Skip to main content

OPINION article

Front. Immunol., 05 October 2022
Sec. Vaccines and Molecular Therapeutics
This article is part of the Research Topic Methods in Vaccines and Molecular Therapeutics: 2022 View all 17 articles

Strategies in developing dimethyl sulfoxide (DMSO)-free cryopreservation protocols for biotherapeutics

  • Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China

Introduction

Cryopreservation is a technology employed in long-term storage of biologics achieved by cooling to cryogenic temperatures (1, 2). This preservation technique has become increasingly relevant especially in the development and commercialization of cellular therapeutic products (3, 4). Conventional cryopreservation protocols involve the application of the permeating cryoprotectant; dimethyl sulfoxide (DMSO) due its ability to restrict ice nucleation and promote post-thaw viability (4). Although DMSO effectively preserves biologics, it can impair functional recovery (57) and induce a variety of mild to severe toxic effects in patients which must be avoided at all cost when administering immunotherapeutic products (8, 9).

The situation is considerably more critical during vitrification; a freezing method that has attracted heightened recognition as a faster and economic substitute to slow freezing as the unorganized liquid state of water is rapidly transformed to a glassy solid state without ice crystallization (10). In vitrification, high cooling rates and high concentrations (4–8 M) of cryoprotectants are usually required (11); enforcing on the exigency of using non-toxic cryoprotectants because increasing DMSO concentration is not advisable.

Efficacious cryopreservation and biobanking requires the development of safe and consistent storage protocols (12, 13). Favorably, such procedures should be devoid of xenogeneic or toxic components and to this effect, many scientists have put forward for the replacement of DMSO. Several groups have discovered/developed safer alternative cryoprotectants with a range of potential cryoprotection mechanisms like ice recrystallization inhibition, osmolality control, cell membrane stabilization and vitrification (14).

Numerous methods have since emerged to lessen the quantity of DMSO used (mostly by supplementation with other cryoprotectants) (1520) or to completely eliminate DMSO application and these methods may also require special adjunct treatments, reagents or freezing protocols as would be discussed here in specifics. Attempts have also been made at preserving cellular products at non-freezing temperatures (2125), advanceable by hypoxia and hypercapnia induced cytoprotection (2628). Although highly beneficial to low-income countries where biobanking facilities are not obtainable, hypothermic storage is however limited to a few days thus fueling the need for safer freezing protocols.

But for the review by Weng et al. (29), studies on replacing DMSO are yet to be critically analyzed; a process that would track research accomplishments, expose novel supplementary techniques applied and encourage further research aimed at improving DMSO-free cryopreservation protocols for different biologicals. Through this article, we wish to draw the attention of researchers to possibility of a DMSO-free preservation era which is achievable in the nearest future. We provide prove of total exclusion of DMSO from cryopreservation solutions and summarize some of the supplementary techniques that have been applied to improve post-thaw viability and function. Our survey has also revealed the commercial availability of DMSO-free cryoprotectant solutions especially those used for cellular therapeutics, but there are limited studies to scrutinize or validate the potency of these products; which might be why most researchers are yet to largely patronizing products without DMSO as there is little to no evidence to back up these product claims. We therefore urge researchers to extend the application of these products to a wider range of biotherapeutics so as to speed up the availability of clinically approved products especially immunotherapeutics which is the answer to many complicated diseases like cancers.

Challenges in cryopreservation with DMSO

Cryopreservation is an important determinant of the stability and activity of biopharmaceutical formulations. Several studies have proven the hypothesis that the application of DMSO can induce temperature-, time-, and concentration-dependent toxicities (7, 30). DMSO causes mitochondrial damage to astrocytes (31), and impacts negatively on cellular membrane/cytoskeleton structure and integrity by interacting with proteins and dehydrating lipids (32) as evident in the increased membrane permeability of erythrocytes (33) and altered chromatin conformation in fibroblasts (30). Also, the presence of DMSO in culture medium can induce unwanted stem cell differentiation (34).

Furthermore, repeated DMSO use even at sub-toxic levels can affect cellular epigenetic profile resulting in undesirable phenotypic disturbances (35). For instance, DMSO interferes with DNA methyltransferases and histone modification enzymes of human pluripotent stem cells causing epigenetic variations and reduction in their pluripotency (36, 37). Similarly, murine embryonic stem cells display disrupted mRNA expression levels of several markers following DMSO treatment (38).

Adverse reactions from cardiac, neurological, and gastrointestinal systems have been reported in patients receiving DMSO-containing cellular products (39, 40). These discoveries have led to the design of several washing procedures to ensure complete DMSO removal. However, the washing protocol usually involves agitation and osmotic/mechanical stresses which are to be avoided due to the fragile and sensitive nature of biologics post-thaw (40). The washing step can also be time consuming, expensive and resource wasting since a significant number of cells are loss in the process.

Strategies in DMSO-free cryopreservation of biotherapeutics

Potent and safe alternative cryoprotectants to DMSO are highly desirable in order to meet the demands in the development and manufacturing of cellular and genetic therapies. In numerous instances, the observed cryoprotective effect is derived from a combination of two or more strategies as discussed below. These strategies and their outcome are also summarized in Table 1.

TABLE 1
www.frontiersin.org

Table 1 Methods/Techniques used in DMSO-free cryopreservation of biotherapeutics.

Alternative cryoprotectants to DMSO

Replacing DMSO with other cryoprotectants is the typical approach in eradicating the use of DMSO in cryopreservation. Kuleshova et al. vitrified neural stem cells using a combination of ethylene glycol (EG) and sucrose. Post-storage evaluations revealed no substantial differences between fresh and vitrified cells in cell markers expression, proliferation or multipotent differentiation (51).

Osmolyte-based freezing solutions containing varying blends of sucrose, glycerol, creatine, isoleucine and mannitol have supported the recovery and survival of mesenchymal stromal cells when compared to conventional preservation with DMSO. These solutions conferred cryoprotection, retained cell differentiation capacity and modulated the cytosine-phosphate-guanine epigenome (54). StemCell Keep™ has been proven effective for the cryopreservation of human induced pluripotent stem cells (hiPSCs) (68), human embryonic stem cells (hESCs) (46) and mesenchymal stem cells (MSCs) (47). Further investigation on the mechanism of cryoprotection showed that the polyampholyte is adsorbed on to the cell membrane suggesting that it can confer protection on cell surface and eliminate the use of proteins and DMSO (47). Other polyampholytes have also shown great potential as DMSO substitutes in storage of murine L929 cells and rat MSCs (69), natural killer cells (59) and other mammalian cell types (70).

A cocktail of non-toxic and Food and Drug Administration (US-FDA)-approved infusible substances including sucrose, glycerol, isoleucine, human serum albumin, and poloxamer 188, have been applied for preservation of hiPSCs (52). In a recent study by Park et al., a block copolymer; PEG−PA (5000−500) has been presented as an excellent cryoprotectant where the recovered stem cells exhibited acceptable survival, proliferation and multilineage differentiation post-thaw (56).

These studies and more presented in Table 1 portray the synergistic activity of several biocompatible cryoprotectants and suggests that this approach may be an innovative paradigm for safe cryopreservation. Presently, commercially manufactured DMSO-free cryosolutions like Pentaisomaltose™, CryoScarless™, CryoNovo P24™, StemCell Keep™, CryoSOfree™ and XT-Thrive™ are available but their quality and capacity to protect a wide range of therapeutics is under-investigated. Further research would involve testing the compatibility of these cryoprotectants with other biotherapeutics. In addition to substituting cryoprotectants, supporting techniques used to improve on solvent-free cryopreservation are discussed below.

Pre-cryopreservation treatment

The pretreatment of biotherapeutics with cryoprotective and stabilizing agents prior to cryopreservation is largely becoming a viable approach to ensuring safe storage. Sugar pretreatment, supplementation of expansion medium with 10% platelet lysate and slow freezing is reportedly an effective protocol in DMSO-less cryopreservation of adipose-derived stromal cells (42). Similarly, sugar pretreatment increased survival, metabolic activity, attachment, proliferation and multilineage differentiation after recultivation of dermal MSCs (43). Improved results are also obtained when the sugars are positioned intracellularly as performed by Mutsenko and coworkers who explored electroporation-aided delivery of cryoprotective sugars in human umbilical cord MSCs (44). Also, pre-incubation of MSCs with osmolyte-based freezing solutions could foster effective cryopreservation (54). These results corroborate the potential advantages of pre-cryopreservation treatment(s).

Programmed freezing methods

Programmed freezing offers improved control over ice nucleation parameters. A technique involving a magnetic field driven freezer termed “Cells Alive System” has been used to prevent formation of intracellular ice for up to three months. The magnetic field vibration function prohibits water molecules from creating clusters during freezing. Although the optimal conditions needed for survival and viability of isolated human periodontal ligament cells (PDL), pulp tissue and tooth using CAS freezers were determined previously (71) (72) with DMSO, the technique proved equally effective without DMSO, promoting greater survival rates over that obtained with conventional freezers (50). Programmed freezing has also been used for cryopreservation of human Wharton’s Jelly Tissue, showing higher post-thaw cell survivability when used in conjunction with a freezing solution consisting of 0.05 M glucose, 0.05 M sucrose and 1.5 M EG in PBS (49).

Matsumura et al. reports a simple, novel slow vitrification method at 4.9 and 10.8°C/min for the cryopreservation of MSC monolayers using a polyampholyte based vitrification solution. Thermal analysis confirmed stable vitrification and post-thaw assessment revealed significantly improved viability and retained differentiation capacity (58).

Thawing protocol

Due to the low thermal conductivity of biological samples, the conventional approach of rewarming large-volume cryopreserved samples in a water bath heated at 37°C is associated with non-uniform distribution of temperature, which can induce thermal stress (41). Preferably, a high heating rate is desirable during thawing, because devitrification and recrystallization may occur if the temperature cannot be elevated rapidly above the sample’s melting point (73). Therefore, both the heating rate and uniformity of heating during rewarming are important to cryopreservation especially vitrification. Nevertheless, attaining the optimal rewarming rate remains a major factor complicating effective vitrification.

Wang et al. proposes magnetic induction heating (MIH) of extracellular Fe3O4 magnetic nanoparticles also called nano-warming technology as a method to amplify rewarming. This technique was successfully applied to rewarming vitrified MSCs where the sample was thawed by plunging the straw into a 0.2 M trehalose supplemented culture medium heated to 37°C. Then, the system was subjected to MIH under alternating magnetic field at a medium frequency for a duration of 10 s. Results obtained reveals the prospective benefits this technique holds as it significantly hindered ice recrystallization/devitrification during rewarming and improved cell viability (41). More recently, Ito et al. also employed the nano-warming technology for thawing of hiPSC using StemCell Keep as cryoprotectant. Similarly, nano-warming showed more uniform and rapid rewarming of vitrified samples, prevented devitrification/recrystallization and improved viability (45).

Another nanotechnology assisted thawing approach involves the utilization of soft liquid metal nanoparticles possessing reproducible photothermal stability, high photothermal conversion efficiency, low cytotoxicity and the ability to suppress ice formation. This technique promotes less ice nucleation during freezing and ultrarapid rewarming while thawing. Human bone marrow stromal cells have been successfully rewarmed with this technique (57).

These studies reveal that the advantages of nanotechnology can be capitalized on to promote safe rewarming post-cryopreservation after verifying the biocompatibility of the nanoparticles.

Conclusion

A critical step prior to the clinical application of biotherapeutics is the optimization of cryopreservation protocols that minimize post-thaw alterations in the stability and potency of preserved materials. Cryosolutions containing 10% of DMSO is a widely used cryopreservative but there is an increasing amount of evidence showing inconsistent results on its impact on post-cryopreservation performance of biologicals. This drawback forms the basis for the development of safer preservation protocols. DMSO-free strategies have the potential to alleviate the aforementioned obstacles as demonstrated by studies discussed in this article. The application of other cryoprotectants, combined with other techniques like programmed freezing, pretreatments and modified thawing protocols have shown good prospects.

In conclusion, the development of effective DMSO-free cryopreservation techniques that will provide high post-thaw viability and preserve original morphology and functioning remains key because this is essential to hastening the industrialization and clinical application of biotherapeutics. We are of the opinion that more research efforts should be put into the development and performance validation of trademarked DMSO-free products. In cases where DMSO elimination is unavoidable, only confirmed safe concentrations should be applied preferably in combination with non-toxic cryoprotectants and other potent strategies.

Author contributions

Conceptualization: ME, CC, ST. Writing—original draft preparation: ME, JX, XL, GB. Writing—review and editing: ME, XL, GB, CC and ST. Supervision and approval: ST. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Pomeroy KO, Comizzoli P, Rushing JS, Lersten IL, Nel-Themaat L. The ART of cryopreservation and its changing landscape. Fertil Steril (2022) 117(3):469–76. doi: 10.1016/j.fertnstert.2022.01.018

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Betsy J, Kumar S. Cryopreservation: History and development. In: Betsy J, Kumar S, editors. Cryopreservation of fish gametes. Singapore: Springer Singapore (2020). p. 135–49.

Google Scholar

3. Hunt CJ. Technical considerations in the freezing, low-temperature storage and thawing of stem cells for cellular therapies. Transfus Med Hemother (2019) 46(3):134–50. doi: 10.1159/000497289

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Whaley D, Damyar K, Witek RP, Mendoza A, Alexander M, Lakey JR. Cryopreservation: An overview of principles and cell-specific considerations. Cell Transplant (2021) 30:963689721999617. doi: 10.1177/0963689721999617

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Li R, Walsh P, Truong V, Petersen A, Dutton JR, Hubel A. Differentiation of human iPS cells into sensory neurons exhibits developmental stage-specific cryopreservation challenges. Front Cell Dev Biol (2021) 9:796960. doi: 10.3389/fcell.2021.796960

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Mizuno M, Matsuzaki T, Ozeki N, Katano H, Koga H, Takebe T, et al. Cell membrane fluidity and ROS resistance define DMSO tolerance of cryopreserved synovial MSCs and HUVECs. Stem Cell Res Ther (2022) 13(1):177. doi: 10.1186/s13287-022-02850-y

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Awan M, Buriak I, Fleck R, Fuller B, Goltsev A, Kerby J, et al. Dimethyl sulfoxide: a central player since the dawn of cryobiology, is efficacy balanced by toxicity? Regener Med (2020) 15(3):1463–91. doi: 10.2217/rme-2019-0145

CrossRef Full Text | Google Scholar

8. Ock SA, Rho GJ. Effect of dimethyl sulfoxide (DMSO) on cryopreservation of porcine mesenchymal stem cells (pMSCs). Cell Transplant (2011) 20(8):1231–9. doi: 10.3727/096368910x552835

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Svalgaard JD, Talkhoncheh MS, Haastrup EK, Munthe-Fog L, Clausen C, Hansen MB, et al. Pentaisomaltose, an alternative to DMSO. engraftment of cryopreserved human CD34(+) cells in immunodeficient NSG mice. Cell Transplant (2018) 27(9):1407–12. doi: 10.1177/0963689718786226

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Sharma A, Rao JS, Han Z, Gangwar L, Namsrai B, Gao Z, et al. Vitrification and nanowarming of kidneys. Adv Sci (Weinh) (2021) 8(19):e2101691. doi: 10.1002/advs.202101691

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Murray KA, Gibson MI. Chemical approaches to cryopreservation. Nat Rev Chem (2022) 6(8):579–93. doi: 10.1038/s41570-022-00407-4

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Zhang TY, Tan PC, Xie Y, Zhang XJ, Zhang PQ, Gao YM, et al. The combination of trehalose and glycerol: An effective and non-toxic recipe for cryopreservation of human adipose-derived stem cells. Stem Cell Res Ther (2020) 11(1):460. doi: 10.1186/s13287-020-01969-0

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Raju R, Bryant SJ, Wilkinson BL, Bryant G. The need for novel cryoprotectants and cryopreservation protocols: Insights into the importance of biophysical investigation and cell permeability. Biochim Biophys Acta Gen Subj (2021) 1865(1):129749. doi: 10.1016/j.bbagen.2020.129749

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Arutyunyan IV, Kananykhina EY, Elchaninov AV, Fatkhudinov TK. Influence of sucrose on the efficiency of cryopreservation of human umbilical cord-derived multipotent stromal cells with the use of various penetrating cryoprotectants. Bull Exp Biol Med (2021) 171(1):150–5. doi: 10.1007/s10517-021-05187-3

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Kaiser D, Otto NM, McCallion O, Hoffmann H, Zarrinrad G, Stein M, et al. Freezing medium containing 5% DMSO enhances the cell viability and recovery rate after cryopreservation of regulatory T cell products ex vivo and in vivo. Front Cell Dev Biol (2021) 9:750286. doi: 10.3389/fcell.2021.750286

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Tuten Sevim E, Arat S. Combining dimethyl sulphoxide (DMSO) with different cryoprotectants ensures better cartilage cell cryopreservation. Cryo Lett (2021) 42(4):220–6.

Google Scholar

17. Gilfanova R, Auclair KM, Hui A, Norris PJ, Muench MO. Reduced dimethyl sulfoxide concentrations successfully cryopreserve human hematopoietic stem cells with multi-lineage long-term engraftment ability in mice. Cytotherapy (2021) 23(12):1053–9. doi: 10.1016/j.jcyt.2021.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Jahan S, Kaushal R, Pasha R, Pineault N. Current and future perspectives for the cryopreservation of cord blood stem cells. Transfus Med Rev (2021) 35(2):95–102. doi: 10.1016/j.tmrv.2021.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Trummer T, Fox R, Koç JR, de Lima M, Otegbeye F. Cryopreservation of hematopoietic cells using a pre-constituted, protein-free cryopreservative solution with 5% dimethyl sulfoxide. Cytotherapy (2020) 22(11):613–6. doi: 10.1016/j.jcyt.2020.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Duchez P, Chevaleyre J, Brunet de la Grange P, Vlaski M, Boiron JM, Wouters G, et al. Cryopreservation of hematopoietic stem and progenitor cells amplified ex vivo from cord blood CD34+ cells. Transfusion (2013) 53(9):2012–9. doi: 10.1111/trf.12015

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Bekadja MA, Boumendil A, Blaise D, Chevallier P, Peggs KS, Salles G, et al. Non-cryopreserved hematopoietic stem cells in autograft patients with lymphoma: a matched-pair analysis comparing a single center experience with the use of cryopreserved stem cells reported to the European society for blood and marrow transplantation registry. Cytotherapy (2021) 23(6):483–7. doi: 10.1016/j.jcyt.2020.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Araújo AB, Soares TB, Schmalfuss T, Angeli MH, Furlan JM, Salton GD, et al. Non-cryopreserved peripheral blood stem cells as a safe and effective alternative for autologous transplantation in multiple myeloma. Transfusion (2022). doi: 10.1111/trf.17090

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Jennane S, Hasnaoui N, Mahtat EM, Merimi F, Bougar S, El Maaroufi H, et al. Non-cryopreserved peripheral blood stem cells autologous transplantation in multiple myeloma: Bicentric study. Transfus Clin Biol (2020) 27(3):152–6. doi: 10.1016/j.tracli.2020.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Piriyakhuntorn P, Tantiworawit A, Rattanathammethee T, Hantrakool S, Chai-Adisaksopha C, Rattarittamrong E, et al. Outcomes of non-cryopreserved versus cryopreserved peripheral blood stem cells for autologous stem cell transplantation in multiple myeloma. Ann Transplant (2020) 25:e927084. doi: 10.12659/aot.927084

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Bittencourt MCB, Mariano L, Moreira F, Schmidt-Filho J, Mendrone-Jr A, Rocha V. Cryopreserved versus non-cryopreserved peripheral blood stem cells for autologous transplantation after high-dose melphalan in multiple myeloma: comparative analysis. Bone Marrow Transplant (2019) 54(1):138–41. doi: 10.1038/s41409-018-0250-1

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Jeanne M, Kovacevic-Filipovic M, Szyporta M, Vlaski M, Hermitte F, Lafarge X, et al. Low-oxygen and high-carbon-dioxide atmosphere improves the conservation of hematopoietic progenitors in hypothermia. Transfusion (2009) 49(8):1738–46. doi: 10.1111/j.1537-2995.2009.02191.x

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Vlaski M, Negroni L, Kovacevic-Filipovic M, Guibert C, Brunet de la Grange P, Rossignol R, et al. Hypoxia/hypercapnia-induced adaptation maintains functional capacity of cord blood stem and progenitor cells at 4°C. J Cell Physiol (2014) 229(12):2153–65. doi: 10.1002/jcp.24678

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Gerby S, Simplicien M, Duchez P, Chevaleyre J, Ivanovic Z, Vlaski-Lafarge M. Hypoxia/hypercapnia prevents iron-dependent cold injuries in cord blood stem and progenitor cells. Cytotherapy (2019) 21(4):460–7. doi: 10.1016/j.jcyt.2019.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Weng L, Beauchesne PR. Dimethyl sulfoxide-free cryopreservation for cell therapy: A review. Cryobiology (2020) 94:9–17. doi: 10.1016/j.cryobiol.2020.03.012

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Verheijen M, Lienhard M, Schrooders Y, Clayton O, Nudischer R, Boerno S, et al. DMSO induces drastic changes in human cellular processes and epigenetic landscape in vitro. Sci Rep (2019) 9(1):4641. doi: 10.1038/s41598-019-40660-0

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Yuan C, Gao J, Guo J, Bai L, Marshall C, Cai Z, et al. Dimethyl sulfoxide damages mitochondrial integrity and membrane potential in cultured astrocytes. PloS One (2014) 9(9):e107447. doi: 10.1371/journal.pone.0107447

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Cheng CY, Song J, Pas J, Meijer LH, Han S. DMSO induces dehydration near lipid membrane surfaces. Biophys J (2015) 109(2):330–9. doi: 10.1016/j.bpj.2015.06.011

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Gironi B, Kahveci Z, McGill B, Lechner BD, Pagliara S, Metz J, et al. Effect of DMSO on the mechanical and structural properties of model and biological membranes. Biophys J (2020) 119(2):274–86. doi: 10.1016/j.bpj.2020.05.037

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Sambo D, Li J, Brickler T, Chetty S. Transient treatment of human pluripotent stem cells with DMSO to promote differentiation. J Vis Exp (2019) 149:1–11. doi: 10.3791/59833

CrossRef Full Text | Google Scholar

35. Cheng H, Han Y, Zhang J, Zhang S, Zhai Y, An X, et al. Effects of dimethyl sulfoxide (DMSO) on DNA methylation and histone modification in parthenogenetically activated porcine embryos. Reproduction. Fertil Dev (2022) 34(8):598–607. doi: 10.1071/RD21083

CrossRef Full Text | Google Scholar

36. Ntai A, La Spada A, De Blasio P, Biunno I. Trehalose to cryopreserve human pluripotent stem cells. Stem Cell Res (2018) 31:102–12. doi: 10.1016/j.scr.2018.07.021

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Fu K, Bonora G, Pellegrini M. Interactions between core histone marks and DNA methyltransferases predict DNA methylation patterns observed in human cells and tissues. Epigenetics (2020) 15(3):272–82. doi: 10.1080/15592294.2019.1666649

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Yi JK, Park S, Ha JJ, Kim DH, Huang H, Park SJ, et al. Effects of dimethyl sulfoxide on the pluripotency and differentiation capacity of mouse embryonic stem cells. Cell Reprogram (2020) 22(5):244–53. doi: 10.1089/cell.2020.0006

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Kollerup Madsen B, Hilscher M, Zetner D, Rosenberg J. Adverse reactions of dimethyl sulfoxide in humans: A systematic review. F1000Res (2018) 7:1746. doi: 10.12688/f1000research.16642.2

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Xie J, Ekpo MD, Xiao J, Zhao H, Bai X, Liang Y, et al. Principles and protocols for post-cryopreservation quality evaluation of stem cells in novel biomedicine. Front Pharmacol (2022) 13:907943. doi: 10.3389/fphar.2022.907943

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Wang J, Zhao G, Zhang Z, Xu X, He X. Magnetic induction heating of superparamagnetic nanoparticles during rewarming augments the recovery of hUCM-MSCs cryopreserved by vitrification. Acta Biomater (2016) 33:264–74. doi: 10.1016/j.actbio.2016.01.026

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Rogulska O, Petrenko Y, Petrenko A. DMSO-free cryopreservation of adipose-derived mesenchymal stromal cells: expansion medium affects post-thaw survival. Cytotechnology (2017) 69(2):265–76. doi: 10.1007/s10616-016-0055-2

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Petrenko YA, Rogulska OY, Mutsenko VV, Petrenko AY. A sugar pretreatment as a new approach to the Me2SO- and xeno-free cryopreservation of human mesenchymal stromal cells. Cryo Lett (2014) 35(3):239–46.

Google Scholar

44. Mutsenko V, Barlič A, Pezić T, Dermol-Černe J, Dovgan B, Sydykov B, et al. Me2SO- and serum-free cryopreservation of human umbilical cord mesenchymal stem cells using electroporation-assisted delivery of sugars. Cryobiology (2019) 91:104–14. doi: 10.1016/j.cryobiol.2019.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Ito A, Yoshioka K, Masumoto S, Sato K, Hatae Y, Nakai T, et al. Magnetic heating of nanoparticles as a scalable cryopreservation technology for human induced pluripotent stem cells. Sci Rep (2020) 10(1):13605. doi: 10.1038/s41598-020-70707-6

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Ota A, Matsumura K, Lee JJ, Sumi S, Hyon SH. StemCell keep™ is effective for cryopreservation of human embryonic stem cells by vitrification. Cell Transplant (2017) 26(5):773–87. doi: 10.3727/096368916x692654

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Matsumura K, Hayashi F, Nagashima T, Hyon SH. Long-term cryopreservation of human mesenchymal stem cells using carboxylated poly-l-lysine without the addition of proteins or dimethyl sulfoxide. J Biomater Sci Polym Ed (2013) 24(12):1484–97. doi: 10.1080/09205063.2013.771318

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Katkov I, Kan N, Cimadamore F, Nelson B, Snyder E, Terskikh A. DMSO-free programmed cryopreservation of fully dissociated and adherent human induced pluripotent stem cells. Stem Cells Int (2011) 981606:8. doi: 10.4061/2011/981606

CrossRef Full Text | Google Scholar

49. Shivakumar SB, Bharti D, Subbarao RB, Jang SJ, Park JS, Ullah I, et al. DMSO- and serum-free cryopreservation of wharton’s jelly tissue isolated from human umbilical cord. J Cell Biochem (2016) 117(10):2397–412. doi: 10.1002/jcb.25563

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Kawata T, Abedini S, Kaku M, Koseki H, Kojima S, Sumi H, et al. Effects of DMSO (Dimethyl sulfoxide) free cryopreservation with program freezing using a magnetic field on periodontal ligament cells and dental pulp tissues. Biomed Res (2012) 23(3):438–43.

Google Scholar

51. Kuleshova LL, Tan FCK, Magalhães R, Gouk SS, Lee KH, Dawe GS. Effective cryopreservation of neural stem or progenitor cells without serum or proteins by vitrification. Cell Transplant (2009) 18(2):135–44. doi: 10.3727/096368909788341298

CrossRef Full Text | Google Scholar

52. Li R, Hornberger K, Dutton JR, Hubel A. Cryopreservation of human iPS cell aggregates in a DMSO-free solution-an optimization and comparative study. Front Bioeng Biotechnol (2020) 8:1. doi: 10.3389/fbioe.2020.00001

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Deller RC, Vatish M, Mitchell DA, Gibson MI. Synthetic polymers enable non-vitreous cellular cryopreservation by reducing ice crystal growth during thawing. Nat Commun (2014) 5:3244. doi: 10.1038/ncomms4244

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Pollock K, Samsonraj RM, Dudakovic A, Thaler R, Stumbras A, McKenna DH, et al. Improved post-thaw function and epigenetic changes in mesenchymal stromal cells cryopreserved using multicomponent osmolyte solutions. Stem Cells Dev (2017) 26(11):828–42. doi: 10.1089/scd.2016.0347

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Pollock K, Yu G, Moller-Trane R, Koran M, Dosa PI, McKenna DH, et al. Combinations of osmolytes, including monosaccharides, disaccharides, and sugar alcohols act in concert during cryopreservation to improve mesenchymal stromal cell survival. Tissue Eng Part C Methods (2016) 22(11):999–1008. doi: 10.1089/ten.TEC.2016.0284

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Park JK, Patel M, Piao Z, Park S-J, Jeong B. Size and shape control of ice crystals by amphiphilic block copolymers and their implication in the cryoprotection of mesenchymal stem cells. ACS Appl Mater Interfaces (2021) 13(29):33969–80. doi: 10.1021/acsami.1c09933

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Hou Y, Lu C, Dou M, Zhang C, Chang H, Liu J, et al. Soft liquid metal nanoparticles achieve reduced crystal nucleation and ultrarapid rewarming for human bone marrow stromal cell and blood vessel cryopreservation. Acta Biomater (2020) 102:403–15. doi: 10.1016/j.actbio.2019.11.023

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Matsumura K, Kawamoto K, Takeuchi M, Yoshimura S, Tanaka D, Hyon SH. Cryopreservation of a two-dimensional monolayer using a slow vitrification method with polyampholyte to inhibit ice crystal formation. ACS Biomater Sci Eng (2016) 2(6):1023–9. doi: 10.1021/acsbiomaterials.6b00150

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Pasley S, Zylberberg C, Matosevic S. Natural killer-92 cells maintain cytotoxic activity after long-term cryopreservation in novel DMSO-free media. Immunol Lett (2017) 192:35–41. doi: 10.1016/j.imlet.2017.09.012

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Mitchell DE, Lovett JR, Armes SP, Gibson MI. Combining biomimetic block copolymer worms with an ice-inhibiting polymer for the solvent-free cryopreservation of red blood cells. Angew Chem Int Ed Engl (2016) 55(8):2801–4. doi: 10.1002/anie.201511454

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Rao W, Huang H, Wang H, Zhao S, Dumbleton J, Zhao G, et al. Nanoparticle-mediated intracellular delivery enables cryopreservation of human adipose-derived stem cells using trehalose as the sole cryoprotectant. ACS Appl Mater Interfaces (2015) 7(8):5017–28. doi: 10.1021/acsami.5b00655

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Duchez P, Rodriguez L, Chevaleyre J, de la Grange PB, Ivanovic Z. Clinical-scale validation of a new efficient procedure for cryopreservation of ex vivo expanded cord blood hematopoietic stem and progenitor cells. Cytotherapy (2016) 18(12):1543–7. doi: 10.1016/j.jcyt.2016.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Li R, Hornberger K, Dutton JR, Hubel A. Cryopreservation of human iPS cell aggregates in a DMSO-free solution–an optimization and comparative study. Front Bioeng Biotechnol (2020) 8:1. doi: 10.3389/fbioe.2020.00001

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Gilfanova R, Callegari A, Childs A, Yang G, Luarca M, Gutierrez AG, et al. A bioinspired and chemically defined alternative to dimethyl sulfoxide for the cryopreservation of human hematopoietic stem cells. Bone Marrow Transplant (2021) 56(11):2644–50. doi: 10.1038/s41409-021-01368-w

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Kaushal R, Jahan S, McGregor C, Pineault N. Dimethyl sulfoxide-free cryopreservation solutions for hematopoietic stem cell grafts. Cytotherapy (2022) 24(3):272–81. doi: 10.1016/j.jcyt.2021.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Svalgaard JD, Haastrup EK, Reckzeh K, Holst B, Glovinski PV, Gørløv JS, et al. Low-molecular-weight carbohydrate pentaisomaltose may replace dimethyl sulfoxide as a safer cryoprotectant for cryopreservation of peripheral blood stem cells. Transfusion (2016) 56(5):1088–95. doi: 10.1111/trf.13543

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Vanichapol T, Pongsakul N, Srisala S, Apiwattanakul N, Chutipongtanate S, Hongeng S. Suppressive characteristics of umbilical cord blood-derived regulatory T cells after ex vivo expansion on autologous and allogeneic T effectors and various lymphoblastic cells. J Immunother (2019) 42(4):110–8. doi: 10.1097/cji.0000000000000262

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Matsumura K, Bae JY, Kim HH, Hyon SH. Effective vitrification of human induced pluripotent stem cells using carboxylated ϵ-poly-l-lysine. Cryobiology (2011) 63(2):76–83. doi: 10.1016/j.cryobiol.2011.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Matsumura K, Hyon S-H. Polyampholytes as low toxic efficient cryoprotective agents with antifreeze protein properties. Biomaterials (2009) 30(27):4842–9. doi: 10.1016/j.biomaterials.2009.05.025

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Matsumura K, Bae JY, Hyon SH. Polyampholytes as cryoprotective agents for mammalian cell cryopreservation. Cell Transplant (2010) 19(6):691–9. doi: 10.3727/096368910x508780

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Abedini S, Kaku M, Kawata T, Koseki H, Kojima S, Sumi H, et al. Effects of cryopreservation with a newly-developed magnetic field programmed freezer on periodontal ligament cells and pulp tissues. Cryobiology (2011) 62(3):181–7. doi: 10.1016/j.cryobiol.2011.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Kaku M, Kamada H, Kawata T, Koseki H, Abedini S, Kojima S, et al. Cryopreservation of periodontal ligament cells with magnetic field for tooth banking. Cryobiology (2010) 61(1):73–8. doi: 10.1016/j.cryobiol.2010.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Baboo J, Kilbride P, Delahaye M, Milne S, Fonseca F, Blanco M, et al. The impact of varying cooling and thawing rates on the quality of cryopreserved human peripheral blood T cells. Sci Rep (2019) 9(1):3417. doi: 10.1038/s41598-019-39957-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cryopreservation, cryoprotectants, biobanking, dimethyl sulfoxide, DMSO-free, biotherapeutics

Citation: Ekpo MD, Boafo GF, Xie J, Liu X, Chen C and Tan S (2022) Strategies in developing dimethyl sulfoxide (DMSO)-free cryopreservation protocols for biotherapeutics. Front. Immunol. 13:1030965. doi: 10.3389/fimmu.2022.1030965

Received: 29 August 2022; Accepted: 20 September 2022;
Published: 05 October 2022.

Edited by:

Mrinmoy Sanyal, Stanford University, United States

Reviewed by:

Xavier Lafarge, Établissement Français du Sang (EFS), France

Copyright © 2022 Ekpo, Boafo, Xie, Liu, Chen and Tan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Songwen Tan, songwen.tan@csu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.