretransplant function analysis towards a 6-h machine preservation”, Transplant International, 19(7):589-593, Jul. 1, 2006 (5 pages).
“The secret of the turtle”, <https://mag.ebmpapst.com/en/industries/medical/the-secret—of-the-turtle_2433/>, mag: The Magazine of ebm-papst, Sep. 2009 (5 pages).
Baker, et al., “Calcium Content of St. Thomas' II Cardioplegic Solution Damages Ischemic Immature Myocardium”, Annals of Thoracic Surgery, 52(4):993-999, Oct. 1991 (7 pages).
Charest, et al., “Design and validation of a clinical-scale bioreactor for long-term isolated lung culture”, Author Manuscript published in Final Edited form as Biomaterials, 52:79-87, Jun. 2015 (22 pages).
De Hart, et al., “An ex vivo platform to simulate cardiac physiology: a new dimension for therapy development and assessment”, The International Journal of Artificial Organs, 34(6):495-505, Jun. 2011 (11 pages).
Definition of Aqueous from Cambridge Dictionary, https://dictionary.cambridge.org/us/dictionary/english/aqueous, accessed Sep. 14, 2023 (2 pages).
Definition of Medium, Collins English Dictionary, https://www.collinsdictionary.com/us/dictionary/english/medium#:˜:text=You%20use%20medium%20to%20describe,middling%20More%20Synonyms%20of%20medium, accessed Sep. 14, 2023 (2 pages).
Dobson, et al., “Adenosine and lidocaine: A new concept in nondepolarizing surgical myocardial arrest, protection, and preservation”, Journal of Thoracic and Cardiovascular Surgery, 127(3):794-805, Mar. 2004 (12 pages).
Ebel, et al., “Lidocaine reduces ischaemic but not reperfusion injury in isolated rat heart”, British Journal Anaesthesia, 86(6):846-852, 2001 (7 pages).
Ely, et al., “Protective Effects of Adenosine in Myocardial Ischemia”, Circulation, 85(3):893-904, Mar. 1992 (12 pages).
European Extended Search Report issued in European Patent Application No. 17805438.3, dated Jan. 28, 2020 (14 pages).
European Extended Search Report issued in European Patent Application No. 15853016.2, dated Mar. 9, 2018 (12 pages).
European Extended Search Report issued in European Patent Application No. 13738530.8, dated Jan. 25, 2016 (9 pages).
European Extended Search Report issued in European Patent Application No. 15767752.7, dated Nov. 30, 2017 (7 pages).
European Extended Search Report issued in European Patent Application No. 15775970.5, dated Oct. 24, 2017 (10 pages).
European Extended Search Report issued in European Patent Application No. 15867786.4, dated Feb. 8, 2019 (14 pages).
European Extended Search Report issued in European Patent Application No. 18879106.5, dated Dec. 17, 2020 (8 pages).
European Search Report issued in European Patent Application No. 15867786.4, dated Sep. 3, 2018 (11 pages).
Gao, et al., “Role of Troponin I Proteolysis in the Pathogenesis of Stunned Myocardium”, Circulation Research, 80(3):393-399, Mar. 1, 1997 (17 pages).
Hearse, et al., “Protection of the myocardium during ischemic arrest”, Journal of Thoracic Cardiovascular Surgery, 81(6):873-879, Jun. 1981 (7 pages).
International Preliminary Report on Patentability issued in International Application No. PCT/CA15/50297, dated Oct. 12, 2016 (6 pages).
International Preliminary Report on Patentability, issued in International Application No. PCT/CA15/51084, dated Feb. 15, 2017 (3 pages).
International Preliminary Report on Patentability, issued in International Application No. PCT/CA2015/051316 dated Apr. 10, 2017 (5 pages).
International Preliminary Report on Patentability, issued in International Application No. PCT/CA2013/000031 dated Apr. 23, 2014 (8 pages).
International Preliminary Report on Patentability, issued in International Application No. PCT/CA2015/050201 dated Sep. 27, 2016 (5 pages).
International Search Report and Written Opinion issued by the Canadian Patent Office as International Searching Authority in International Application No. PCT/CA2015/051316, dated Mar. 16, 2016 (8 pages).
International Search Report and Written Opinion issued by Canadian Patent Office as International Searching Authority in International Application No. PCT/CA13/00031, dated Apr. 15, 2013 (9 pages).
International Search Report and Written Opinion issued by Canadian Patent Office as International Searching Authority in International Application No. PCT/CA18/51474, dated Mar. 4, 2019 (6 pages).
International Search Report and Written Opinion issued by the Canadian Patent Office as International Searching Authority in International Application No. PCT/CA15/50201, dated Jun. 10, 2015 (8 pages).
International Search Report and Written Opinion issued by the Canadian Patent Office as International Searching Authority in International Application No. PCT/CA15/50297, dated Jul. 13, 2015 (8 pages).
International Search Report and Written Opinion issued by the Canadian Patent Office as International Searching Authority in International Application No. PCT/CA15/51084, dated Feb. 5, 2016 (8 pages).
Jakobsen, et al., “Adenosine instead of supranormal potassium in cardioplegia: It is safe, efficient, and reduces the incidence of postoperative atrial fibrillation. A randomized clinical trial”, Journal of Thoracic and Cardiovascular Surgery, 145(3):812-818, Mar. 2013 (7 pages).
Lim, et al., “Computational analysis of the effect of the type of LVAD flow on coronary perfusion and ventricular afterload”, J. Physiol Sci., 59:307-316, 2009 (10 pages).
Mehaffey, et al., “Airway pressure release ventilation during ex vivo lung perfusion attenuates injury”, Journal Thoracic Cardiovascular Surgery, 153(1):197-204, Jan. 2017 (8 pages).
Muhlbacher, et al., “Preservation Solutions for Transplantation”, Transplantation Proceedings, 31(5):2069-2070, Aug. 1999 (2 pages).
Nelson, et al., “Abstract 736: Determination of Optimum Ventilation Strategy for Ex-Vivo Lung Perfusion: Comparing Negative and Positive Pressure Ventilation”, Journal of Heart and Lung Transplantation, 34(4 Supplement):S270, Apr. 2015 (1 page).
O'Blenes, et al., “Protecting the aged heart during cardiac surgery: The potential benefits of del Nido cardioplegia”, Journal Thoracic and Cardiovascular Surgery, 141(3):762-770, Mar. 2011 (9 pages).
Popov, et al., “Ex Vivo Lung Perfusion—State of the Art in Lung Donor Pool Expansion”, Medical Science Monitor Basic Research, 21:9-14, Feb. 3, 2015 (6 pages).
Raymondos, et al., “Combined Negative- and Positive-Pressure Ventilation for the Treatment of ARDS”, Case Reports in Critical Care, Article ID714902, 2015 (5 pages).
Robinson, et al., “Lowering the calcium concentration in St. Thomas' Hospital cardioplegic solution improves protection during hypothermic ischemia”, Journal Thoracic and Cardiovascular Surgery, 101(2):314-325, Feb. 1991 (12 pages).
Rudd, et al., “Eight hours of cold static storage with adenosine and lidocaine (Adenocaine) heart preservation solutions: Toward therapeutic suspended animation”, Journal of Thoracic Cardiovascular Surgery, 142(6):1552-1561, Dec. 2011 (10 pages).
Rudd, et al., “Toward a new cold and warm nondepolarizing, normokalemic arrest paradigm for orthotopic heart transplantation”, The Journal of Thoracic and Cardiovascular Surgery, 137(1):198-207, Jan. 2009 (10 pages).
Sutherland, et al., “The Isolated Blood And Perfusion Fluid Perfused Heart”, https://www.southalabama.edu/ishr/help/hearse/, Cardiovascular Research—The Centre for Cardiovascular Biology and Medicine, The Rayne Institute, King's College, St Thomas' Hospital, London, UK; originally retrieved on Oct. 16, 2017, accessed Aug. 28, 2023 (12 pages).
Takemoto, et al., “The reciprocal protective effects of magnesium and calcium in hyperkalemic cardioplegic solutions on ischemic myocardium”, Basic Research in Cardiology, 87(6):559-569, 1992 (11 pages).
Tane, et al., “Ex Vivo Lung Perfusion: A Key Tool for Translational Science in the Lungs”, Chest, 151(6):1220-1228, Jun. 2017 (9 pages).
Taylor, et al., “Registry of the International Society for Heart and Lung Transplantation: Twenty-sixth Official Adult Heart Transplant Report—2009”, Journal of Heart and Lung Transplantation, 28(10):1007-1022, Oct. 2009 (16 pages).
Tipton, et al., “The use of Langendorff preparation to study the bradycardia of training”, Medicine and Science in Sports, 9(4):220-230, 1977 (11 pages).
unitslab.com, Online Converter, Lidocaine, https://unitslab.com/node/178, retrieved Aug. 29, 2023 (3 pages).
White et al., “Abstract 735: Impact of Initial Acidic Reperfusion on the Functional Recovery of DCD Hearts During Ex Vivo Heart Perfusion”, Journal of Heart and Lung Transplantation, 34(4S):S269-S270, Apr. 2015 (2 pages).
White, et al., “Abstract 385: Impact of Initial Acidic Reperfusion on the Functional Recovery of DCD Hearts During Ex Vivo Heart Perfusion”, Canadian Journal Cardiology, 30:S251-252, 2014 (2 pages).
White, et al., “Impact of Reperfusion Calcium and pH on the Resuscitation of Hearts Donated After Circulatory Death”, Annals of Thoracic Surgery, 103:122-130, Jan. 2017 (9 pages).
Wild et al., “PEEP and CPAP”, British Journal of Anaesthesia, 1(3):89-92, 2001 (4 pages).
Zhong, et al., “The management experience of long duration roller pump ventricular assist device,” Chinese Journal of ECC, 9(3):134-137, Sep. 15, 2011 (4 pages)—English Abstract Only.
Related Publications (1)
Number Date Country
20220361482 A1 Nov 2022 US
Provisional Applications (1)
Number Date Country
61475524 Apr 2011 US
Divisions (1)
Number Date Country
Parent 13446706 Apr 2012 US
Child 17869701 US