Immune Netw. 2023 Dec;23(6):e44. English.
Published online Nov 23, 2023.
Copyright © 2023. The Korean Association of Immunologists
Review

Comparing the Benefits and Drawbacks of Stem Cell Therapy Based on the Cell Origin or Manipulation Process: Addressing Immunogenicity

Sung-Ho Chang,1 and Chung Gyu Park2,3,4,5
    • 1Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Korea.
    • 2Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.
    • 3Transplantation Research Institute, Medical Research center, Seoul National University College of Medicine, Seoul 03080, Korea.
    • 4Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
    • 5Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea.
Received October 12, 2023; Accepted November 12, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Mesenchymal stem cells (MSCs) are effective in treating autoimmune diseases and managing various conditions, such as engraftment of allogeneic islets. Additionally, autologous and HLA-matched allogeneic MSCs can aid in the engraftment of human allogeneic kidneys with or without low doses of tacrolimus, respectively. However, HLA alloantigens are problematic because cell therapy uses more HLA-mismatched allogeneic cells than autologous for convenience and standardization. In particular, HLA-mismatched MSCs showed increased Ag-specific T/B cells and reduced viability faster than HLA-matched MSCs. In CRISPR/Cas9-based cell therapy, Cas9 induce T cell activation in the recipient's immune system. Interestingly, despite their immunogenicity being limited to the cells with foreign Ags, the accumulation of HLA alloantigen-sensitized T/B cells may lead to allograft rejection, suggesting that alloantigens may have a greater scope of adverse effects than foreign Ags. To avoid alloantigen recognition, the β2-microglobulin knockout (B2MKO) system, eliminating class-I MHC, was able to avoid rejection by alloreactive CD8 T cells compared to controls. Moreover, universal donor cells in which both B2M and Class II MHC transactivator (CIITA) were knocked out was more effective in avoiding immune rejection than single KO. However, B2MKO and CIITA KO system remain to be controlled and validated for adverse effects such as the development of tumorigenicity due to deficient Ag recognition by CD8 T and CD4 T cells, respectively. Overall, better HLA-matching or depletion of HLA alloantigens prior to cell therapy can reduce repetitive transplantation through the long-term survival of allogeneic cell therapy, which may be especially important for patients seeking allogeneic transplantation.

Keywords
Immunogenicity; HLA-matched/mismatched allogeneic cell therapy; CRISPR/Cas9; B2MKO; Universal cell therapy; Opportunity cost

INTRODUCTION

Mesenchymal stem cells (MSCs) can be obtained from a variety of tissues, such as bone marrow, adipose tissue, and umbilical cord blood, and organs, such as, the spleen, thymus, and kidney, with adipose-derived mesenchymal stem cells (ADSCs), bone marrow-MSCs (BM-MSCs), and umbilical cord-MSCs (UC-MSCs) being the most commonly used (1). In addition to their immunomodulatory effects, MSCs are multipotent progenitor cells that can differentiate into a variety of cell types and have been shown to be therapeutic in a variety of diseases (2, 3, 4, 5, 6). However, the HLA-ABC alloantigens expressed on allogeneic MSCs are highly immunogenic (7, 8, 9), which is a common issue in cell therapy utilizing allogeneic cells (10). Mechanistically, in alloantigen recognition, T cells of the recipient immune system can recognize and act on HLA alloantigens through direct or indirect pathways (Supplementary Fig. 1) (11). Ab-mediated rejection (ABMR) by alloreactive B cells is a widely recognized cause of allograft rejection (11, 12).

CRISPR/Cas9 is the most widely used method for epigenome editing, owing to its efficiency and convenience (13). Specifically, cell therapies involving MSCs and induced pluripotent stem cells (iPSCs) use CRISPR/Cas9 for genome editing (14). However, it is a system of bacterial origin, which raises concerns regarding its potential immunogenicity (15). The expression of foreign Ags from genome-edited cell therapy can trigger an immune response in the recipient's immune system, which can lead to a decrease in therapeutic efficacy and accumulation of sensitized immune cells (16, 17). These foreign Ags can be recognized by T cells via class I and II MHC pathways on Ag presenting cells (18).

This study first examined the various efficacies of MSCs for disease treatment and whether there were differences based on their origin. Second, cell therapy-induced immunogenicity and its causes were investigated in detail. Third, we investigated the benefits and disadvantages of HLA-matched allogeneic cell therapies. Fourth, we discuss the ability of the β2-microglobulin knockout (B2MKO) and universal cell therapies system to evade recipient immune responses to HLA alloantigens. We also review the currently available options for suppressing Ag-specific recipient immune responses. This review of immunogenic-cell therapy will contribute to the identification of the most effective therapeutic approaches while minimizing their adverse effects.

THE UTILITY OF MSCs AS A TREATMENT FOR A VARIETY OF DISEASES

MSCs have therapeutic effects in various diseases, including autoimmune diseases such as systemic lupus erythematosus, severe refractory gingivostomatitis, and oral lichen planus (OLP) (Table 1; reports from 2016 to 2022). Specifically, feline chronic gingivostomatitis (FCGS), an autoimmune disease similar to OLP, is a painful and debilitating inflammatory disease of the oral mucosa that requires lifelong treatment with antibiotics and corticosteroids; however, when MSCs were used to treat FCGS, 4 out of 7 animals (57%) reported clinical improvement (Table 1). HLA-mismatched MSCs (cured by 12–20 months) were also effective in the treatment of FCGS, but less effective than autologous MSCs (cured by 3–9 months) (concentrations of serum IFNγ and neutrophil counts, both p=0.057) (19). In addition, MSCs contributed to improved treatment of ankle non-union in diabetic patients (improving non-unions, p=0.04; reduced infection rate, p<0.01), engraftment of allogeneic islets (improving graft survival, p<0.01), knee osteoarthritis (p<0.05), and radiation-induced xerostomia (increase of 0.06 ml per minute, p=0.0009) (Table 1).

Table 1
Effects of MSCs on the treatment of various diseases

MSCs IN ENGRAFTMENT OF HUMAN KIDNEY TRANSPLANTS: AUTOLOGOUS & HLA-MATCHED ALLOGENEIC CELLS

In human organ transplants, calcineurin inhibitors (CNI; cyclosporine [1980s], tacrolimus [1990s]) provide better protection against acute rejection (25). However, the long-term use of these inhibitors increases the risk of infection and malignancy, which is an obstacle to long-term survival; therefore, MSCs treatment has been utilized as an alternative to CNI (Table 2; reports from 2016 to 2022). Human MSCs can reduce or replace the use of CNI in the engraftment of HLA-matched allogeneic kidneys and suppress rejection for a long time. Specifically, autologous BM-MSCs were able to prevent rejection after the tacrolimus withdrawal, and HLA-matched allogeneic BM-MSCs were able to prevent rejection with a low dose of tacrolimus for two years (Table 2).

Table 2
Effect of autologous- or HLA-matched allogeneic-MSCs on engraftment of human kidney transplantation

STEM CELL THERAPY IMMUNOGENICITY

Causes of immunogenicity: HLA-alloantigens, xenogeneic molecules, and foreign Ags from genome editing system

Cell therapy is promising for the treatment of various diseases; however, immunogenicity remains challenging (Table 3; reports from 2015 to 2023). Thus, understanding the causes and impact of immunogenicity is essential for the safe use of cell therapy. Kol et al. (30) reported that HLA-mismatched MSCs induce an immune response in alloreactive Abs or cytotoxic CD8 T cells. These results are supported by reports that HLA-ABC alloantigens expressed by MSCs induce alloreactive CD8 T cell activity primarily through a direct pathway (Supplementary Fig. 1) (8). Interestingly, rejection of HLA-mismatched MSCs can be analyzed both in vivo and in vitro approximately 3 weeks after injection, compared to mixed lymphocyte responses in donors and recipients, which are detectable at approximately 7 days (8, 31, 34, 41). These results correlate with MSCs having immunomodulatory activity and relatively low expression of HLA alloantigens, which may be a hallmark of MSCs, leading to a later onset of rejection (11). However, T cells specific for alloantigens on allogeneic MSCs do not respond to immunosuppressants (36, 38). Večerić-Haler et al. (29) also reported that autologous MSCs used for immunomodulation were ineffective as inhibitors of chronic ABMR, suggesting that, even if MSCs have immunomodulatory effects, they are limited in suppressing Ag-specific rejection (Tables 14) (8). These results are also supported by reports that HLA-mismatched allogeneic cell therapy results in a lower survival rate than autologous cell therapy (Table 3). In iPSC cell therapy, it has been reported that HLA-matched iPSC-neurons have a higher survival rate than mismatched cells in the brain, a tissue known to have a low immune response, proving that the same applies to allogeneic cell therapy (17, 48).

Table 4
Comparative analysis of immunogenicity for autologous-versus allogeneic-cell therapy

Other causes of immunogenicity include foreign Ags and xenogeneic molecules used in the genetic manipulation and culture of cell therapy, respectively. First, the Cas9 protein from CRISPR/Cas9, which is used to generate iPSCs, can induce the activity of CD8 T cells as well as Ag-specific CD4 T cells (36). This is further supported by recent reports that plasmacytoid dendritic cells (DCs), as well as conventional DCs, which are known to be potent cross-presenting dendritic cells, can cross-present extracellular Ags to Class I MHC (18). However, it is encouraging that Wagner et al. (36) have shown that Ag-specific Treg cells are effective in suppressing the activity of Cas9-specific T cells. Second, fetal bovine serum, commonly used for the culture and proliferation of MSCs, promotes cytotoxic CD8 T cell activation and kills HLA-mismatched MSCs more rapidly than human serum (8, 35). These results correlate with reports that albumin can bind to a variety of tissues and cells and be absorbed intracellularly (49, 50). This means that fetal bovine serum must be replaced with human serum or xenofree material before use (8, 51).

Comparison of immunogenicity in autologous– versus HLA-mismatched allogeneic–cell therapy

Data comparing the immunogenicity of allogeneic and autologous cells in MSCs therapy have been examined (Table 4; reports from 2012 to 2023). The results showed that HLA-mismatched MSCs induced an increase in B cells and the production of alloreactive Abs compared to autologous MSCs (42, 44). In addition, HLA-mismatched MSCs induce significant T lymphocyte infiltration compared to autologous MSCs (44, 45, 46). Ultimately, the immunogenicity of HLA-mismatched MSCs resulted in a significant decrease in the survival rate compared to that of autologous MSCs (8, 24). Interestingly, even immunogenic HLA-mismatched MSCs were effective in prolonging the survival of allogeneic islets without serious adverse effects (Table 1) (19, 24). These results suggest that HLA-mismatched MSCs may partially contribute their immunomodulatory effects, even if they trigger Ag-specific immune responses in the recipient immune system. However, immunogenic-cell therapy may eventually be eliminated by rejection by the recipient’s immune system, which may cause routine repeated administration and result in adverse effects (Tables 3 and 4).

BENEFITS OR DEFECTS OF HLA-MATCHED ALLOGENEIC-CELL THERAPY

Autologous-derived cell therapies may be the most effective choice from an immunological safety and functional standpoint. However, they have the disadvantage of not being immediately available. In particular, patient-derived autologous cell therapy may not be functionally effective (21). Thus, HLA-matched allogeneic-cell therapies may be an alternative to autologous cell therapies. Table 5 compares the advantages and disadvantages of HLA-matched cell therapy based on Tables 14. Most importantly, HLA-matched allogeneic cells can lower the induction of alloreactive memory T/B cells in recipients compared to mismatched allogeneic cells (47). In addition, the long-term survival of HLA-matched allogeneic cells reduces the number of repeated cell treatments (24, 47, 52, 53, 54). Although HLA-matched allogeneic cells have disadvantages in terms of convenience, they have the potential to be used effectively once national/international cell banking systems are well established, as their immunogenicity can be minimized (Fig. 1) (55).

Table 5
Benefits and defects of HLA-matched allogeneic cell therapy

Figure 1
Overview of effective HLA matching in preparing allogeneic cell therapy. (a-c) Cultivation of allogeneic cell therapy (replacing FBS with human serum) and manipulation using genome editing system. (d) For better HLA-matched cell therapy, national/international cell banking systems may be effective (17, 55). (e) Preparation of autologous cell therapy. (f) Treat patients with ready-made cell therapy. This figure was created using BioRender.com.

DIFFERENCES IN THE IMMUNOGENICITY OF ALLOGENEIC– AND GENOME-EDITED–THERAPEUTIC CELLS

Both allogeneic and genome-edited cell therapies cause immunogenicity in patients upon transplantation (Tables 3 and 4); however, there are differences in the consequences of their immunogenicity. Table 6 presents the differences in immunogenicity according to the origin of the cell therapy and genetic manipulation based on Tables 35. Specifically, HLA-matched therapeutic cells with edited genomes are primarily associated with cytotoxic responses and the accumulation of memory cells in response to foreign Ags rather than the rejection of alloantigens (14, 36, 38, 53, 56, 57). In other words, the effect of immunogenicity due to foreign Ags is limited to the cell populations associated with these Ags. In contrast, cell therapy using allogeneic cells sensitizes memory T/B cells to HLA-alloantigens, leading to their accumulation (36, 58, 59). Thus, immunogenicity problems caused by HLA alloantigens may be associated with a much broader range of adverse effects than those caused by foreign Ags.

Table 6
Comparison of immunogenicity differences between allogeneic- and genome edited-therapeutic cells

HOW TO AVOID HLA-ALLOANTIGEN SENSITIZATION 1: B2MKO CELL THERAPY

Recently, attempts have been made to eliminate class I MHC as a major cause of allogeneic rejection. B2MKO iPSCs/iMSCs lacking class I HLA presentation were able to evade rejection by alloreactive CD8 T cells compared to wild-type controls (60). However, cells lacking class I HLA may have been targeted by NK cells (61, 62), thus increasing their susceptibility to NK cell lysis (p<0.001) (60). NK cell lysis in B2MKO cell therapy could be suppressed by knock-in of HLA-E or HLA-G at the B2M locus, which would not induce allogeneic responses (62, 63). However, B2MKO cell therapy may be fundamentally vulnerable because it is difficult for the recipient's immune system to function in the event of tumorigenesis or exposure to infection. Defects in B2M gene expression have been reported as the basic mechanism of drug resistance in patients with lung cancer (64, 65). Thus, B2MKO cell therapy may be considered for the application of a safety system, such as a suicide system introduced into CAR-T cells, to control the occurrence of serious adverse effects (66). Three types of suicide systems have been reported:1) HSV-1 thymidine kinase suicide genes with immunogenicity (67, 68), 2) human origin of the iCaspase 9 suicide system (69, 70), and 3) uridine monophosphate synthetase-knockout cell line system for auxotrophy to uridine; suicide switches without transduction may avoid immunogenicity issues (71).

HOW TO AVOID HLA-ALLOANTIGEN SENSITIZATION 2: UNIVERSAL DONOR CELL THERAPY

CIITA is the master transcription factor for class II MHC genes, and it has been reported that ablation of class II MHC can alleviate rejection by CD4 T cells (72). Universal donor cells have been proposed that remove both class I and II MHC, the greatest source of immunogenicity in allogeneic transplantation, and include MSCs (73), iPSCs (74, 75), and CAR-T (66, 76). Wang et al. (74) demonstrated in a monkey xenogeneic model that human universal iPSCs with dual KO of B2M and CIITA can further reduce infiltration of T/B lymphocytes than single KO. Dexamethasone was also able to increase the expression of prostaglandin E-2 (PGE-2), indoleamine-2,3-dioxygenase (IDO), and HLA-G in MSCs, which is interesting because it may contribute to improved function of the cells as well as reduced NK lysis susceptibility (77). However, safeguards such as suicide systems may also be required to control these cells, which are outside the host’s immune system.

DISCUSSION

Because of their immunomodulatory and progenitor cell capabilities, MSCs are attractive therapeutic agents for treating various diseases (Tables 1 and 2). Notably, in human kidney transplantation, autologous MSCs replaced tacrolimus to reduce the adverse effects of CNI, and HLA-matched allogeneic MSCs suppressed rejection at lower concentrations of tacrolimus (Table 2). However, HLA-mismatched cell therapy, used for convenience and standardization, causes rejection owing to the activation of alloantigen-specific T/B cells preexisting in the recipient’s immune system (Tables 3 and 4). Fortunately, there are few reports of recipient rejection of these cell therapies directly causing serious adverse effects (78, 79). However, these results may lead to the repeated use of cell therapy from allogeneic sources without a deeper understanding of the trade-offs between adverse effects and therapeutic benefits. A second repeat transplantation of allogeneic MSCs into horses or monkeys has been associated with the development of adverse clinical effects thought to be due to adaptive immunity compared to controls (35, 44, 47). These results are supported by reports showing that HLA-mismatched allogeneic cells cause an increase in alloreactive memory CD8 T cells, an increase in Ag-specific Abs, decreased survival, and faster immune rejection (44) (Tables 3 and 4). Thus, these results suggest that reducing alloantigen-sensitized T/B cells through an HLA-matching design is the most important factor in improving engraftment and reducing the repeated use of any cell therapy that uses allogeneic cells (Fig. 1) (14, 35, 53).

The removal of class I MHC from the surface of cell therapy is being attempted as another way to avoid rejection due to the use of HLA-mismatched cell therapy. The neoantigen peptides bind to a class-I MHC molecule consisting of an α chain and a β chain of B2M, which is presented on the cell surface and recognized by the TCR of CD8 T cells (Supplementary Fig. 1) (65). Thus, B2MKO cell therapy was able to function while avoiding rejection by alloreactive T cells, compared to wild-type controls (60). However, since the B2MKO system avoids Ag recognition by CD8 T cells, it may be difficult to deal with the development of adverse effects, such as tumorigenicity or infection of genome-edited cells (64, 65). Thus, this problem may be solved by applying a suicide system to prevent the adverse effects caused by the excessive immune response of CAR-T cells (66, 67, 69, 70, 71).

Allogeneic CAR-T cells are the most successful cell therapies approved by the FDA, but they are not free from rejection due to immunogenicity (66). However, universal–CAR-T, –MSCs, and –iPSC cell therapies in which both B2M and CIITA are knocked out have been shown to effectively evade immune rejection (66, 73, 74, 75, 76). Even more encouraging is that a number of universal cell therapies are being evaluated as functionally normal or well differentiated (66, 74, 75, 76, 80). These results show that universal cell therapy has the potential to be an effective treatment that overcomes the disadvantages of autologous or HLA-matched cells. Additionally, these results may provide hope for MSCs, which have failed to achieve efficacy in several clinical trials due to tissue source, donor heterogeneity, and heterogeneous manufacturing (81). However, the control of these cells outside the host’s immune system may require safeguards such as artificial suicide systems. In addition, HLA-KO cell therapies are expected to suppress acute rejection, but immunogenicity to foreign Ags, which is expected to be relatively weak, remains (17, 32, 36, 48).

CONCLUSION

The use of autologous or HLA-matched allogeneic cells in cell therapy may have disadvantages in terms of convenience but may reduce repeated use due to the long-term survival of therapeutic cells. In addition, the use of these cell therapies is expected to increase curative opportunities and success rates in patients who are candidates for allogeneic transplantation (24, 47, 52, 53, 54). These results are attributed to the fact that although HLA-mismatched allogeneic cells such as MSCs and iPSCs have immunomodulatory effects, they are ineffective in suppressing the activity of Ag-specific memory cells in the recipient's immune system (Tables 3 and 4) (29). To prevent the rejection of immunogenic-cell therapy, the introduction of Ag-specific Tregs effectively suppresses the recipient's immune response to Cas9 and alloantigens (36, 82, 83). However, to produce Ag-specific Tregs, there are obstacles, such as high-level technology that has not yet been established, cost, and time consumption (84). Thus, B2MKO- and universal-cell therapies, with proven safety, as well as validated HLA-matched cells, has the potential to be an effective way to reduce immunogenicity. However, the number of times a patient should receive a validated HLA-matched-, B2MKO-, or universal-cell therapy compared with autologous treatment to minimize adverse effects remains to be evaluated. In addition, the use of xenogeneic-free serum benefits the long-term survival of therapeutic cells by reducing unnecessary activity of the recipient’s immune system (8, 35, 51). This suggests that immunogenic cell therapy may have an opportunity cost in terms of the limited number of treatments between adverse effects and therapeutic benefit, so efforts to eliminate the causes of immunogenicity need to be sustained.

SUPPLEMENTARY MATERIAL

Supplementary Figure 1

Recognition pathway of recipient T cells against HLA-alloantigen. (A) A direct pathway is the recognition of HLA-alloantigens presented on the surface of donor cells by recipient T cells. This pathway is primarily associated with acute allograft rejection (S1,S2). (B) In the indirect pathway, HLA-alloantigens from donor cells are recognized by recipient T cells by presentation of recipient APCs. This pathway is involved in chronic allograft rejection (S1,S2). This figure was created using BioRender.com.

Click here to view.(308K, ppt)

Notes

Conflict of Interest:The authors declare no potential conflicts of interest.

Author Contributions:

  • Conceptualization: Chang SH, Park CG.

  • Investigation: Chang SH, Park CG.

  • Writing - original draft: Chang SH, Park CG.

  • Writing - review & editing: Chang SH, Park CG.

Abbreviations

ABMR antibody-mediated rejection
ADSC adipose-derived mesenchymal stem cell
B2MKO β2-microglobulin knockout
BM-MSC bone marrow-MSC
CIITA class II MHC transactivator
CNI calcineurin inhibitors
DC dendritic cell
FCGS feline chronic gingivostomatitis
GE genome edited
GFP green fluorescent protein
IAI intra-articular injection
iEC differentiation of iPSCs into endothelial cell
iPSC induced pluripotent stem cell
IV Intravenous
KTp kidney transplantation
MSC mesenchymal stem cell
OLP oral lichen planus
SIBG standard iliac bone graft
SLE systemic lupus erythematosus
SRG severe refractory gingivostomatitis
UC-MSC umbilical cord-MSC

ACKNOWLEDGEMENTS

This study was supported by the National Research Foundation of Korea (Daejun, Korea) through grants 2020R1A2C1100163 awarded to Sung-Ho Chang.

References

    1. Saler M, Caliogna L, Botta L, Benazzo F, Riva F, Gastaldi G. hASC and DFAT, multipotent stem cells for regenerative medicine: a comparison of their potential differentiation in vitro . Int J Mol Sci 2017;18:2699.
    1. Weiss AR, Dahlke MH. Immunomodulation by mesenchymal stem cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs. Front Immunol 2019;10:1191.
    1. Lynggaard CD, Grønhøj C, Christensen R, Fischer-Nielsen A, Melchiors J, Specht L, Andersen E, Mortensen J, Oturai P, Barfod GH, et al. Intraglandular off-the-shelf allogeneic mesenchymal stem cell treatment in patients with radiation-induced xerostomia: a safety study (MESRIX-II). Stem Cells Transl Med 2022;11:478–489.
    1. Chen CF, Hu CC, Wu CT, Wu HH, Chang CS, Hung YP, Tsai CC, Chang Y. Treatment of knee osteoarthritis with intra-articular injection of allogeneic adipose-derived stem cells (ADSCs) ELIXCYTE®: a phase I/II, randomized, active-control, single-blind, multiple-center clinical trial. Stem Cell Res Ther 2021;12:562.
    1. Meucci MC, Reinders ME, Groeneweg KE, Bezstarosti S, Ajmone Marsan N, Bax JJ, De Fijter JW, Delgado V. Cardiovascular effects of autologous bone marrow-derived mesenchymal stromal cell therapy with early tacrolimus withdrawal in renal transplant recipients: an analysis of the randomized triton study. J Am Heart Assoc 2021;10:e023300
    1. Mo Y, Kim Y, Bang JY, Jung J, Lee CG, Elias JA, Kang HR. Mesenchymal stem cells attenuate asthmatic inflammation and airway remodeling by modulating macrophages/monocytes in the il-13-overexpressing mouse model. Immune Netw 2022;22:e40
    1. Wang Y, Tian M, Wang F, Heng BC, Zhou J, Cai Z, Liu H. Understanding the immunological mechanisms of mesenchymal stem cells in allogeneic transplantation: From the aspect of major histocompatibility complex class i. Stem Cells Dev 2019;28:1141–1150.
    1. Chang SH, Park CG. Allogeneic ADSCs induce CD8 T cell-mediated cytotoxicity and faster cell death after exposure to xenogeneic serum or proinflammatory cytokines. Exp Mol Med 2019;51:1–10.
    1. Mukonoweshuro B, Brown CJ, Fisher J, Ingham E. Immunogenicity of undifferentiated and differentiated allogeneic mouse mesenchymal stem cells. J Tissue Eng 2014;5:2041731414534255
    1. Li C, Chen S, Zhou Y, Zhao Y, Liu P, Cai J. Application of induced pluripotent stem cell transplants: autologous or allogeneic? Life Sci 2018;212:145–149.
    1. Karahan GE, Claas FH, Heidt S. Pre-existing alloreactive t and b cells and their possible relevance for pre-transplant risk estimation in kidney transplant recipients. Front Med (Lausanne) 2020;7:340.
    1. Lefaucheur C, Loupy A. Antibody-mediated rejection of solid-organ allografts. N Engl J Med 2018;379:2580–2582.
    1. Pulecio J, Verma N, Mejía-Ramírez E, Huangfu D, Raya A. Crispr/cas9-based engineering of the epigenome. Cell Stem Cell 2017;21:431–447.
    1. Gorecka J, Kostiuk V, Fereydooni A, Gonzalez L, Luo J, Dash B, Isaji T, Ono S, Liu S, Lee SR, et al. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res Ther 2019;10:87.
    1. Pei WD, Zhang Y, Yin TL, Yu Y. Epigenome editing by CRISPR/Cas9 in clinical settings: possibilities and challenges. Brief Funct Genomics 2020;19:215–228.
    1. Scheiner ZS, Talib S, Feigal EG. The potential for immunogenicity of autologous induced pluripotent stem cell-derived therapies. J Biol Chem 2014;289:4571–4577.
    1. Morizane A, Kikuchi T, Hayashi T, Mizuma H, Takara S, Doi H, Mawatari A, Glasser MF, Shiina T, Ishigaki H, et al. MHC matching improves engraftment of iPSC-derived neurons in non-human primates. Nat Commun 2017;8:385.
    1. Embgenbroich M, Burgdorf S. Current concepts of antigen cross-presentation. Front Immunol 2018;9:1643.
    1. Arzi B, Clark KC, Sundaram A, Spriet M, Verstraete FJ, Walker NJ, Loscar MR, Fazel N, Murphy WJ, Vapniarsky N, et al. Therapeutic efficacy of fresh, allogeneic mesenchymal stem cells for severe refractory feline chronic gingivostomatitis. Stem Cells Transl Med 2017;6:1710–1722.
    1. Arzi B, Mills-Ko E, Verstraete FJ, Kol A, Walker NJ, Badgley MR, Fazel N, Murphy WJ, Vapniarsky N, Borjesson DL. Therapeutic efficacy of fresh, autologous mesenchymal stem cells for severe refractory gingivostomatitis in cats. Stem Cells Transl Med 2016;5:75–86.
    1. Kamen DL, Wallace C, Li Z, Wyatt M, Paulos C, Wei C, Wang H, Wolf BJ, Nietert PJ, Gilkeson G. Safety, immunological effects and clinical response in a phase I trial of umbilical cord mesenchymal stromal cells in patients with treatment refractory SLE. Lupus Sci Med 2022;9:e000704
    1. Jolly M, Garris CP, Mikolaitis RA, Jhingran PM, Dennis G, Wallace DJ, Clarke A, Dooley MA, Parke A, Strand V, et al. Development and validation of the Lupus Impact Tracker: a patient-completed tool for clinical practice to assess and monitor the impact of systemic lupus erythematosus. Arthritis Care Res (Hoboken) 2014;66:1542–1550.
    1. Hernigou P, Guissou I, Homma Y, Poignard A, Chevallier N, Rouard H, Flouzat Lachaniette CH. Percutaneous injection of bone marrow mesenchymal stem cells for ankle non-unions decreases complications in patients with diabetes. Int Orthop 2015;39:1639–1643.
    1. Oliveira RL, Chagastelles PC, Sesterheim P, Pranke P. In vivo immunogenic response to allogeneic mesenchymal stem cells and the role of preactivated mesenchymal stem cells cotransplanted with allogeneic islets. Stem Cells Int 2017;2017:9824698
    1. Coemans M, Süsal C, Döhler B, Anglicheau D, Giral M, Bestard O, Legendre C, Emonds MP, Kuypers D, Molenberghs G, et al. Analyses of the short- and long-term graft survival after kidney transplantation in Europe between 1986 and 2015. Kidney Int 2018;94:964–973.
    1. Pan GH, Chen Z, Xu L, Zhu JH, Xiang P, Ma JJ, Peng YW, Li GH, Chen XY, Fang JL, et al. Low-dose tacrolimus combined with donor-derived mesenchymal stem cells after renal transplantation: a prospective, non-randomized study. Oncotarget 2016;7:12089–12101.
    1. Dreyer GJ, Groeneweg KE, Heidt S, Roelen DL, van Pel M, Roelofs H, Huurman VA, Bajema IM, Moes DJ, Fibbe WE, et al. Human leukocyte antigen selected allogeneic mesenchymal stromal cell therapy in renal transplantation: the Neptune study, a phase I single-center study. Am J Transplant 2020;20:2905–2915.
    1. Reinders ME, Groeneweg KE, Hendriks SH, Bank JR, Dreyer GJ, de Vries AP, van Pel M, Roelofs H, Huurman VA, Meij P, et al. Autologous bone marrow-derived mesenchymal stromal cell therapy with early tacrolimus withdrawal: the randomized prospective, single-center, open-label TRITON study. Am J Transplant 2021;21:3055–3065.
    1. Večerić-Haler Ž, Sever M, Kojc N, Halloran PF, Boštjančič E, Mlinšek G, Oblak M, Poženel P, Švajger U, Hartman K, et al. Autologous mesenchymal stem cells for treatment of chronic active antibody-mediated kidney graft rejection: report of the phase i/ii clinical trial case series. Transpl Int 2022;35:10772.
    1. Kol A, Wood JA, Carrade Holt DD, Gillette JA, Bohannon-Worsley LK, Puchalski SM, Walker NJ, Clark KC, Watson JL, Borjesson DL. Multiple intravenous injections of allogeneic equine mesenchymal stem cells do not induce a systemic inflammatory response but do alter lymphocyte subsets in healthy horses. Stem Cell Res Ther 2015;6:73.
    1. Owens SD, Kol A, Walker NJ, Borjesson DL. Allogeneic mesenchymal stem cell treatment induces specific alloantibodies in horses. Stem Cells Int 2016;2016:5830103
    1. Kawamura T, Miyagawa S, Fukushima S, Maeda A, Kashiyama N, Kawamura A, Miki K, Okita K, Yoshida Y, Shiina T, et al. Cardiomyocytes derived from MHC-homozygous induced pluripotent stem cells exhibit reduced allogeneic immunogenicity in MHC-matched non-human primates. Stem Cell Reports 2016;6:312–320.
    1. Nakagawa M, Taniguchi Y, Senda S, Takizawa N, Ichisaka T, Asano K, Morizane A, Doi D, Takahashi J, Nishizawa M, et al. A novel efficient feeder-free culture system for the derivation of human induced pluripotent stem cells. Sci Rep 2014;4:3594.
    1. Chang SH, Kim HJ, Park CG. Allogeneic ADSCs induce the production of alloreactive memory-CD8 T cells through HLA-ABC antigens. Cells 2020;9:1246.
    1. Joswig AJ, Mitchell A, Cummings KJ, Levine GJ, Gregory CA, Smith R 3rd, Watts AE. Repeated intra-articular injection of allogeneic mesenchymal stem cells causes an adverse response compared to autologous cells in the equine model. Stem Cell Res Ther 2017;8:42.
    1. Wagner DL, Amini L, Wendering DJ, Burkhardt LM, Akyüz L, Reinke P, Volk HD, Schmueck-Henneresse M. High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population. Nat Med 2019;25:242–248.
    1. Arruda VR, Favaro P, Finn JD. Strategies to modulate immune responses: a new frontier for gene therapy. Mol Ther 2009;17:1492–1503.
    1. Deuse T, Hu X, Agbor-Enoh S, Koch M, Spitzer MH, Gravina A, Alawi M, Marishta A, Peters B, Kosaloglu-Yalcin Z, et al. De novo mutations in mitochondrial DNA of iPSCs produce immunogenic neoepitopes in mice and humans. Nat Biotechnol 2019;37:1137–1144.
    1. He Y, Wu J, Dressman DC, Iacobuzio-Donahue C, Markowitz SD, Velculescu VE, Diaz LA Jr, Kinzler KW, Vogelstein B, Papadopoulos N. Heteroplasmic mitochondrial DNA mutations in normal and tumour cells. Nature 2010;464:610–614.
    1. Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci U S A 1997;94:514–519.
    1. Badr Eslam R, Croce K, Mangione FM, Musmann R, Leopold JA, Mitchell RN, Waxman AB. Persistence and proliferation of human mesenchymal stromal cells in the right ventricular myocardium after intracoronary injection in a large animal model of pulmonary hypertension. Cytotherapy 2017;19:668–679.
    1. Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, et al. Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA 2012;308:2369–2379.
    1. Pigott JH, Ishihara A, Wellman ML, Russell DS, Bertone AL. Investigation of the immune response to autologous, allogeneic, and xenogeneic mesenchymal stem cells after intra-articular injection in horses. Vet Immunol Immunopathol 2013;156:99–106.
    1. Isakova IA, Lanclos C, Bruhn J, Kuroda MJ, Baker KC, Krishnappa V, Phinney DG. Allo-reactivity of mesenchymal stem cells in rhesus macaques is dose and haplotype dependent and limits durable cell engraftment in vivo . PLoS One 2014;9:e87238
    1. Colbath AC, Dow SW, Hopkins LS, Phillips JN, McIlwraith CW, Goodrich LR. Allogeneic vs. autologous intra-articular mesenchymal stem cell injection within normal horses: clinical and cytological comparisons suggest safety. Equine Vet J 2020;52:144–151.
    1. Hwang JW, Lee NK, Yang JH, Son HJ, Bang SI, Chang JW, Na DL. A comparison of immune responses exerted following syngeneic, allogeneic, and xenogeneic transplantation of mesenchymal stem cells into the mouse brain. Int J Mol Sci 2020;21:3052.
    1. Rowland AL, Miller D, Berglund A, Schnabel LV, Levine GJ, Antczak DF, Watts AE. Cross-matching of allogeneic mesenchymal stromal cells eliminates recipient immune targeting. Stem Cells Transl Med 2021;10:694–710.
    1. Hoornaert CJ, Le Blon D, Quarta A, Daans J, Goossens H, Berneman Z, Ponsaerts P. Concise review: innate and adaptive immune recognition of allogeneic and xenogeneic cell transplants in the central nervous system. Stem Cells Transl Med 2017;6:1434–1441.
    1. Merlot AM, Kalinowski DS, Richardson DR. Unraveling the mysteries of serum albumin-more than just a serum protein. Front Physiol 2014;5:299.
    1. Tiruppathi C, Finnegan A, Malik AB. Isolation and characterization of a cell surface albumin-binding protein from vascular endothelial cells. Proc Natl Acad Sci U S A 1996;93:250–254.
    1. Haque N, Kasim NH, Rahman MT. Optimization of pre-transplantation conditions to enhance the efficacy of mesenchymal stem cells. Int J Biol Sci 2015;11:324–334.
    1. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol 2014;32:252–260.
    1. Stoddard-Bennett T, Pera RR. Stem cell therapy for Parkinson’s disease: safety and modeling. Neural Regen Res 2020;15:36–40.
    1. Verboket R, Leiblein M, Seebach C, Nau C, Janko M, Bellen M, Bönig H, Henrich D, Marzi I. Autologous cell-based therapy for treatment of large bone defects: from bench to bedside. Eur J Trauma Emerg Surg 2018;44:649–665.
    1. Taylor CJ, Peacock S, Chaudhry AN, Bradley JA, Bolton EM. Generating an iPSC bank for HLA-matched tissue transplantation based on known donor and recipient HLA types. Cell Stem Cell 2012;11:147–152.
    1. Lu Y, Xue J, Deng T, Zhou X, Yu K, Deng L, Huang M, Yi X, Liang M, Wang Y, et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat Med 2020;26:732–740.
    1. Charlesworth CT, Deshpande PS, Dever DP, Camarena J, Lemgart VT, Cromer MK, Vakulskas CA, Collingwood MA, Zhang L, Bode NM, et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat Med 2019;25:249–254.
    1. Xian B, Huang B. The immune response of stem cells in subretinal transplantation. Stem Cell Res Ther 2015;6:161.
    1. Dudek AM, Porteus MH. Answered and unanswered questions in early-stage viral vector transduction biology and innate primary cell toxicity for ex-vivo gene editing. Front Immunol 2021;12:660302
    1. Zha S, Tay JC, Zhu S, Li Z, Du Z, Wang S. Generation of mesenchymal stromal cells with low immunogenicity from human PBMC-derived beta2 microglobulin knockout induced pluripotent stem cells. Cell Transplant 2020;29:963689720965529
    1. Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer 2016;16:7–19.
    1. Shin MH, Kim J, Lim SA, Kim J, Kim SJ, Lee KM. NK cell-based immunotherapies in cancer. Immune Netw 2020;20:e14
    1. Gornalusse GG, Hirata RK, Funk SE, Riolobos L, Lopes VS, Manske G, Prunkard D, Colunga AG, Hanafi LA, Clegg DO, et al. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat Biotechnol 2017;35:765–772.
    1. Wang C, Wang Z, Yao T, Zhou J, Wang Z. The immune-related role of beta-2-microglobulin in melanoma. Front Oncol 2022;12:944722
    1. Zhao Y, Cao Y, Chen Y, Wu L, Hang H, Jiang C, Zhou X. B2M gene expression shapes the immune landscape of lung adenocarcinoma and determines the response to immunotherapy. Immunology 2021;164:507–523.
    1. Wagner DL, Fritsche E, Pulsipher MA, Ahmed N, Hamieh M, Hegde M, Ruella M, Savoldo B, Shah NN, Turtle CJ, et al. Immunogenicity of CAR T cells in cancer therapy. Nat Rev Clin Oncol 2021;18:379–393.
    1. Jensen MC, Popplewell L, Cooper LJ, DiGiusto D, Kalos M, Ostberg JR, Forman SJ. Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant 2010;16:1245–1256.
    1. Berger C, Flowers ME, Warren EH, Riddell SR. Analysis of transgene-specific immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified donor T cells after allogeneic hematopoietic cell transplantation. Blood 2006;107:2294–2302.
    1. Zhou X, Dotti G, Krance RA, Martinez CA, Naik S, Kamble RT, Durett AG, Dakhova O, Savoldo B, Di Stasi A, et al. Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation. Blood 2015;125:4103–4113.
    1. Di Stasi A, Tey SK, Dotti G, Fujita Y, Kennedy-Nasser A, Martinez C, Straathof K, Liu E, Durett AG, Grilley B, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med 2011;365:1673–1683.
    1. Wiebking V, Patterson JO, Martin R, Chanda MK, Lee CM, Srifa W, Bao G, Porteus MH. Metabolic engineering generates a transgene-free safety switch for cell therapy. Nat Biotechnol 2020;38:1441–1450.
    1. Kagoya Y, Guo T, Yeung B, Saso K, Anczurowski M, Wang CH, Murata K, Sugata K, Saijo H, Matsunaga Y, et al. Genetic ablation of HLA class I, class II, and the T-cell receptor enables allogeneic T cells to be used for adoptive T-cell therapy. Cancer Immunol Res 2020;8:926–936.
    1. Halm D, Leibig N, Martens J, Stark GB, Groß T, Zimmermann S, Finkenzeller G, Lampert F. Direct comparison of the immunogenicity of major histocompatibility complex-I and -II deficient mesenchymal stem cells in vivo . Biol Chem 2021;402:693–702.
    1. Wang X, Lu M, Tian X, Ren Y, Li Y, Xiang M, Chen S. Diminished expression of major histocompatibility complex facilitates the use of human induced pluripotent stem cells in monkey. Stem Cell Res Ther 2020;11:334.
    1. Mattapally S, Pawlik KM, Fast VG, Zumaquero E, Lund FE, Randall TD, Townes TM, Zhang J. Human leukocyte antigen class I and II knockout human induced pluripotent stem cell-derived cells: universal donor for cell therapy. J Am Heart Assoc 2018;7:e010239
    1. Lee J, Sheen JH, Lim O, Lee Y, Ryu J, Shin D, Kim YY, Kim M. Abrogation of HLA surface expression using CRISPR/Cas9 genome editing: a step toward universal T cell therapy. Sci Rep 2020;10:17753.
    1. Rawat S, Dadhwal V, Mohanty S. Dexamethasone priming enhances stemness and immunomodulatory property of tissue-specific human mesenchymal stem cells. BMC Dev Biol 2021;21:16.
    1. Kot M, Baj-Krzyworzeka M, Szatanek R, Musiał-Wysocka A, Suda-Szczurek M, Majka M. The importance of HLA assessment in “off-the-shelf” allogeneic mesenchymal stem cells based-therapies. Int J Mol Sci 2019;20:5680.
    1. Shah K, Shah N, Ghassemi F, Ly C, George T, Lutz C, Sumer H. Alloreactivity of allogeneic mesenchymal stem/stromal cells and other cellular therapies: a concise review. Stem Cells Int 2022;2022:9589600
    1. Mo F, Watanabe N, McKenna MK, Hicks MJ, Srinivasan M, Gomes-Silva D, Atilla E, Smith T, Ataca Atilla P, Ma R, et al. Engineered off-the-shelf therapeutic T cells resist host immune rejection. Nat Biotechnol 2021;39:56–63.
    1. Srinivasan A, Sathiyanathan P, Yin L, Liu TM, Lam A, Ravikumar M, Smith RA, Loh HP, Zhang Y, Ling L, et al. Strategies to enhance immunomodulatory properties and reduce heterogeneity in mesenchymal stromal cells during ex vivo expansion. Cytotherapy 2022;24:456–472.
    1. Juneja T, Kazmi M, Mellace M, Saidi RF. Utilization of Treg cells in solid organ transplantation. Front Immunol 2022;13:746889
    1. Proics E, David M, Mojibian M, Speck M, Lounnas-Mourey N, Govehovitch A, Baghdadi W, Desnouveaux J, Bastian H, Freschi L, et al. Preclinical assessment of antigen-specific chimeric antigen receptor regulatory T cells for use in solid organ transplantation. Gene Ther 2023;30:309–322.
    1. Atif M, Conti F, Gorochov G, Oo YH, Miyara M. Regulatory T cells in solid organ transplantation. Clin Transl Immunology 2020;9:e01099

Metrics
Share
Figures

1 / 1

Tables

1 / 6

Funding Information
PERMALINK