Mesenchymal stromal cells (MSCs) are multipotent homeostatic regulators of the body that have both multilineage differentiation potential and self-renewal capabilities [1]. These ’stem-like’ cells are readily available and exist in the stroma of the bone marrow, adipose tissue, the synovium of the joints, dental pulps, the umbilical cord and the liver among other tissues [1,2]. MSCs are immune-evasive allowing for off-the-shelf use. Given MSC availability, their low immunogenicity, significant immunomodulatory and regenerative capabilities, MSCs are considered an attractive option for conversion to cellular therapies for treatment of immune disease [1,2].
Despite the promising potential of MSCs for the treatment of immune disease ranging from graft-vs.-host diseases (GVHD) to Crohn’s and amyotrophic lateral sclerosis, there have been no resultant US FDA approvals to date. However, MSCs have been approved for the treatment of steroid refractory acute GVHD and implications of colitis in other countries including Japan, Canada, Europe and New Zealand [2,3]. These approved MSC therapy products span from autologous to allogeneic sources and are derived from the bone marrow, adipose and umbilical cord sources. Among the clinical products furthest along in the path to FDA acceptance are an allogeneic bone marrow derived MSC, called Mesoblast (remestemcel-L) for use in pediatric steroid refractory GVHD. Most recently, Mesoblast treatment led to a 74.1% overall survival rate at day 100 in a phase 3, prospective, single-arm, multicenter study (NCT02336230) treating 54 children with primary steroid refractory GVHD [4].
Nonetheless, full approval in the USA has mostly been thwarted by lack of consistent MSC therapeutic efficacy across clinical trials [3,5,6]. MSC failures are characterized by low survival, engraftment, homing to damaged tissues as well as insufficient immunosuppression at inflammatory sites following their ex vivo culture [2,3,5]. Additionally, MSCs have high heterogeneity from donor to donor, as well as from different tissue and isolation sources, which further complicates the consistency of their therapeutic efficacy [6]. These underwhelming findings and untapped MSC therapeutic potential call forth the need to increase efforts in MSC cellular engineering to enhance therapeutic outcomes in the clinic.
To directly improve the shortcomings associated with MSC therapeutic efficacy, various researchers have employed genetic and nongenetic engineering strategies to enhance MSC function. Cellular engineering has been used to help overexpress beneficial proteins for MSC therapeutic function including the promotion of MSC survival, pro-apoptotic proteins, enhanced migration to target sites and enhanced immunosuppressive function [3,7,8].
1. Nongenetic MSC engineering
Among the most prevalent nongenetic engineering strategy used to improve MSC function is MSC priming. Priming approaches have been employed with cytokines, growth factors, alternative culture conditions, hypoxia, herbal components and biomaterials such as pharmacologic drugs, small molecules, among other factors [9,10].
1.1. Cytokine priming
Preconditioning MSCs with IFNy, among other inflammatory cytokines, leads to an upregulation of immunosuppressive IDO1 gene, priming the MSC to secrete immunomodulatory molecules PGE2, HGF, TGF-β and CCL2 as a feedback mechanism [9,11].
1.2. Culture conditioning
MSCs have also shown improvements based on their culture conditions. It has been reported that 3D culturing techniques of MSCs more closely mimics the in vivo state of MSCs as compared with 2D culture. As such, the development of 3D MSC spheroids has led to improved immunosuppressive outcomes in murine peritonitis, colitis and rheumatoid arthritis [12].
1.3. Hypoxic priming
Another strategy to improve MSC proliferative capabilities is through culturing MSCs in hypoxic environments [9]. MSCs adapt to the reduced oxygen in hypoxia by increasing their proliferative capabilities, reducing their rates of apoptosis and increasing HIF1a-associated expression of chemokines involved in MSC trafficking [9,13].
Altogether, these strategies are used in attempt to mimic the inflammatory environment and other cell stressors that MSCs react to in vivo, but in an ex vivo cell culture environment [12,14].
2. Genetic engineering
Advances in genetic engineering tools have allowed various transient and stable protein delivery to improve MSC therapy:
2.1. MSC viability
One notable limitation observed in some instances of MSC therapy is low viability following MSC transplantation into hosts. Researchers have transiently overexpressed growth factors in MSCs to improve this viability. For example, FGF-2 gene has been overexpressed in MSCs ex vivo to improve cardiac tissue repair. This MSC genetic modification results in a threefold increase in cell viability and cardiac marker expression for greater therapeutic outcomes in myocardial infarction repair [7,15]. Integrin-linked kinase has also been overexpressed in MSCs to increase their survival for myocardial infarction repair leading to improved angiogenesis at week three post-MSC transplantation [7,16].
2.2. MSC homing
Another major therapeutic limitation addressed through MSC cellular engineering is the limited homing and migration of MSC to target regions following administration [3,5]. The induction of CXCR4 and CXCR7 has been applied for enhanced MSC migration. CXCR4 and CXCR7 chemokine receptors are known to play a pivotal role in canonical MSC migration and adhesion capacity. As such, these studies reported that the overexpression of CXCR4/CXCR7 in adipose derived MSCs improved their paracrine, proliferative and migratory abilities yielding a powerful therapeutic improvement to MSC treatment [8,17].
2.3. MSC immunosuppression
Cellular engineering has also been applied to directly improve MSC immunosuppressive function to better treat inflammatory disease. This has been done through the stable lentiviral overexpression of potent anti-inflammatory cytokine IL10 in combination with CXCR4 creating CXCR4-IL10-MSCs. These cells not only induced expression of functional CXCR4 receptor on the MSC cell surface, but also allowed for the increased secretion of IL10 [17,18]. In preclinical GVHD mouse models, CXCR4-IL10-MSCs displayed increased therapeutic capacity and marked reduction in the number of pro-inflammatory Th1 and Th17 CD3+ cells with polarization toward anti-inflammatory IL10+ CD3+ T-cells [18]. Engineered MSCs have also been created to overexpress both inhibitory marker PDL1 as well as pro survival kinase AKT (adMSC-PDL1-AKT) simultaneously [19,20]. This dual engineering not only allowed for enhanced proliferation rates and resistance to oxidative stress by the MSCs through AKT, but also led to a robust upregulation of immunosuppressive regulatory T-cell phenotypes through increased interaction with PDL1 on the MSCs to PD1 on T-cells [19]. These modifications further highlight the functional and therapeutic potential of genetically engineered MSCs.
3. Synthetic biology
Most recently, our group has applied synthetic biology to MSCs creating chimeric antigen receptor MSCs (CAR-MSCs). A CAR is a synthetic protein composed of an extracellular antigen binding domain fused to a functional intracellular signaling domain. CAR was first applied with resounding success to the field of hematological malignancies through the creation and eventual FDA approval of CART-cell therapy [21,22]. CART became the most successful cellular therapy to date by taking known functions of immunogenic T-cells and enhancing them to target B-cell malignancies. Specifically, the first FDA approved CART-cells contained CARs designed to recognize CD19 antigen expressed almost exclusively on malignant B-cells and was fused to a intracellular 4-1BB and CD3z signaling domain to mediate activation and killing of target cancer cells by T-cells [21,22].
By creating CAR-MSCs, we equipped the MSCs with a gain of more than one function including enhanced 1) homing to target tissues due to a tissue-specific antigen binding domain and 2) immunosuppression due to inclusion of an immunosuppressive intracellular signaling domain [23].
The bioengineering of CAR-MSCs was verified for safety and maintenance of stemness as well as high lentiviral transduction efficiency of CAR at >80% in human adipose-derived MSCs. As a proof-of-concept model, CAR-MSCs were engineered to target E-cadherin (Ecad) for the treatment of GVHD. CAR-MSCs displayed Ecad antigen-specific immunosuppression of T-cells in vitro and in vivo GVHD Xenograft Models. Compared with standard nontransduced MSC therapy, CAR-MSC treatment led to a significant prevention of mouse weight loss, decreased clinical symptom scores, increased human T-cell suppression, differentiation of T-cells toward a Treg-like phenotype and improved overall survival of mice [23]. A distinct upregulation of immunosuppressive gene signatures was found in Ecad stimulated CAR-MSCs, further supporting functionality of the CAR receptor. Finally, anti-Ecad CAR-MSCs displayed preferential homing to mouse Ecad+ colonic tissue and greater efficacy as compared to nonspecific control scFv CAR-MSCssupporting the importance of CAR scFv design [23].
The stability and increased functionality of MSCs engineered to overexpress genes and synthetic CAR receptors represents a promising step for cell therapy technologies used to treat immune disease. They directly address the shortcomings of previous MSC clinical trials and display promising superiority to the current standard of care. The possibilities here are endless where MSC functionality can be mediated by intracellular signaling domains or inclusion of immunosuppression cytokines [18], while MSC homing and migration can be mediated by increased expression of chemokine receptors or incorporation of a target tissue specific antigen-binding domain [8,17,23].
Engineered MSCs have only recently entered early phase clinical trials. Although terminated early due to insufficient trial participants, autologous MSCs retrovirally expressing tumor-specific herpes simplex virus thymidine kinase were tested in a phase I/II clinical trial (EuDRA CT Numberi: 2012-003741-15) for patients with gastrointestinal tumors and reported a median overall survival of 15.6 months [24,25]. A second completed phase I/II clinical trial for patients with relapsed solid tumors utilized autologous BM-MSCs engineered to carry oncolytic adenovirus called Celyvir. Celyvir showed minimal clinical efficacy with 83% of patients demonstrating progressive disease following treatment. However, the engineered Celyvir product demonstrated robust safety and tolerability in patients guiding future opportunities for selecting off-the-shelf MSCs from healthy donors with clinically favorable characteristics in future trials [25,26].
These foundations and early clinical findings in MSC cellular engineering open the door for the creation of novel therapeutic platforms that can be designed and expanded to treat a wide range of immune diseases. The successful transduction of CAR-MSCs with evidence of improved antigen-specific immunosuppression serves as a proof-of-concept study to be implemented in other disease-specific contexts. For example, cytokine secreting or differentiation-promoting intracellular signaling domains could be incorporated into CAR-MSCs to initiate an anti-inflammatory milieu and encourage tissue regeneration. Additionally, these functionalities could be directed to a wide range of tissues including the heart, lungs and liver based on CAR-MSC antigen-binding domain or chemokine design ultimately broadening the therapeutic scope of engineered MSCs.
Acknowledgments
This study was partly funded through NSF-GRFP 2021321972 (O Sirpilla), Mayo Clinic Center for Individualized Medicine (SS Kenderian), Mayo Clinic President’s Strategic Initiative Funds (SS Kenderian), Mayo Clinic Center for Regenerative Biotherapeutics (SS Kenderian), National Institutes of Health grants R01AI179974 (O Sirpilla and SS Kenderian), K12CA090628 (SS Kenderian) and R37CA266344-01 (SS Kenderian), Department of Defense grant CA201127 (SS Kenderian), Predolin Foundation (O Sirpilla and SS Kenderian) and the generosity of Donald Porteous (SS Kenderian).
Funding Statement
This study was supported by the NIHMS grant R37CA266344, R01AI179974.
Author contributions
O Sirpilla and SS Kenderian wrote, edited and approved the final version of the manuscript.
Financial disclosure
This study was supported by the NIHMS grant R37CA266344, R01AI179974. The authors have no financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
Competing interests disclosure
SS Kenderian is an inventor on patents in the field of CAR immunotherapy that are licensed to Novartis (through an agreement between Mayo Clinic, University of Pennsylvania and Novartis), Humanigen (through Mayo Clinic), Mettaforge (through Mayo Clinic). SS Kenderian receives research funding from Kite, Gilead, Juno, BMS, Novartis, Humanigen, MorphoSys, Tolero, Sunesis/Viracta, LifEngine Animal Health Laboratories Inc and Lentigen. SS Kenderian has participated in advisory meetings with Kite/Gilead, Humanigen, Juno/BMS, Capstan Bio and Novartis. SS Kenderian has served on the data safety and monitoring board with Humanigen and Carisma. SS Kenderian has severed a consultant for Torque, Calibr, Novartis, Capstan Bio, BMS, Kite/Gilead and Humanigen. O Sirpilla and SS Kenderian have intellectual property in the CAR-MSC technology.
Writing disclosure
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as: • of interest; •• of considerable interest
- 1.Ma S, Xie N, Li W, et al. Immunobiology of mesenchymal stem cells. Cell Death Differ. 2014;21(2):216–225. doi: 10.1038/cdd.2013.158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Krampera M, Le Blanc K. Mesenchymal stromal cells: putative microenvironmental modulators become cell therapy. Cell Stem Cell. 2021;28(10):1708–1725. doi: 10.1016/j.stem.2021.09.006 [DOI] [PubMed] [Google Scholar]
- 3.Levy O, Kuai R, Siren EMJ, et al. Shattering barriers toward clinically meaningful MSC therapies. Sci Adv. 2020;6(30):eaba6884. doi: 10.1126/sciadv.aba6884 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kurtzberg J, Abdel-Azim H, Carpenter P, et al. A phase 3, single-arm, prospective study of remestemcel-L, ex vivo culture-expanded adult human mesenchymal stromal cells for the treatment of pediatric patients who failed to respond to steroid treatment for acute graft-versus-host disease. Biol Blood Marrow Transplant. 2020;26(5):845–854. doi: 10.1016/j.bbmt.2020.01.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Galipeau J, Sensebe L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22(6):824–833. doi: 10.1016/j.stem.2018.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Robb KP, Galipeau J, Shi Y, et al. Failure to launch commercially-approved mesenchymal stromal cell therapies: what’s the path forward? Proceedings of the International Society for Cell & Gene Therapy (ISCT) Annual Meeting Roundtable held in May 2023, Palais des Congres de Paris, Organized by the ISCT MSC Scientific Committee. Cytotherapy. 2024;26(5):413–417. doi: 10.1016/j.jcyt.2023.09.001 [DOI] [PubMed] [Google Scholar]
- 7.Hodgkinson CP, Gomez JA, Mirotsou M, et al. Genetic engineering of mesenchymal stem cells and its application in human disease therapy. Hum Gene Ther. 2010;21(11):1513–1526. doi: 10.1089/hum.2010.165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ocansey DKW, Pei B, Yan Y, et al. Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med. 2020;18(1):42. doi: 10.1186/s12967-020-02234-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Noronha NC, Mizukami A, Caliari-Oliveira C, et al. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther. 2019;10(1):131. doi: 10.1186/s13287-019-1224-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Afkhami H, Mahmoudvand G, Fakouri A, et al. New insights in application of mesenchymal stem cells therapy in tumor microenvironment: pros and cons. Front Cell Dev Biol. 2023;11:1255697. doi: 10.3389/fcell.2023.1255697 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kim DS, Jang IK, Lee MW, et al. Enhanced immunosuppressive properties of human mesenchymal stem cells primed by interferon-gamma. EBioMedicine. 2018;28:261–273. doi: 10.1016/j.ebiom.2018.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yen BL, Hsieh CC, Hsu PJ, et al. Three-dimensional spheroid culture of human mesenchymal stem cells: offering therapeutic advantages and in vitro glimpses of the in vivo state. Stem Cells Transl Med. 2023;12(5):235–244. doi: 10.1093/stcltm/szad011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kim Y, Jin HJ, Heo J, et al. Small hypoxia-primed mesenchymal stem cells attenuate graft-versus-host disease. Leukemia. 2018;32(12):2672–2684. doi: 10.1038/s41375-018-0151-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Saparov A, Ogay V, Nurgozhin T, et al. Preconditioning of human mesenchymal stem cells to enhance their regulation of the immune response. Stem Cells Int. 2016;2016:3924858. doi: 10.1155/2016/3924858 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Song H, Kwon K, Lim S, et al. Transfection of mesenchymal stem cells with the FGF-2 gene improves their survival under hypoxic conditions. Mol Cells. 2005;19(3):402–407. doi: 10.1016/S1016-8478(23)13186-4 [DOI] [PubMed] [Google Scholar]
- 16.Zeng B, Liu L, Wang S, et al. ILK regulates MSCs survival and angiogenesis partially through AKT and mTOR signaling pathways. Acta Histochem. 2017;119(4):400–406. doi: 10.1016/j.acthis.2017.04.003 [DOI] [PubMed] [Google Scholar]
- 17.Lu F, Liu L, Yu DH, et al. Therapeutic effect of Rhizoma Dioscoreae Nipponicae on gouty arthritis based on the SDF-1/CXCR 4 and p38 MAPK pathway: an in vivo and in vitro study. Phytother Res. 2014;28(2):280–288. doi: 10.1002/ptr.4997 [DOI] [PubMed] [Google Scholar]
- 18.Hervas-Salcedo R, Fernandez-Garcia M, Hernando-Rodriguez M, et al. Improved efficacy of mesenchymal stromal cells stably expressing CXCR4 and IL-10 in a xenogeneic graft versus host disease mouse model. Front Immunol. 2023;14:1062086. doi: 10.1016/S1465-3249(23)00217-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This article demonstrates a new and emerging engineered mesenchymal stromal cell (MSC) product overexpressing CXCR4 and IL10 aiding MSCs with two canonically cited mechanisms of improved therapeutic efficacy featured in this editorial.
- 19.Lin YK, Hsiao LC, Wu MY, et al. PD-L1 and AKT overexpressing adipose-derived mesenchymal stem cells enhance myocardial protection by upregulating CD25(+) T cells in acute myocardial infarction rat model. Int J Mol Sci. 2023;25(1):134. doi: 10.3390/ijms25010134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Zhou L, Liu S, Wang Z, et al. Bone marrow-derived mesenchymal stem cells modified with Akt1 ameliorates acute liver GVHD. Biol Proced Online. 2019;21:24. doi: 10.1186/s12575-019-0112-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.June CH, O'Connor RS, Kawalekar OU, et al. CAR T-cell immunotherapy for human cancer. Science. 2018;359(6382):1361–1365. doi: 10.1126/science.aar6711 [DOI] [PubMed] [Google Scholar]
- 22.Sakemura R, Cox MJ, Hefazi M, et al. Resistance to CART-cell therapy: lessons learned from the treatment of hematological malignancies. Leuk Lymphoma. 2021;62(9):2052–2063. doi: 10.1080/10428194.2021.1894648 [DOI] [PubMed] [Google Scholar]
- 23.Sirpilla O, Sakemura RL, Hefazi M, et al. Mesenchymal stromal cells with chimaeric antigen receptors for enhanced immunosuppression. Nat Biomed Eng. 2024;8(4):443–460. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This article focuses on one of the newest technologies in engineered cellular therapies by applying chimeric antigen receptors to MSCs and highlights a dual positive functional outcome highlighted in this editorial.
- 24.Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol. Sci. 2020;41(9):653–664. doi: 10.1016/j.tips.2020.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhou T, Yuan Z, Weng J, et al. Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol. 2021;14(1):24. doi: 10.1186/s13045-021-01037-x [DOI] [PMC free article] [PubMed] [Google Scholar]; • This review article provides a comprehensive up to date overview of past and present MSC clinical trials, including nonengineered MSCs from a wide variety of tissue sources and culture techniques as well as emerging engineered MSC clinical trials.
- 26.Ruano D, Lopez-Martin JA, Moreno L, et al. First-in-human, first-in-child trial of autologous MSCs carrying the oncolytic virus icovir-5 in patients with advanced tumors. Mol Ther. 2020;28(4):1033–1042. doi: 10.1016/j.ymthe.2020.01.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
