Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 May 15.
Published in final edited form as: J Immunol. 2022 Nov 15;209(10):1811–1816. doi: 10.4049/jimmunol.2200542

Therapeutic Opportunities for Immunoreceptor-Engineered T cell Therapy for Modulation of Alloimmunity

Kavitha Muralidharan 1, Divyansh Agarwal 2, Ali Naji 3, Vijay G Bhoj 1,
PMCID: PMC9680015  NIHMSID: NIHMS1835663  PMID: 36344114

Abstract

Achieving immunosuppression-free immune tolerance to an allograft is one of the central goals of transplantation. Here, we review recent developments in the fields of T cell-based therapies and T cell engineering using chimeric antigen receptors (CARs), and their potential for effective and targeted immune modulation of T and B cell activity in an effort to eliminate pre-existing alloantibodies (desensitization) and achieve long-term tolerance. Approaches that span preclinical to early clinical studies in transplantation will be reviewed, with specific emphasis on advances in T cell immunotherapy that have shown promise. Lastly, we conclude with a forward-looking discussion of how T cell-based therapies in other fields of medicine can be potentially applied to solid organ transplantation.

Introduction

Solid organ transplantation is the standard of care and definitive treatment option for end stage diseases of various organs, including kidney, liver, heart and lung. Current armamentarium of pharmacologic and biologic immunosuppression has achieved remarkable patient and allograft survival rates in the first few years post transplantation(1-5). However, a number of critical challenges have limited successful long-term outcomes in solid organ transplant, namely, toxicities associated with chronic and non-specific immunosuppression(6-9), and graft loss due to antibody mediated rejection (ABMR) despite immunosuppression(10-12). Furthermore, a distinct but related immune barrier to transplant is the presence of pre-formed anti-HLA allo-antibodies that preclude sensitized recipients from the life-saving option of transplantation but also impact graft survival following transplant(13-15). In this regard, current strategies for desensitization targeting B cells (anti-CD20)(16-18), plasma cells (protease inhibitors)(19, 20) and antibodies (IVIG, plasmapheresis)(21, 22) have been largely ineffective. Thus, given the limited availability of precious organs, there is a clear need for creative approaches to achieve safe and effective immunomodulation that will realize the goal of “one [organ] for life”(23)

Over the last decade, T cell therapies using both endogenous and gene-edited T cells have yielded remarkable breakthroughs in cancer biology spurring the development of several T cell platforms for application in other disease arenas including auto- and allo-immunity. T cell therapies in immuno-oncology have reached clinical phase development, ushering an exciting era in the field of allo-immune modulation(24, 25). In the context of organ transplantation, both regulatory as well as engineered CAR T cells may afford therapeutic benefit. Here, we discuss the potential of polyclonal and antigen-specific T cells (Tregs) in modulating alloimmune responses, as well as engineered CAR T cells that could be harnessed for tolerance induction or desensitization.

Induction of Immunologic Tolerance Using Regulatory T cell (Treg) Therapy

The therapeutic potential of Tregs has been long recognized since the phenotypic identification of CD4+CD25+ T cells with immune suppressive properties by Sakaguchi and colleagues(26). Subsequent work identified FoxP3 as a critical transcription factor for Tregs and highlighted the pleiotropic suppressive effects of Tregs by virtue of their impact on antigen-presenting cell, T cell, and B cell functions. The demonstration that adoptively-transferred Tregs can prolong the survival of skin allografts had clear therapeutic implications and has prompted clinical development of Treg therapies for the induction of immunologic tolerance in solid organ transplantation(27, 28). Indeed, correlations between the level or frequency of circulating endogenous Tregs and rejection-free graft survival have been demonstrated(29-33).

Clinical evaluation of Treg therapies for organ transplantation began with infusion of polyclonal Tregs(34). Once this was determined to be safe in the setting of kidney and liver transplant, several trials have been undertaken to evaluate the efficacy of polyclonal Treg therapy in transplant, and whether this approach could help minimize the immunosuppressive regimen, recently reviewed in “Regulatory T cells: tolerance induction in solid organ transplantation.”(35). The ONE Study conducted by the multi-center consortium separately evaluated seven cell-based therapies using Tregs, tolerogenic dendritic cells, or macrophages as suppressive agents in living-donor kidney transplant recipients(36). While there was no statistically significant difference in the rate of biopsy-proven rejection compared to a control cohort, there was importantly no increased incidence of infection supporting the safety of polyclonal Treg therapy. Another phase-I study conducted in recipients of cadaveric liver transplants who received autologous polyclonal Tregs 2-16 months after transplant also demonstrated safety of the approach(37). While there were no cases of rejection during the 6-month follow-up, there was no clear evidence of efficacy attributable to the infused cells. These early studies which support feasibility and safety have primarily employed non-specific, polyclonal Tregs. Collectively, their results have inspired studies of antigen-specific or donor-reactive Tregs in both kidney and liver transplant recipients(36, 37). Compared to non-specific polyclonal Tregs, a product enriched for alloantigen-specific Tregs has multiple theoretical advantages. For instance, specificity may limit global immunodeficiency which could be a precursor to infection and malignancy. Its improved efficacy has been demonstrated in pre-clinical studies(38, 39).

Collectively, although Tregs have been shown to be safe in the preclinical and clinical studies performed thus far, antigen receptor-engineered Tregs with defined specificities has the potential to provide a more effective and safer therapy to modulate alloimmune responses.

Chimeric Antigen Receptor T Cell Engineering

Autologous T-cells, engineered to express a CAR, have provided breakthroughs in oncology(40). CARs are synthetic receptors that provided defined-target specificity in an MHC-independent manner. This is achieved by linking variable domains of an antibody via a transmembrane domain to intracellular T cell activation domains. First-generation CARs employed the zeta chain of the CD3 complex on the intracellular side to induce T cell activation. Optimized CARs include a co-stimulatory domain promoting complete T cell activation. These so-called second-generation CARs have proven highly efficacious against hematological malignancies. Several CD19-directed CAR T cells have now been FDA-approved for treatment of B-cell lineage malignancies and additional CAR therapies (e.g., anti-BCMA) are expected to be approved soon. These successes lay the foundation for expansion of CAR-based approaches beyond oncology, including the field of organ transplantation.

Induction of immunologic tolerance

The efficacy of CAR Tregs to dampen alloimmune response was demonstrated by CAR Tregs recognizing HLA-A2 in mouse models, which proved effective in preventing graft versus host disease (GVHD)(41). Similarly, CAR Tregs were demonstrated to be more potent than polyclonal Tregs at minimizing graft injury in immunodeficient mice engrafted with human HLA-A2+ skin(42-44). More recently, donor-specific CAR Tregs were evaluated for their ability to prevent skin allograft rejection in naïve and in previously sensitized mice (40). Six weeks post-transplant, despite the absence of any detectable CAR T cells in circulation, the recipients had a significantly lower concentration of DSAs, suggesting that antigen-specific CAR T cells can potentially prevent DSA formation by inhibiting the generation of donor reactive memory B and T cells(45). However, this approach is likely limited to prevention as the CAR Tregs were ineffective at eliminating established DSA in sensitized mice.

Although preclinical murine models have demonstrated the efficacy of CAR Tregs in the context of solid organ transplantation, most of these models have targeted HLA-A2 reactive immune cells(46); expanding this approach to other HLA epitopes is warranted. For instance, an increasing body evidence supports the finding that antibodies against HLA-C, which were previously thought to be clinically insignificant, may contribute to the evolution of antibody mediated rejection(47, 48). Additionally, studies have found that antibodies commonly arise against HLA-A24, B27, and B7 alleles at frequencies comparable to those against HLA-A2(49, 50). Thus, in order to create therapies that have a wide applicability, a method that can target DSAs that arise to HLA epitopes beyond HLA-A2 will be necessary.

CAR T cell therapy to achieve desensitization

CAR T cells targeting CD19 have resulted in durable remission of B cell-lineage malignancies. While global B cell depletion is an undesirable off-tumor on-target side-effect, it can be leveraged as a tool to deplete alloreactive or autoreactive pathogenic B cells. Indeed, Dr. Schett and colleagues employed CD19 targeted CAR T therapy solely for the purpose of eliminating autoimmune B cells in a patient with severe systemic lupus erythematosus refractory to numerous chemo-immunotherapies including the B cell depleting agents rituximab and belimumab(24). Administration of a single dose of CAR T cells resulted in B cell aplasia which was sustained for at least 44 days and which was accompanied by decline of anti-dsDNA antibodies to undetectable level, restoration of complement component levels, and normalization of proteinuria, all within 44 days of CAR T infusion. These were also accompanied by a remarkable improvement in SLEDAI score of disease activity to the point of complete remission by day 44. Certainly, evaluation of more patients with longer post-CAR T follow-up is needed. Importantly, the safety profile appears favorable for further evaluation of CAR T cells in these and other non-cancer patient cohorts.

It was important to determine if a similar approach would also be effective in elimination of pre-formed anti-HLA alloantibodies. This was studied in patients who achieved remission following CART-19 treatment with ongoing B cell aplasia. We and others demonstrated persistence of numerous pre-existing protective vaccine/pathogen specific antibodies and anti-HLA alloantibodies in patients who achieved durable B cell aplasia following CD19-directed CAR T cell therapy(51-53). These findings highlight the importance of CD19-negative long-lived plasma cells (LLPCs) in maintaining such immune responses. Thus, elimination of anti-HLA alloantibodies likely requires depletion of both B cells as well as LLPCs(51, 54, 55). In this regard, several CAR platforms currently in development for the treatment of multiple myeloma, a malignancy of PCs, might be effective. For instance, a CAR T cell targeting the B-cell maturation antigen (BCMA) has already demonstrated efficacy in patients with multiple myeloma, resulting in depletion of malignant plasma cells in the bone marrow(56), and possibly even physiological PCs(57, 58), though more evidence is needed to conclude the latter definitively. Combination of a CAR T cell targeting BCMA with a CAR T cell targeting CD19 has resulted in depletion of anti-HLA allo-Abs in human subjects(59), and this strategy can be readily adapted to desensitize candidates on the waitlist for solid organ transplantation with high allo-Ab burden. For example, for kidney transplant alone, 3,000 candidates are very highly allo-sensitized with anti-HLA alloantibodies against 99.9-100% of the potential donor population. For this subset of patients who find it extremely challenging to find a compatible organ for transplant, CAR T cell therapy for depletion of donor-specific B cells and PCs may result in a clinically meaningful reduction of allo-Abs. This strategy is expected to be tested in an upcoming clinical trial (Clinical Trials in Organ Transplant, NIH/NIAID). It is also worth mentioning that bispecific antibodies that bind CD3 and BCMA, thus bridging cytolytic T cells with PCs are also being evaluated as a desensitization strategy in transplant candidates (clinicaltrials.gov, NCT05137054).

CAR T cell therapies that target lineage-specific antigens (e.g., CD19) are unfortunately relatively non-specific. To avoid depletion of protective B cells and PCs, a novel CAR platform that targets antigen-specific B cells is a more attractive option. Proof-of-concept studies have utilized target auto- or allo-antigens, such as desmoglein 3 or Factor VIII, as the extracellular CAR domain to re-direct engineered T cells to B cells expressing only cognate antigen-specific BCRs(60, 61). Could such a strategy be used to target the highly-polyclonal pool of allo-HLA specific B cells and/or T cells? In this setting, HLA being the immune target, the platform would call for converting HLA molecules into T cell activating receptors. For example, β2M complexes with the heavy chain of HLA class I and, thus, could be engineered to deliver activation signals. β2M linked to an intracellular T cell-activating domain (β2M-CAR) could theoretically associate with endogenous HLA class I heavy chain, endowing the engineered T cells to selectively target HLA class I allo-reactive B and/or T cells. A similar strategy may be used to convert HLA class II molecules into CARs but would require allele-specific constructs given the absence of a monomorphic binding partner such as β2M.

Indeed, this strategy of leveraging MHC molecules as CARs has been tested in the context of autoimmunity to target diabetogenic T cells. Gross and colleagues linked β2M-CD3ζ to peptides such as ones derived from b cell autoantigens IGRP and insulin(62, 63). T cells, from NOD mice, expressing this CAR construct were able to target autoreactive T cells, and reduced insulitis and hyperglycemia(62).

Application of this and similar strategies to target allo-reactive T cells in the context of transplantation must contend with additional important challenges such as the vast and undefined pMHC specificity of the allo-reactive T cell pool. In a peptide-agnostic manner, Quach et. al. used the β2M-CAR without peptide-linkage, relying on its complex with endogenously-loaded MHC-I heavy chains. In vitro studies demonstrated the ability of CAR T cells to suppress activation and proliferation of allo-reactive T cells in mixed lymphocyte reactions(64). Interestingly, they found suppression of both allo-reactive CD8 and CD4 T cells despite using only an HLA class I-based CAR, which may point to direct and indirect methods of activity. Harnessing HLA complexes as activating receptors has also been explored for targeting MHC-II directed pathogenic autoimmunity(65). Transgenic mice in which hCD2 promoter driven expression of class II chains (IASα and IASβ) each linked to CD3ζ, along with linkage to MBP peptide (to IASβ), resulted in CAR expressing CD4 and CD8 T cells. These T cells exhibited pMHC-specific in vitro cytotoxicity and, in vivo, were able to suppress expansion of MBP-specific T cells and suppress EAE in both prevention and treatment models.

While engineered T cell platforms to target allo-reactive T cells are still in a very early phase of development, it is important to consider potential application challenges that are unique to transplant. One, for instance, is the choice between use of engineered recipient (autologous T cell product) versus donor T cells (allogeneic T cell product). While only the latter expresses allo-HLA that could be CAR-modified, additional modification, such as TCR deletion, may be needed to eliminate the risk of GVHD. The alternative use of recipient T cells presents a distinct challenge – that multiple allo-HLA molecules must be co-delivered. However, advances in CAR manufacturing (see Future directions), particularly in the delivery of CAR genes/mRNA may make this a feasible approach. Regardless of the T cell source, another consideration is the timing of CAR treatment relative to transplantation. Strategies focused on pre-transplant desensitization are clinically feasible whereas attempting to eliminate allo-reactive cells after transplant to treat rejection must address concomitant T cell immunosuppression. Certainly, post-transplant immunosuppression may also impact the persistence and long-term efficacy of pre-transplant administered T cell therapies.

To date, no study has reported the use of HLA-based CAR T strategies to target allo-specific B cells (Figure 1). A B cell targeting strategy could be used as a desensitization approach prior to transplantation and may even provide clinically meaningful effect with a single or limited set of HLA alleles to create “windows” in a patient’s broad allo-immune repertoire. Targeting humoral immune responses in an antigen-specific manner is confounded by IgG-secreting LLPCs, which may not display their specificity to engineered T cells as surface BCR(66). This potential challenge will need to be tested empirically; if they are found to be resistant to elimination by antigen-specific CAR T cells, non-specific PC-targeted approaches (e.g. BCMA CAR T cells) may have to be combined with the antigen-specific B-cell targeted methods.

Figure 1. Schematics of MHC-based CARs targeting alloreactive lymphocytes.

Figure 1.

(A) Conventional CARs such as CD19-directed CARs target lineage antigens and mediate non-specific B cell elimination. Antigen-based CARs may be used to recognize only antigen-specific B cells. An engineered beta-2-microglobulin that is linked to T cell signaling domains associates with endogenous MHC-I heavy chain, converting the MHC-I complex into a CAR-like receptor which could engage allo-reactive B and T cells. (B) Both MHC-I and MHC-II may be converted to T cell activating complexes by fusing signal activating domains to different components of each complex. Both autologous and allogeneic platforms are conceivable with each presenting unique advantages and challenges.

Future Directions

Developments in basic science and translational T cell immunology and bioengineering have enormous potential of providing more effective precision therapies for modulation of alloimmunity. Among the most exciting advances in the field are those related to manufacturing engineered T cells. For example, recent studies that have shortened CAR T cell manufacturing from 10-14 days to a 1-2 day process provide an opportunity for ease of delivery and also improves the biologic potency of the product (i.e. less ex-vivo T cell expansion-induced exhaustion)(67-69). All three platforms demonstrate robust in vivo proliferation of infused CAR T cells, including in 1st in human clinical trials conducted by two of the groups(68, 69). Preclinical studies reported by Ghassemi et. al. and Yang et. al. also demonstrated superiority of their shortened manufacturing process compared to conventionally manufactured CAR T cells in preclinical models. Conventional CAR T manufacturing involves ex-vivo T cell activation typically followed by CAR gene introduction employing lentiviral or retroviral vectors. In contrast, Ghassemi et. al. described a process in which T cells may be transduced, using a modified formulation of the culture medium and the surface area-to-volume ratio of the culture vessel, to yield efficient lentiviral transduction without the need for T cell activation. One potential advantage of their method is the avoidance of T cell activation ex-vivo which may further mitigate adverse effects of activation using artificial anti-CD3/anti-CD28 reagents.

Other exciting developments in the CAR T field are approaches for in-vivo CAR T cell “manufacturing” completely avoiding all issues related to ex vivo T cell expansion(70, 71). Recently, Rurik et. al. combined mRNA therapeutics, targeted lipid nanoparticle (LNP) delivery, and CAR technology to treat pathogenic cardiac fibrosis following myocardial injury. Delivery of CD5-targeted LNP encapsulating mRNA encoding a CAR targeting fibroblast activation protein (FAP-CAR) created FAP-CAR T cells in vivo which were effective at reducing fibrosis that had already been established in a murine model of hypertension induced cardiac injury(71). The inherently transient nature of these mRNA CAR T cells is also an attractive feature for such non-cancer applications such as solid organ transplant.

Conclusions

Given the vast complexities and challenges of allo-immunity, it is heartening to acknowledge that CAR T approaches are one among several other technologies that are currently in development for desensitization and tolerance induction. As in the field of oncology, we may need a combination of approaches to achieve successful long-term graft survival. Combining CAR T therapy with standard immunosuppression in the context of organ transplant presents unique challenges that will require careful consideration and evaluation. For instance, while desensitization strategies in the pre-transplant setting may be more straightforward, attempting to eliminate allo-reactive cells after transplant to treat rejection must address concomitant T cell immunosuppression, which may impact the persistence and long-term efficacy of CAR T cells (72). Nonetheless, the recent developments in basic biology and clinical applications of CAR T cell therapy make this an attractive tool in the armamentarium of immune intervention strategies. As these enter the clinical arena, the immunology community will be witness to the elimination of two major barriers in the field of organ transplantation (desensitization and tolerance induction).

Acknowledgements.

This work was supported by the Institute for Translational Medicine and Therapeutics at Penn (Dr. Muralidharan), Gift of Life Transplant Foundation (Drs. Naji and Bhoj), Burroughs Wellcome Fund (Dr. Bhoj), and The Colton Center for Autoimmunity at Penn (Dr. Bhoj).

References

  • 1.Doberer K, Duerr M, Halloran PF, Eskandary F, Budde K, Regele H, Reeve J, Borski A, Kozakowski N, Reindl-Schwaighofer R, Waiser J, Lachmann N, Schranz S, Firbas C, Muhlbacher J, Gelbenegger G, Perkmann T, Wahrmann M, Kainz A, Ristl R, Halleck F, Bond G, Chong E, Jilma B, and Bohmig GA. 2021. A Randomized Clinical Trial of Anti-IL-6 Antibody Clazakizumab in Late Antibody-Mediated Kidney Transplant Rejection. J Am Soc Nephrol 32: 708–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ravichandran AK, Schilling JD, Novak E, Pfeifer J, Ewald GA, and Joseph SM. 2013. Rituximab is associated with improved survival in cardiac allograft patients with antibody-mediated rejection: a single center review. Clin Transplant 27: 961–967. [DOI] [PubMed] [Google Scholar]
  • 3.Everly MJ, Everly JJ, Susskind B, Brailey P, Arend LJ, Alloway RR, Roy-Chaudhury P, Govil A, Mogilishetty G, Rike AH, Cardi M, Wadih G, Tevar A, and Woodle ES. 2008. Bortezomib provides effective therapy for antibody- and cell-mediated acute rejection. Transplantation 86: 1754–1761. [DOI] [PubMed] [Google Scholar]
  • 4.Stegall MD, Diwan T, Raghavaiah S, Cornell LD, Burns J, Dean PG, Cosio FG, Gandhi MJ, Kremers W, and Gloor JM. 2011. Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am J Transplant 11: 2405–2413. [DOI] [PubMed] [Google Scholar]
  • 5.Kaposztas Z, Podder H, Mauiyyedi S, Illoh O, Kerman R, Reyes M, Pollard V, and Kahan BD. 2009. Impact of rituximab therapy for treatment of acute humoral rejection. Clin Transplant 23: 63–73. [DOI] [PubMed] [Google Scholar]
  • 6.Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, Maciuca R, Zhang D, Garg JP, Brunetta P, Appel G, and L. I. Group. 2012. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum 64: 1215–1226. [DOI] [PubMed] [Google Scholar]
  • 7.Eskandary F, Regele H, Baumann L, Bond G, Kozakowski N, Wahrmann M, Hidalgo LG, Haslacher H, Kaltenecker CC, Aretin MB, Oberbauer R, Posch M, Staudenherz A, Handisurya A, Reeve J, Halloran PF, and Bohmig GA. 2018. A Randomized Trial of Bortezomib in Late Antibody-Mediated Kidney Transplant Rejection. J Am Soc Nephrol 29: 591–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cabanillas F, Liboy I, Pavia O, and Rivera E. 2006. High incidence of non-neutropenic infections induced by rituximab plus fludarabine and associated with hypogammaglobulinemia: a frequently unrecognized and easily treatable complication. Ann Oncol 17: 1424–1427. [DOI] [PubMed] [Google Scholar]
  • 9.Pallier A, Hillion S, Danger R, Giral M, Racape M, Degauque N, Dugast E, Ashton-Chess J, Pettre S, Lozano JJ, Bataille R, Devys A, Cesbron-Gautier A, Braudeau C, Larrose C, Soulillou JP, and Brouard S. 2010. Patients with drug-free long-term graft function display increased numbers of peripheral B cells with a memory and inhibitory phenotype. Kidney Int 78: 503–513. [DOI] [PubMed] [Google Scholar]
  • 10.Redfield RR, Ellis TM, Zhong W, Scalea JR, Zens TJ, Mandelbrot D, Muth BL, Panzer S, Samaniego M, Kaufman DB, Astor BC, and Djamali A. 2016. Current outcomes of chronic active antibody mediated rejection - A large single center retrospective review using the updated BANFF 2013 criteria. Hum Immunol 77: 346–352. [DOI] [PubMed] [Google Scholar]
  • 11.Dorje C, Midtvedt K, Holdaas H, Naper C, Strom EH, Oyen O, Leivestad T, Aronsen T, Jenssen T, Flaa-Johnsen L, Lindahl JP, Hartmann A, and Reisaeter AV. 2013. Early versus late acute antibody-mediated rejection in renal transplant recipients. Transplantation 96: 79–84. [DOI] [PubMed] [Google Scholar]
  • 12.Sun Q, Liu ZH, Ji S, Chen J, Tang Z, Zeng C, Zheng C, and Li LS. 2006. Late and early C4d-positive acute rejection: different clinico-histopathological subentities in renal transplantation. Kidney Int 70: 377–383. [DOI] [PubMed] [Google Scholar]
  • 13.Coutance G, Nguyen L, Lebreton G, Ouldamar S, Rouvier P, Saheb S, Bouglé A, Bréchot N, Leprince P, and Varnous S. 2019. Pre-formed donor specific antibodies>3000 MFI managed at the time of transplantation predicts early antibody-mediated rejection after heart transplantation in a large cohort of patients. Archives of Cardiovascular Diseases Supplements 11: 140–141. [Google Scholar]
  • 14.Eng HS, Bennett G, Chang SH, Dent H, McDonald SP, Bardy P, Coghlan P, Russ GR, and Coates PT. 2011. Donor human leukocyte antigen specific antibodies predict development and define prognosis in transplant glomerulopathy. Hum Immunol 72: 386–391. [DOI] [PubMed] [Google Scholar]
  • 15.Ziemann M, Altermann W, Angert K, Arns W, Bachmann A, Bakchoul T, Banas B, von Borstel A, Budde K, Ditt V, Einecke G, Eisenberger U, Feldkamp T, Gorg S, Guthoff M, Habicht A, Hallensleben M, Heinemann FM, Hessler N, Hugo C, Kaufmann M, Kauke T, Koch M, Konig IR, Kurschat C, Lehmann C, Marget M, Muhlfeld A, Nitschke M, Pego da Silva L, Quick C, Rahmel A, Rath T, Reinke P, Renders L, Sommer F, Spriewald B, Staeck O, Stippel D, Susal C, Thiele B, Zecher D, and Lachmann N. 2019. Preformed Donor-Specific HLA Antibodies in Living and Deceased Donor Transplantation: A Multicenter Study. Clin J Am Soc Nephrol 14: 1056–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bailly E, Ville S, Blancho G, Morelon E, Bamoulid J, Caillard S, Chatelet V, Malvezzi P, Tourret J, Vuiblet V, Anglicheau D, Bertrand D, Grimbert P, Haidar F, Hazzan M, Kamar N, Merville P, Mousson C, Pernin V, Pouteil-Noble C, Purgus R, Sayegh J, Westeel PF, Sautenet B, Gatault P, and Buchler M. 2020. An extension of the RITUX-ERAH study, multicenter randomized clinical trial comparing rituximab to placebo in acute antibody-mediated rejection after renal transplantation. Transpl Int 33: 786–795. [DOI] [PubMed] [Google Scholar]
  • 17.Redfield RR, Jordan SC, Busque S, Vincenti F, Woodle ES, Desai N, Reed EF, Tremblay S, Zachary AA, Vo AA, Formica R, Schindler T, Tran H, Looney C, Jamois C, Green C, Morimoto A, Rajwanshi R, Schroeder A, Cascino MD, Brunetta P, and Borie D. 2019. Safety, pharmacokinetics, and pharmacodynamic activity of obinutuzumab, a type 2 anti-CD20 monoclonal antibody for the desensitization of candidates for renal transplant. Am J Transplant 19: 3035–3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Macklin PS, Morris PJ, and Knight SR. 2017. A systematic review of the use of rituximab for the treatment of antibody-mediated renal transplant rejection. Transplant Rev (Orlando) 31: 87–95. [DOI] [PubMed] [Google Scholar]
  • 19.Moreno Gonzales MA, Gandhi MJ, Schinstock CA, Moore NA, Smith BH, Braaten NY, and Stegall MD. 2017. 32 Doses of Bortezomib for Desensitization Is Not Well Tolerated and Is Associated With Only Modest Reductions in Anti-HLA Antibody. Transplantation 101: 1222–1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Waiser J, Duerr M, Schonemann C, Rudolph B, Wu K, Halleck F, Budde K, and Lachmann N. 2016. Rituximab in Combination With Bortezomib, Plasmapheresis, and High-Dose IVIG to Treat Antibody-Mediated Renal Allograft Rejection. Transplant Direct 2: e91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mella A, Gallo E, Messina M, Caorsi C, Amoroso A, Gontero P, Verri A, Maletta F, Barreca A, Fop F, and Biancone L. 2018. Treatment with plasmapheresis, immunoglobulins and rituximab for chronic-active antibody-mediated rejection in kidney transplantation: Clinical, immunological and pathological results. World J Transplant 8: 178–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Moreso F, Crespo M, Ruiz JC, Torres A, Gutierrez-Dalmau A, Osuna A, Perello M, Pascual J, Torres IB, Redondo-Pachon D, Rodrigo E, Lopez-Hoyos M, and Seron D. 2018. Treatment of chronic antibody mediated rejection with intravenous immunoglobulins and rituximab: A multicenter, prospective, randomized, double-blind clinical trial. Am J Transplant 18: 927–935. [DOI] [PubMed] [Google Scholar]
  • 23.Kawai T, Leventhal J, Madsen JC, Strober S, Turka LA, and Wood KJ. 2014. Tolerance: one transplant for life. Transplantation 98: 117–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mougiakakos D, Kronke G, Volkl S, Kretschmann S, Aigner M, Kharboutli S, Boltz S, Manger B, Mackensen A, and Schett G. 2021. CD19-Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus. N Engl J Med 385: 567–569. [DOI] [PubMed] [Google Scholar]
  • 25.Patel U, Abernathy J, Savani BN, Oluwole O, Sengsayadeth S, and Dholaria B. 2022. CAR T cell therapy in solid tumors: A review of current clinical trials. EJHaem 3: 24–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, and Toda M. 1995. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155: 1151–1164. [PubMed] [Google Scholar]
  • 27.Issa F, Hester J, Goto R, Nadig SN, Goodacre TE, and Wood K. 2010. Ex vivo-expanded human regulatory T cells prevent the rejection of skin allografts in a humanized mouse model. Transplantation 90: 1321–1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Vokaer B, Charbonnier LM, Lemaitre PH, Spilleboudt C, and Le Moine A. 2013. IL-17A and IL-2-expanded regulatory T cells cooperate to inhibit Th1-mediated rejection of MHC II disparate skin grafts. PLoS One 8: e76040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kwun J, Matignon M, Manook M, Guendouz S, Audard V, Kheav D, Poullot E, Gautreau C, Ezekian B, Bodez D, Damy T, Faivre L, Menouche D, Yoon J, Park J, Belhadj K, Chen D, Bilewski AM, Yi JS, Collins B, Stegall M, Farris AB, Knechtle S, and Grimbert P. 2019. Daratumumab in Sensitized Kidney Transplantation: Potentials and Limitations of Experimental and Clinical Use. J Am Soc Nephrol 30: 1206–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bestard O, Cruzado JM, Rama I, Torras J, Goma M, Seron D, Moreso F, Gil-Vernet S, and Grinyo JM. 2008. Presence of FoxP3+ regulatory T Cells predicts outcome of subclinical rejection of renal allografts. J Am Soc Nephrol 19: 2020–2026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bestard O, Cunetti L, Cruzado JM, Lucia M, Valdez R, Olek S, Melilli E, Torras J, Mast R, Goma M, Franquesa M, and Grinyo JM. 2011. Intragraft regulatory T cells in protocol biopsies retain foxp3 demethylation and are protective biomarkers for kidney graft outcome. Am J Transplant 11: 2162–2172. [DOI] [PubMed] [Google Scholar]
  • 32.Grimbert P, Mansour H, Desvaux D, Roudot-Thoraval F, Audard V, Dahan K, Berrehar F, Dehoulle-Poillet C, Farcet JP, Lang P, and Le Gouvello S. 2007. The regulatory/cytotoxic graft-infiltrating T cells differentiate renal allograft borderline change from acute rejection. Transplantation 83: 341–346. [DOI] [PubMed] [Google Scholar]
  • 33.San Segundo D, Fernandez-Fresnedo G, Rodrigo E, Ruiz JC, Gonzalez M, Gomez-Alamillo C, Arias M, and Lopez-Hoyos M. 2012. High regulatory T-cell levels at 1 year posttransplantation predict long-term graft survival among kidney transplant recipients. Transplant Proc 44: 2538–2541. [DOI] [PubMed] [Google Scholar]
  • 34.Todo S, Yamashita K, Goto R, Zaitsu M, Nagatsu A, Oura T, Watanabe M, Aoyagi T, Suzuki T, Shimamura T, Kamiyama T, Sato N, Sugita J, Hatanaka K, Bashuda H, Habu S, Demetris AJ, and Okumura K. 2016. A pilot study of operational tolerance with a regulatory T-cell-based cell therapy in living donor liver transplantation. Hepatology 64: 632–643. [DOI] [PubMed] [Google Scholar]
  • 35.Vaikunthanathan T, Safinia N, Boardman D, Lechler RI, and Lombardi G. 2017. Regulatory T cells: tolerance induction in solid organ transplantation. Clin Exp Immunol 189: 197–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sawitzki B, Harden PN, Reinke P, Moreau A, Hutchinson JA, Game DS, Tang Q, Guinan EC, Battaglia M, Burlingham WJ, Roberts ISD, Streitz M, Josien R, Boger CA, Scotta C, Markmann JF, Hester JL, Juerchott K, Braudeau C, James B, Contreras-Ruiz L, van der Net JB, Bergler T, Caldara R, Petchey W, Edinger M, Dupas N, Kapinsky M, Mutzbauer I, Otto NM, Ollinger R, Hernandez-Fuentes MP, Issa F, Ahrens N, Meyenberg C, Karitzky S, Kunzendorf U, Knechtle SJ, Grinyo J, Morris PJ, Brent L, Bushell A, Turka LA, Bluestone JA, Lechler RI, Schlitt HJ, Cuturi MC, Schlickeiser S, Friend PJ, Miloud T, Scheffold A, Secchi A, Crisalli K, Kang SM, Hilton R, Banas B, Blancho G, Volk HD, Lombardi G, Wood KJ, and Geissler EK. 2020. Regulatory cell therapy in kidney transplantation (The ONE Study): a harmonised design and analysis of seven non-randomised, single-arm, phase 1/2A trials. Lancet 395: 1627–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sanchez-Fueyo A, Whitehouse G, Grageda N, Cramp ME, Lim TY, Romano M, Thirkell S, Lowe K, Fry L, Heward J, Kerr A, Ali J, Fisher C, Lewis G, Hope A, Kodela E, Lyne M, Farzaneh F, Kordasti S, Rebollo-Mesa I, Jose Lozano J, Safinia N, Heaton N, Lechler R, Martinez-Llordella M, and Lombardi G. 2020. Applicability, safety, and biological activity of regulatory T cell therapy in liver transplantation. Am J Transplant 20: 1125–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ratnasothy K, Jacob J, Tung S, Boardman D, Lechler RI, Sanchez-Fueyo A, Martinez-Llordella M, and Lombardi G. 2019. IL-2 therapy preferentially expands adoptively transferred donor-specific Tregs improving skin allograft survival. Am J Transplant 19: 2092–2100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sagoo P, Ali N, Garg G, Nestle FO, Lechler RI, and Lombardi G. 2011. Human regulatory T cells with alloantigen specificity are more potent inhibitors of alloimmune skin graft damage than polyclonal regulatory T cells. Sci Transl Med 3: 83ra42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Phimister EG, and Rubin EJ. 2022. Targeting Cytotoxic T Cells to Tumor. N Engl J Med 386: 2145–2148. [DOI] [PubMed] [Google Scholar]
  • 41.MacDonald KG, Hoeppli RE, Huang Q, Gillies J, Luciani DS, Orban PC, Broady R, and Levings MK. 2016. Alloantigen-specific regulatory T cells generated with a chimeric antigen receptor. J Clin Invest 126: 1413–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Boardman DA, Philippeos C, Fruhwirth GO, Ibrahim MA, Hannen RF, Cooper D, Marelli-Berg FM, Watt FM, Lechler RI, Maher J, Smyth LA, and Lombardi G. 2017. Expression of a Chimeric Antigen Receptor Specific for Donor HLA Class I Enhances the Potency of Human Regulatory T Cells in Preventing Human Skin Transplant Rejection. Am J Transplant 17: 931–943. [DOI] [PubMed] [Google Scholar]
  • 43.Dawson NA, Lamarche C, Hoeppli RE, Bergqvist P, Fung VC, McIver E, Huang Q, Gillies J, Speck M, Orban PC, Bush JW, Mojibian M, and Levings MK. 2019. Systematic testing and specificity mapping of alloantigen-specific chimeric antigen receptors in regulatory T cells. JCI Insight 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Noyan F, Zimmermann K, Hardtke-Wolenski M, Knoefel A, Schulde E, Geffers R, Hust M, Huehn J, Galla M, Morgan M, Jokuszies A, Manns MP, and Jaeckel E. 2017. Prevention of Allograft Rejection by Use of Regulatory T Cells With an MHC-Specific Chimeric Antigen Receptor. Am J Transplant 17: 917–930. [DOI] [PubMed] [Google Scholar]
  • 45.Sicard A, Lamarche C, Speck M, Wong M, Rosado-Sanchez I, Blois M, Glaichenhaus N, Mojibian M, and Levings MK. 2020. Donor-specific chimeric antigen receptor Tregs limit rejection in naive but not sensitized allograft recipients. Am J Transplant 20: 1562–1573. [DOI] [PubMed] [Google Scholar]
  • 46.Martin A, Daris M, Johnston JA, and Cui J. 2021. HLA-A*02:01-directed chimeric antigen receptor/forkhead box P3-engineered CD4+ T cells adopt a regulatory phenotype and suppress established graft-versus-host disease. Cytotherapy 23: 131–136. [DOI] [PubMed] [Google Scholar]
  • 47.Aubert O, Bories MC, Suberbielle C, Snanoudj R, Anglicheau D, Rabant M, Martinez F, Scemla A, Legendre C, and Sberro-Soussan R. 2014. Risk of antibody-mediated rejection in kidney transplant recipients with anti-HLA-C donor-specific antibodies. Am J Transplant 14: 1439–1445. [DOI] [PubMed] [Google Scholar]
  • 48.Visentin J, Couzi L, and Taupin JL. 2021. Clinical relevance of donor-specific antibodies directed at HLA-C: A long road to acceptance. HLA 97: 3–14. [DOI] [PubMed] [Google Scholar]
  • 49.Fu Q, Wang C, Zeng W, and Liu L. 2012. The correlation of HLA allele frequencies and HLA antibodies in sensitized kidney transplantation candidates. Transplant Proc 44: 217–221. [DOI] [PubMed] [Google Scholar]
  • 50.Zheng J, Kuang PD, Zhang Y, Zhao Q, He XL, Ding XM, and Xue WJ. 2019. [Relationship of distribution frequency of HLA antigen/antibody and PIRCHE score with DSA production and AMR occurrence]. Zhonghua Yi Xue Za Zhi 99: 901–906. [DOI] [PubMed] [Google Scholar]
  • 51.Zhang Z, Schuster SJ, Lacey SF, Milone MC, Monos D, and Bhoj VG. 2020. Stable HLA antibodies following sustained CD19+ cell depletion implicate a long-lived plasma cell source. Blood Adv 4: 4292–4295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bhoj VG, Arhontoulis D, Wertheim G, Capobianchi J, Callahan CA, Ellebrecht CT, Obstfeld AE, Lacey SF, Melenhorst JJ, Nazimuddin F, Hwang WT, Maude SL, Wasik MA, Bagg A, Schuster S, Feldman MD, Porter DL, Grupp SA, June CH, and Milone MC. 2016. Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood 128: 360–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Walti CS, Krantz EM, Maalouf J, Boonyaratanakornkit J, Keane-Candib J, Joncas-Schronce L, Stevens-Ayers T, Dasgupta S, Taylor JJ, Hirayama AV, Bar M, Gardner RA, Cowan AJ, Green DJ, Boeckh MJ, Maloney DG, Turtle CJ, and Hill JA. 2021. Antibodies against vaccine-preventable infections after CAR-T cell therapy for B cell malignancies. JCI Insight 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Redfield R, Parsons R, Naji A, and Abt PL. 2011. Bortezomib alone is sufficient to cause sustained elimination of alloreactive plasma cells and donor-specific antibody in mice. Journal of the American College of Surgeons 213: S68–S69. [Google Scholar]
  • 55.Su H, Zhang CY, Lin JH, Hammes HP, and Zhang C. 2019. The Role of Long-Lived Plasma Cells in Antibody-Mediated Rejection of Kidney Transplantation: An Update. Kidney Dis (Basel) 5: 211–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Teoh PJ, and Chng WJ. 2021. CAR T-cell therapy in multiple myeloma: more room for improvement. Blood Cancer J 11: 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Markmann C, and Bhoj VG. 2021. On the road to eliminating long-lived plasma cells-"are we there yet?". Immunol Rev 303: 154–167. [DOI] [PubMed] [Google Scholar]
  • 58.Wang Y, Li C, Xia J, Li P, Cao J, Pan B, Tan X, Li H, Qi K, Wang X, Shi M, Jing G, Yan Z, Cheng H, Zhu F, Sun H, Sang W, Li D, Zhang X, Li Z, Zheng J, Liang A, Zhou J, and Xu K. 2021. Humoral immune reconstitution after anti-BCMA CAR T-cell therapy in relapsed/refractory multiple myeloma. Blood Adv 5: 5290–5299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Liu F, Zhang H, Wang X, Xiong Y, Cao Y, Su Y, Yi H, Feng J, Zhang W, Ma Y, Wada M, and Ma Y. 2019. First-in-Human Trial of Bcma-CD19 Compound CAR with Remarkable Donor-Specific Antibody Reduction. Blood 134: 38. [Google Scholar]
  • 60.Ellebrecht CT, Bhoj VG, Nace A, Choi EJ, Mao X, Cho MJ, Di Zenzo G, Lanzavecchia A, Seykora JT, Cotsarelis G, Milone MC, and Payne AS. 2016. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science 353: 179–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Parvathaneni K, and Scott DW. 2018. Engineered FVIII-expressing cytotoxic T cells target and kill FVIII-specific B cells in vitro and in vivo. Blood Adv 2: 2332–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Fishman S, Lewis MD, Siew LK, De Leenheer E, Kakabadse D, Davies J, Ziv D, Margalit A, Karin N, Gross G, and Wong FS. 2017. Adoptive Transfer of mRNA-Transfected T Cells Redirected against Diabetogenic CD8 T Cells Can Prevent Diabetes. Mol Ther 25: 456–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Margalit A, Fishman S, Berko D, Engberg J, and Gross G. 2003. Chimeric beta2 microglobulin/CD3zeta polypeptides expressed in T cells convert MHC class I peptide ligands into T cell activation receptors: a potential tool for specific targeting of pathogenic CD8(+) T cells. Int Immunol 15: 1379–1387. [DOI] [PubMed] [Google Scholar]
  • 64.Quach DH, Becerra-Dominguez L, Rouce RH, and Rooney CM. 2019. A strategy to protect off-the-shelf cell therapy products using virus-specific T-cells engineered to eliminate alloreactive T-cells. J Transl Med 17: 240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Jyothi MD, Flavell RA, and Geiger TL. 2002. Targeting autoantigen-specific T cells and suppression of autoimmune encephalomyelitis with receptor-modified T lymphocytes. Nat Biotechnol 20: 1215–1220. [DOI] [PubMed] [Google Scholar]
  • 66.Pinto D, Montani E, Bolli M, Garavaglia G, Sallusto F, Lanzavecchia A, and Jarrossay D. 2013. A functional BCR in human IgA and IgM plasma cells. Blood 121: 4110–4114. [DOI] [PubMed] [Google Scholar]
  • 67.Ghassemi S, Durgin JS, Nunez-Cruz S, Patel J, Leferovich J, Pinzone M, Shen F, Cummins KD, Plesa G, Cantu VA, Reddy S, Bushman FD, Gill SI, O'Doherty U, O'Connor RS, and Milone MC. 2022. Rapid manufacturing of non-activated potent CAR T cells. Nat Biomed Eng 6: 118–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yang J, He J, Zhang X, Li J, Wang Z, Zhang Y, Qiu L, Wu Q, Sun Z, Ye X, Yin W, Cao W, Shen L, Sersch M, and Lu P. 2022. Next-day manufacture of a novel anti-CD19 CAR-T therapy for B-cell acute lymphoblastic leukemia: first-in-human clinical study. Blood Cancer J 12: 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sperling AS, Nikiforow S, Nadeem O, Mo CC, Laubach JP, Anderson KC, Alonso A, Ikegawa S, Prabhala R, Hernandez Rodriguez D, Daley H, Shaw KL, Arihara Y, Ansari S, Quinn DS, Pearson D, Hack A, Treanor LM, Bu D, Mataraza J, Rispoli L, Credi M, Ritz J, De Vita S, and Munshi NC. 2021. Phase I Study of PHE885, a Fully Human BCMA-Directed CAR-T Cell Therapy for Relapsed/Refractory Multiple Myeloma Manufactured in <2 Days Using the T-Charge TM Platform. Blood 138: 3864–3864. [Google Scholar]
  • 70.Agarwal S, Hanauer JDS, Frank AM, Riechert V, Thalheimer FB, and Buchholz CJ. 2020. In Vivo Generation of CAR T Cells Selectively in Human CD4(+) Lymphocytes. Mol Ther 28: 1783–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rurik JG, Tombacz I, Yadegari A, Mendez Fernandez PO, Shewale SV, Li L, Kimura T, Soliman OY, Papp TE, Tam YK, Mui BL, Albelda SM, Pure E, June CH, Aghajanian H, Weissman D, Parhiz H, and Epstein JA. 2022. CAR T cells produced in vivo to treat cardiac injury. Science 375: 91–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Myburgh R, Kiefer JD, Russkamp NF, Magnani CF, Nunez N, Simonis A, Pfister S, Wilk CM, McHugh D, Friemel J, Muller AM, Becher B, Munz C, van den Broek M, Neri D, and Manz MG. 2020. Anti-human CD117 CAR T-cells efficiently eliminate healthy and malignant CD117-expressing hematopoietic cells. Leukemia 34: 2688–2703. [DOI] [PubMed] [Google Scholar]

RESOURCES