throbber
JOURNAL OF HEMATOTHERAPY 1:233-250 (1992)
`Mary Ann Liebert, Inc., Publishers
`
`Hematopoietic Stem Cell Cryopreservation: A Review of
`Current Techniques
`
`SCOTT D. ROWLEY
`
`ABSTRACT
`Hematopoietic stem cells (HSC) can be stored for prolonged periods at cryogenic tempera-
`tures. The techniques currently used were derived from the initial report
`in 1949 of
`cryopreservation of bovine sperm in glycerol. The addition of this penetrating cryoprotectant
`protected the cells from the injury associated with ice formation. Current cryopreservation
`techniques (with minor variations) suspend cells in an aqueous solution of salts, protein, and
`one or more cryoprotectants. Cells are frozen at slow rates and stored generally below
`120°C
`in mechanical freezers or nitrogen refrigerators. That these techniques are successful
`in
`maintaining HSC viability is evident from the engraftment of these cells in patients treated
`with marrow-lethal conditioning regimens. However, issues such as the composition of the
`cryoprotectant solution, cell concentration during freezing, cryoprotectant toxicity, and
`storage temperatures have not been adequately studied, primarily because of a lack of
`appropriate assays for HSC cryosurvival. HSC cryobiology will become an increasingly
`important subject as new HSC collection and processing techniques are developed. Improved
`cryosurvival of HSC using modified cryoprotectant solutions may improve engraftment
`kinetics and decrease the cost and morbidity of autologous transplantation.
`
`—
`
`INTRODUCTION
`
`Cryopreservation of bone marrow or peripheral blood-derived hematopoietic stem cells (HSC) permits
`
`the administration of intensive chemo-radiotherapy to patients with dose-responsive malignancies.
`Although cryopreservation is not a requirement for autologous transplantation, and patients have recovered
`marrow function after reinfusion of marrow cells stored for several days at 4°C, a progressive loss of
`hematopoietic stem cells occurs during nonfrozen storage. Definition of ideal conditions (e.g., temperature,
`initial processing of the cells, additives) for nonfrozen storage of cells may slow this loss. However, merely
`maintaining survival, much less growing additional HSC, is beyond the current capabilities of in vitro cell
`culture techniques. Therefore, most transplant centers cryopreserve HSC intended for autologous transplan-
`tation. Cryopreservation allows administration of multiple-day transplant conditioning regimens as well as
`prophylactic storage for patients to be transplanted months to years later. Although some unavoidable loss of
`
`Fred Hutchinson Cancer Research Center, Seattle, WA 98104.
`
`233
`
`Downloaded by University of Michigan from www.liebertpub.com at 05/27/20. For personal use only.
`
`Novo Nordisk Ex. 2040, P. 1
`Mylan Institutional v. Novo Nordisk
`IPR2020-00324
`
`

`

`ROWLEY
`HSC occurs with marrow or peripheral blood stem cell (PBSC) processing and cryopreservation, progressive
`loss over months of proper storage is not obvious and may not occur.
`That HSC can be successfully cryopreserved is evident from the success ofautologous HSC transplantation
`in regenerating marrow function after marrow-lethal conditioning regimens. Engraftment failure is generally
`not attributed to HSC cryopreservation, although some investigators correlated poor HSC cryopreservation
`with delayed engraftment after transplantation (Gorin, 1986; Rowley et al, 1989). Most cryopreservation
`laboratories use a variation of the technique outlined in Table 1. Cryopreservation research, however, has not
`been a focus of the transplantation teams, and HSC cryopreservation is one of the least understood aspects of
`HSC processing. This lack of understanding of cryobiology hinders the ability to adapt techniques for
`handling unusually small or large collections of cells or other accidental or intentional deviations from
`laboratory protocol. Although easily performed by a number oftransplant centers, HSC cryopreservation and
`reinfusion are not without risk of toxicity to both the stem cell inoculum and the recipient.
`It is the purpose of this commentary to review the development of our current cryobiology techniques,
`examining the parameters depicted in Table 1. For this review, HSC is intended to refer to both primitive
`hematopoietic stem cells responsible for durable engraftment as well as the more mature progenitor cells that
`can be cultured in vitro, regardless of source (e.g., bone marrow, peripheral blood).
`CRYOPRESERVATION THEORY
`Preservation of viability after cryopreservation and thawing became possible after the discovery that
`glycerol could be used for the freezing of bovine sperm (Polge et al, 1949). Subsequent experiments
`demonstrated that bone marrow frozen with glycerol could be used to reconstitute the bone marrow of
`irradiated mice (Barnes & Loutit, 1955). Considerable exploration of the cryobiology of various mammalian
`and nonmammalian cells ensued. Those studies defined the mechanism of cell damage incurred during
`cryopreservation, and techniques to moderate that damage. The current understanding of cryobiology
`contends that ice crystal formation during cooling is the primary cause of cell damage (Karow & Webb,
`1965a). Intracellular ice crystals may form at rapid rates of cooling, resulting in mechanical rupture of cell
`structures. At slower rates of cooling, ice crystal formation occurs in the extracellular space, resulting in
`increasing osmolality as free water is incorporated into the ice crystals. This loss of free water results in the
`concentration of extracellular solutes, such as sodium ions, that do not freely penetrate the cell membrane (or
`are actively excluded), extreme hyperosmolality, and dehydration injury. For example, the molality of Na+
`10CC is about 2.8 molal, and at -20°C about 5.6 molal (Mazur, 1970).
`in a saline solution at
`
`-
`
`Cell processing:
`Cell concentration:
`Storage container:
`Sample volume:
`Cryoprotectant:
`Protein:
`Electrolytes:
`Rate of cooling:
`Storage temperature:
`Rate of thawing:
`Post-thaw manipulation:
`
`Table 1. Seattle HSC Cryopreservation Technique
`Buffy-coat or light-density cells
`>2 x 107 cells/ml final concentration
`Blood storage bag tolerant of cryogenic temperatures
`Dependent upon bag size, generally 40-60 ml per bag
`10% (vol/vol) dimethylsulfoxide final concentration
`20% autologous plasma
`Tissue culture medium
`l°C/minute to —40°C with compensation for heat of fusion, then
`10°C/minute to
`-80°C
`135°C in nitrogen vapor-phase refrigerators or mechanical freezers
`Below
`Rapidly (>100°C/minute) in a 37°C water bath
`Addition of ACD (20% vol/vol of product volume), then infusion immediately after
`thawing without further processing; filtration through blood administration set
`(170 u.) if clumped after thawing
`
`—
`
`—
`
`234
`
`Downloaded by University of Michigan from www.liebertpub.com at 05/27/20. For personal use only.
`
`Novo Nordisk Ex. 2040, P. 2
`Mylan Institutional v. Novo Nordisk
`IPR2020-00324
`
`

`

`—
`
`CRYOPRESERVATION OF HEMATOPOIETIC CELLS
`Intracellular ice formation may be limited by cooling cells slowly. At slow rates, it is probable that ice
`nucleation will first occur in the larger volume outside the cell. Progressive dehydration of the cell will result
`if the rate ofcooling is slow enough to allow water to shift to the extracellular space and be incorporated into
`the growing ice crystals. The permeability ofthe cell to water appears to define the optimal cooling rate for any
`particular cell (Mazur, 1966). Dehydration with concentration of intracellular solutes protects the cells from
`intracellular ice formation and growth.
`Glycerol and dimethylsulfoxide (DMSO) are colligative cryoprotectants that prevent dehydration injury by
`moderating the increased concentration ofnonpenetrating extracellular solutes such as sodium ions during ice
`formation, and by decreasing the amount of water absorbed by (in equilibrium with) the ice crystals at a
`defined temperature. Colligative refers to properties dependent upon the number ofparticles (solute), not the
`composition of the particles. Freezing is the crystallization of water, and the freezing point is the temperature
`at which ice crystals can be sustained in equilibrium with water. The freezing point of water is depressed by
`the addition of solute. For any particular mixture of solute(s) and water, there will be a defined temperature at
`which ice crystals can initially form. Unlike pure water, ice crystal formation and growth in aqueous solutions
`occurs over a temperature range. Growing ice crystals absorb free water and exclude solute particles. The
`incorporation of water into ice results in concentration of the solute and further depression of the freezing
`temperature oftheremaining water, thereby preventing additional ice formation unless further cooling occurs.
`Thus, temperature (and pressure) defines the equilibrium between ice and the nonfrozen solution. With further
`cooling, a temperature is eventually reached at which the solute itself crystallizes (eutectic point). The
`molality of a solution in equilibrium with ice is determined by the temperature of the solution (and not the
`20°C has a Na+ molality of
`initial concentration of the solute). In our example above, a saline solution at
`about 5.6 molal. For a two-component system such as DMSO and saline, both solutes will contribute to the
`molality of the unfrozen solution. The molality (before freezing) of 10% DMSO (vol/vol) in solution is about
`1.6 molal, about 10 times the molality ofNa+ in the medium. At -20°C, this molal ratio between DMSO and
`Na+ will be maintained. Therefore, the molality of the Na+ will be about 0.56 molal, or a little less than four
`times the molality ofNa+inasolution without ice. The addition of a penetrating cryoprotectant to an aqueous
`saline solution, therefore, reduces the osmotic stress across the cell membrane at
`20°C from about 36 times
`to less than four times that of an ice-free solution. According to this theory, colligative cryoprotectants must
`be capable ofpenetrating the cell to avoid merely contributing to the molality of the extracellular medium (and
`must be nontoxic to the cells at the concentration required for cryopreservation).
`The tolerance of cells to freezing, therefore, depends on the ability of the cells to withstand osmotic stress.
`The osmotic tolerance of granulocytes is much less than that for lymphocytes or mature hematopoietic
`progenitors, probably accounting at least in part for the difference in survival after cryopreservation (Dooley
`& Takahashi, 1981 ; Law et al, 1983). With DMSO concentrations less than 10%, the degree of dehydration
`caused by concentration of the nonpenetrating solutes during freezing will be greater because nonpenetrating
`solutes will contribute proportionately more to the molality of the nonfrozen solution in the extracellular
`medium. With greater concentrations of DMSO,the osmotic stress will be less. The optimal concentration for
`acolligative cryoprotectant, therefore, depends upon the osmotic tolerance of the cell to be frozen, the toxicity
`to the cell of high concentrations of the cryoprotectant itself (Karow & Webb, 1965b; Fahy, 1986; Arakawa
`et al, 1990), and the presence of other cryoprotectants.
`Colligative properties do not explain the cryopreservation achieved by freezing cells in solutions of
`macromolecules such as hydroxyethyl starch (HES). Solutions of high-molecular-weight, polymeric
`cryoprotectants contain relatively few particles and, moreover, do not freely penetrate the cell. These
`cryoprotectants may protect the cell by forming a viscous, glassy shell that retards the movement of water,
`thereby preventing progressive dehydration as water is incorporated into the extracellular ice crystals
`(Takahashi etal, 1988). Solutions of some compounds, when present in sufficiently high concentrations, will
`solidify to an amorphous glass without first forming ice crystals, a process termed "vitrification" (Fahy et al,
`1984). The "glass-transition temperature" (Tg) depends upon both the structure and concentration of the
`solute. Pure water forms a glass at about
`139°C (Grout, 1991). The addition of cryoprotectants such as
`DMSO, glycerol, or HES raises the Tg (Luyet, 1960). At very high concentrations ofcryoprotectants, (6.3 M
`for DMSO [Fahy et al, 1984]), the T is higher (warmer) than the temperature at which ice crystals can form,
`thereby preventing crystallization during cooling and its resulting mechanical and osmotic stresses. The
`235
`
`—
`
`—
`
`Downloaded by University of Michigan from www.liebertpub.com at 05/27/20. For personal use only.
`
`Novo Nordisk Ex. 2040, P. 3
`Mylan Institutional v. Novo Nordisk
`IPR2020-00324
`
`

`

`ROWLEY
`practical difficulty with achieving vitrification arises from the necessity to use very high concentrations of
`cryoprotectants. Vitrification has been used for the cryopreservation of murine embryos, for example, using
`a solution of 20% DMSO, 15.5% acetamide, 10% propylene glycol, and 6% polyethylene glycol (Rail &
`Fahy, 1985). Prolonged exposure to this solution was toxic, however, with complete loss ofviability after 30
`min at 4°C. With lower concentrations of cryoprotectants, the crystallization temperature is higher than the
`vitrification temperature and ice crystals will form during cooling. Yet, glass transition may occur even in the
`presence of ice crystallization. With the formation of ice crystals and resulting loss' in free water,
`the
`concentration of the cryoprotectant in the nonfrozen water will increase. With decreasing temperature causing
`increasing cryoprotectant concentration, a point will be reached at which the cryoprotectant forms a glass. At
`this "limiting glass transition temperature" (T'g), the solution suddenly becomes viscous, retarding, if not
`stopping, the flow of water through the extracellular matrix. This has been proposed as the mechanism for
`In one study of
`cryoprotection afforded by the extracellular cryoprotectants (Takahashi et al, 1988).
`peripheral blood monocyte cryopreservation using extracellular cryoprotectants, a limiting glass-transition
`20°C was optimal, and this was achieved using a 20% solution of HES (Takahashi et al,
`temperature of
`1988). Substances forming glasses at higher or lower temperatures were less effective. This model of
`cryopreservation requires adequate dehydration to occur to concentrate intracellular solutes and decrease the
`probability of intracellular ice formation, but glass formation at an appropriate temperature (please recall that
`temperature defines the osmolality of the unfrozen aqueous solution) to prevent excessive dehydration of the
`cell.
`Cells are not frozen in simple, two-component solutions, but rather in complex solutions containing salts,
`sugars, penetrating cryoprotectants with or without extracellular cryoprotectants, and plasma proteins (Table
`2). Phase transition temperatures (such as T'g) for these solutions have not been determined. The improved
`cryosurvival observed with increasing protein concentration (Ragab et al, 1977) and combined penetrating
`and extracellular cryoprotectants (Stiff et al, 1983; Conscience & Fischer, 1985) may be explained at least in
`part by these theories on ice and glass formation and their effects on the cell. Moreover, the existence ofglass
`formation may explain the relationship between storage temperature and survival of HSC over time. Below
`the T'g temperature, ice crystal growth cannot occur and the cells are protected from mechanical disruption by
`growing ice crystals. The optimal storage temperature is below the T'g for the cryoprotectant solution used.
`
`—
`
`Other
`Heparin
`
`Table 2. Published Cryopreservation Techniques
`Final concentration
`End-freeze
`Cell
`Pre-chill
`DMSO
`Protein
`(per ml)
`(%)
`(%)
`Author
`cells
`Medium
`temperature
`0.2-1 x 108
`-120°C
`Weiner & Gross
`25, FFP
`HBSS
`Yes
`10
`4 x 107
`10, AP
`-90°C
`Soken
`Yes
`TCI 99
`10
`-90°C
`5, PPF
`TCI 99
`Janssen & Lee
`Yes
`NS
`10
`<108
`Meagher, Herzig & Herzig
`-90°C
`5, AP
`RPMI-1640
`Yes
`10
`<108
`Hill, Robertson & Dickson
`-100°C
`0.5, HSA
`MEM
`Yes
`10
`-90°C
`NS, AP
`TCI 99
`Hollinsworth
`Yes
`10
`NS
`2-4 x 107
`Dyson, Haylock & To
`-80°C
`20, AP
`HBSS
`Yes
`10
`NS, AP
`Heparin
`Yao
`TCI 99
`NS
`NS
`Yes
`10
`4 x 107
`Bouzgarou et al.
`140°C
`NS, AP
`HBSS
`Yes
`10
`Normosol-R*
`Oldenburg & Stiff
`3.6, HSA
`NA
`6% HES
`NS
`Yes
`Plasma-Lyte"
`<108
`Rosina & Jiang
`5, HSA
`NA
`6% HES
`Yes
`All techniques were recently published in a manual of marrow processing techniques (Areman, Deeg & Sacher, 1992).
`Only technical details without supporting clinical data documenting cryopreservation efficacy were provided. Abbrevi-
`ations used are: FFP = fresh frozen plasma, AP = autologous plasma, PPF = plasma protein fraction, HSA = human
`serum albumin, NS = not specified, NA = not applicable.
`
`-
`
`236
`
`55
`
`Downloaded by University of Michigan from www.liebertpub.com at 05/27/20. For personal use only.
`
`Novo Nordisk Ex. 2040, P. 4
`Mylan Institutional v. Novo Nordisk
`IPR2020-00324
`
`

`

`CRYOPRESERVATION OF HEMATOPOIETIC CELLS
`Warmer temperatures may be used, but at risk of cell damage, a risk that is dependent upon the temperature
`of storage and the stability (viscosity) of the solution at that temperature.
`This simplified review of the effects of freezing and the cryoprotectant properties of penetrating and
`polymeric cryoprotectants does not completely explain the processes involved during freezing, and more
`detailed reviews of the freezing of aqueous solutions and mammalian cells have been published (Karow &
`Pegg, 1981; Fuller& Grout, 1991). In addition to mechanical and dehydration injuries, cooling itself may be
`to explain the
`damaging to the cell (Fuller, 1991). Also, colligative effects alone are not sufficient
`cryoprotectant properties of DMSO or glycerol. Other freely penetrating chemicals such as urea and DMS02
`do not function as cryoprotectants for mammalian cells (Vos & Kaalen, 1965; McGann & Walterson, 1987),
`and some chemicals such as ethanol and guanidine may actually function as cryosensitizers (Kruuv et al,
`1990). Obviously, the chemical structure of the cryoprotectant is important in the cryosurvival of mammalian
`cells, and a molecular interaction between the cryoprotectant and protein or lipid molecules appears necessary
`foroptimal cryopreservation (Carpenter & Crowe, 1988; Crowe et al, 1987, 1990; Anchordoguy et al, 1991).
`The damaging effect of freezing on membrane proteins or structure compared to the survival of the cell is
`illustrated by the ability of HSC to proliferate in vitro when stimulated by peripheral blood feeder layers but
`lack of response to GCT-conditioned medium (Gilmore, 1983). Similar "resistance" to recombinant growth
`factors by thawed bone marrow progenitors, suggests a loss of cytokine receptors despite the survival of the
`cell (Rowley & Hattenburg, 1990). The ideal cryoprotectant solution would also protect the various cell
`organdíes and molecular species from sublethal damage.
`
`PROCESSING OF BONE MARROW FOR CRYOPRESERVATION
`important concepts of clinical HSC cryopreservation that must be realized is the
`One of the most
`heterogeneity of marrow and PBSC populations. HSC cryopreservation involves not only the cryobiology of
`the HSC, but also of the mature blood cells contained in the harvested product. The minimum number of cells
`required for marrow reconstitution after autologous transplantation is not known. Generally, 1-3 x 108
`nucleated marrow cells per kilogram of patient weight are harvested. Less certain is the minimum amount of
`cells required for PBSC transplantation; under certain conditions, the quantity collected may far exceed the
`number harvested for marrow transplantation. Regardless, hematopoietic stem cells comprise a very small
`portion (< 1 %) ofeither the marrow orperipheral blood product. Bone marrow consists ofHSC, mature blood
`cells, and noncellular material such as fat. Cryopreservation techniques that are optimal for HSC may destroy
`mature blood cells. The standard technique for red cell cryopreservation uses glycerol (Sputteck & Korber,
`1991); granulocytes cannot be cryopreserved successfully. The presence of mature blood cells affects HSC
`cryopreservation in at least three ways. First, the large proportion of mature blood cells collected may hinder
`the laboratory processing ifclumping before or after thawing is induced by damaged granulocytes or platelets.
`We observed the loss of about 50% of cells when clumping occurred, and infusion through a standard blood
`administration set may be required to prevent pulmonary embolism. Second, damaged cells may also
`predispose toward infusion-related toxicity. The infusion of marrow that was frozen without depletion of
`mature blood cells was associated with acute renal failure in 3 of 33 patients in one study (Smith et al, 1987);
`this presumably resulted from massive hemolysis of red cells. Other investigators correlated the quantity of
`cells cryopreserved and infused to the post-infusion toxicity observed. In one study, infusion ofcryopreserved
`buffy-coat cells incurred greater toxicity than infusion of light-density cells, which were essentially depleted
`of mature blood cells (Davis et al, 1990). Kessinger et al reported a high incidence of infusion-related toxicity
`for recipients of large quantities of peripheral blood stem cell products (Kessinger et al, 1990). Third, if the
`cell concentration affects HSC cryosurvival, then the presence oflarge numbers of mature blood cells requires
`that the cells be frozen in large volumes. It is possible that the toxicity reported by Kessinger et al and Davis
`et al may relate to the quantity of DMSO infused, and not specifically from lysis of mature blood cells.
`Therefore, cryopreservation of HSC can be facilitated by the prefreeze depletion of mature blood elements.
`Collection of buffy-coat cells is the minimum processing required for cryopreservation of bone marrow. A
`number of apheresis or cell-washing devices are capable of processing the large quantities of cells harvested.
`237
`
`Downloaded by University of Michigan from www.liebertpub.com at 05/27/20. For personal use only.
`
`Novo Nordisk Ex. 2040, P. 5
`Mylan Institutional v. Novo Nordisk
`IPR2020-00324
`
`

`

`ROWLEY
`Isolation of buffy-coat cells achieves volume reduction and depletion of red cells. Plasma collected during
`fractionation may be used for subsequent processing or cryopreservation. Some apheresis devices may
`provide enrichment of mononuclear cells relative to granulocytes, providing a "cleaner" preparation for
`processing and cryopreservation. Density-gradient separation of light-density cells further enriches for HSC,
`although at the expense of additional cell losses and cost ofprocessing. At the extreme ofprefreeze processing
`is the extensive purification of HSC by isolation of CD34+ cells (Berenson et al, 1991). This technology may
`be especially advantageous in the cryopreservation of peripheral blood-derived HSC collected from
`hematopoietic growth factor-stimulated donors, from whom large quantities of cells are collected with each
`apheresis.
`
`CRYOPROTECTANT SOLUTIONS
`
`Dimethylsulfoxide
`Glycerol and DMSO are colligative cryoprotectants that protect the cell from excessive dehydration as
`extracellular water is drawn into growing ice crystals. The cryoprotectant properties of glycerol were
`described in 1949 (Polge et al, 1949), and those of DMSO 10 years later (Lovelock & Bishop, 1959). Both
`have been used for the cryopreservation of bone marrow. It is because of the rapid diffusion of DMSO through
`the cell membrane, and the difficulty in removing glycerol before reinfusion (infusion of DMSO-containing
`products without washing is generally tolerated), that DMSO became the favored agent for HSC cryopreser-
`vation.
`Dimethylsulfoxide, a byproduct of paper manufacturing,
`is a hygroscopic polar compound developed
`originally as a solvent for chemicals such as insecticides, fungicides, and herbicides (David, 1972). Pure
`DMSO is a colorless, virtually odorless liquid (sp. gr. 1.108, m.w. 78.13 g/mole), although industrial grades
`may have a strong sulfur odor (Willhite & Katz, 1984). The serum half-life of DMSO is about 20 hr, although
`that of dimethylsulfone (DMS02), a renal-excreted metabolite, 72 hr (Willhite & Katz, 1984). A small
`proportion of DMSO is reduced to dimethylsulfide (DMS), which is expired through the lungs for about 24 hr
`after administration, and which is responsible for the characteristic odor resulting from DMSO infusion. The
`LD50 values (amount of DMSO required to kill 50% of test animals) reported for intravenous infusion of
`DMSO are 3.1-9.2 g/kg for mice, and 2.5 gm/kg for dogs (Willhite & Katz, 1984). The acute toxic dose of
`DMSO for humans has not been determined.
`The optimal concentration of either DMSO or glycerol for the cryoprotection of HSC appears to be about
`10%. In their original report, Lovelock and Bishop demonstrated a dose-response with improving red cell
`survival as the concentration ofDMSO was increased to 15% (Lovelock & Bishop, 1959). The effect ofhigher
`concentrations was not reported. Subsequently, Ragab et al studied the survival of hematopoietic progenitor
`cells (CFC) from human donors after freezing with various concentrations of DMSO (Ragab et al, 1977).
`They found a significant increase in CFC recovery when the concentration ofDMSO was increased from 7.5%
`to 10%. No further improvement was found with an increase to 12.5%. The improvement in CFC recovery
`resulted from a 2-fold increase in nucleated cell recovery from 17.2% to 35.6% and 32.1 %, respectively, after
`washing. Cell recovery before the post-thaw wash did not differ, nor did the numbers of colonies per 105 cells
`plated. The loss ofcells during the wash steps may not reflect direct intravenous infusion, however, and these
`data suggest that HSC tolerate at least a limited range in DMSO concentration.
`High concentrations ofcryoprotectants may incur direct toxicity to the cells being cryopreserved (cf. effects
`In general, cells are more tolerant
`of vitrification solutions on murine embryos discussed above).
`to
`cryoprotectants at reduced temperatures. For example, DMSO is concentrated also during the formation of
`ice. Optimal cryopreservation requires a balance between protection from freeze damage and the occurrence
`of cryoprotectant-induced toxicity. The toxicity of DMSO to the HSC or to the patient has not been fully
`defined. Using in vitro cultures, Douay et al described a 23.5% recovery of CFU-C after 60-min exposure (at
`4°C, without cryopreservation) and a 15% recovery after 120 min (Douay et al, 1982). We were unable to
`confirm their findings of DMSO-induced progenitor cell loss in similar studies (Rowley, 1991a). Recoveries
`of nucleated cells, granulocyte-macrophage colony-forming units (CFU-GM), and erythroid burst-forming
`238
`
`Downloaded by University of Michigan from www.liebertpub.com at 05/27/20. For personal use only.
`
`Novo Nordisk Ex. 2040, P. 6
`Mylan Institutional v. Novo Nordisk
`IPR2020-00324
`
`

`

`CRYOPRESERVATION OF HEMATOPOIETIC CELLS
`units (BFU-E) were virtually 100% after exposure to 10% DMSO at either 4°C or 37°C. Only at 20%
`concentration of DMSO did cell clumping cause a decrease in cell recovery to 21.7% (and total CFU-GM
`quantity to 27.1%). The numbers ofmyeloid and erythroid progenitors per 5 x 104 cells plated dropped only
`after exposure to 40% DMSO (3.3% of control after 60 min incubation). Similarly, we found no loss of
`progenitor cells after thawing if the removal of DMSO was delayed, even up to 60 min. One possible
`explanation for the differences between these studies is the purity ofDMSO, a strong solvent. Others have also
`reported no toxicity to hematopoietic progenitors with short exposure to DMSO (Ma et al, 1982), although
`direct addition of DMSO to the culture medium at
`1 % or greater concentration was toxic to cell culture
`(Goldman et al, 1978). On the basis of these studies, we no longer precool the freezing chamber or chill the
`cells before addition of the cryoprotectant solution (although that solution is cooled because of the exothermic
`reaction caused by the initial dilution of DMSO in aqueous solutions).
`In contrast, a high incidence ofgenerally mild, infusion-related morbidity with the reinfusion ofeither bone
`marrow or peripheral blood-derived stem cells has been reported by several centers (Davis et al, 1990;
`Kessinger et al, 1990; Stroncek et al, 1991). DMSO itself has a variety of pharmacologie effects (David,
`1972), which may be compounded by the presence of lysed blood cells and foreign proteins or contaminants
`from nonpharmaceutical grades ofreagents used in the processing. Rarely, anaphylaxis or cardiac dysfunction
`may occur. Sudden and severe hypotension can result from the intravenous infusion of DMSO, presumably
`from histamine-induced vasodilatation (David, 1972). Skin flushing, dyspnea, abdominal cramping, nausea,
`and diarrhea, reported to varying degrees after HSC reinfusion, can also all be attributed to DMSO-induced
`histamine release. In a series of 82 patients who were premedicated with diphenhydramine, Davis et al
`observed increased blood pressure and decreased heart rate, which were maximal about
`1 hr after the
`completion of the marrow infusion (Davis et al, 1990). This probably resulted from the cardiac and peripheral
`vascular effects of DMSO. Cardiac dysrythmias were not observed in that study, although one subsequent
`patient developed a transient 2° heart block about 1 hr after the marrow infusion that persisted for about 6 hr
`before spontaneous resolution (unpublished observation). Published reports of cardiac or pulmonary arrest
`during marrow infusion are rare (Vriesendorp et al, 1984; Rapoport etal, 1991). Painful irritation may occur
`if thawed cells are infused through a peripheral vein. Marrows frozen in 10% DMSO have an average
`osmolality of 1794 mOsm/kg H20 after thawing (Rowley, 1992b), and infusion through a central venous
`catheter is preferred. Despite the hyperosmolality of the products, serum osmolality was not greatly affected
`for patients receiving less than about 1 gram of DMSO per kilogram of body weight (Table 3). Although
`intravascularhemolysis has been reported after DMSO administration to cats (DiStefano & Klahn, 1965), and
`possibly humans (Samoszuk etal, 1983), Davis et al did not detect significant hemolysis in their clinical study
`as reflected by a major change in hematocrit. Hemoglobinuria occurs frequently (Kessinger et al, 1990),
`
`Patient
`1
`2
`3
`4
`5
`6
`7
`8
`
`Table 3. Serum Osmolality Changes Associated With Cryopreserved HSC Reinfusion
`Osmolality
`(mOsmlkg H20)
`Patient weight
`Infusion duration
`Volume
`DMSO
`(kg)
`(gm)
`(ml)
`(min)
`Pre
`Post
`59.9
`100
`289
`293
`11.1
`17
`67.2
`9
`100
`11.1
`291
`303
`80.4
`100
`282
`14
`277
`11.1
`100
`64.3
`290
`31
`11.1
`277
`87.0
`116
`95.0
`856
`315
`ND
`96.4
`100
`28
`11.1
`ND
`277
`79
`95.0
`857
`ND
`297
`147
`96.8
`303
`150
`48.3
`436
`ND
`Blood for serum osmolality testing was obtained immediately before initiation of HSC reinfusion (before mannitol
`administration for patient 4) and immediately (at 6 hours for patient 5) after the completion of the infusion. All patients
`were premedicated with 12.5 gm of mannitol, 50 mg of diphenhydramine, 250 mg of hydrocortisone, and saline
`hydration. Serum osmolality was determined by measuring freezing-point depression. ND = not done.
`239
`
`Downloaded by University of Michigan from www.liebertpub.com at 05/27/20. For personal use only.
`
`Novo Nordisk Ex. 2040, P. 7
`Mylan Institutional v. Novo Nordisk
`IPR2020-00324
`
`

`

`ROWLEY
`presumably resulting from hemolysis of red cells within the HSC inoculum. If large quantities of cells frozen
`in DMSO are to be infused, the infusion can be separated over 2 days to avoid complications from infusion of
`excessive amounts of DMSO.
`Hydroxyethyl starch
`HES is a polymeric substance containing chains of different molecular weights. Initially explored as a
`cryoprotectant for red blood cells, it was found also to be effective for a variety of other cells (Ashwood-Smith
`etal, 1972; Takahashi etal, 1988). The major focus in the study of HSC cryopreservation using extracellular
`cryoprotectants has been their use in combination with penetrating cryoprotectants. In one early study, the
`addition ofpolyvinylpyrrolidone (PVP) to glycerol or DMSO improved the cryopreservation of murine cells
`compared to the use of a penetrating agent alone (van Putten, 1968). Stiff et al froze human cells in a
`combination of 5% DMSO, 6% HES, and 4% human serum albumin, and reported improved progenitor cell
`survival as determined using in vitro cultures (Stiff et al, 1983). They subsequently successfully used this
`mixture of cryoprotectants to cryopreserve the bone marrow of 60 patients (Stiff etal, 1987). No engraftment
`failure was attributed to this technique, or after the cryopreservati

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket