Long-Term Clinical and Immunological Profile of Kidney Transplant Patients Given Mesenchymal Stromal Cell Immunotherapy

Norberto Perico, Federica Casiraghi, Marta Todeschini, Monica Cortinovis, Eliana Gotti, Valentina Portalupi, Marilena Mister, Flavio Gaspari, Alessandro Villa, Sonia Fiori, Martino Introna, Elena Longhi, Giuseppe Remuzzi, Norberto Perico, Federica Casiraghi, Marta Todeschini, Monica Cortinovis, Eliana Gotti, Valentina Portalupi, Marilena Mister, Flavio Gaspari, Alessandro Villa, Sonia Fiori, Martino Introna, Elena Longhi, Giuseppe Remuzzi

Abstract

We report here the long-term clinical and immunological results of four living-donor kidney transplant patients given autologous bone marrow-derived mesenchymal stromal cells (MSCs) as part of a phase 1 study focused on the safety and feasibility of this cell therapy. According to study protocols implemented over time, based on initial early safety findings, the patients were given MSC at day 7 posttransplant (n = 2) or at day -1 pretransplant (n = 2) and received induction therapy with basiliximab and low-dose rabbit anti-thymocyte globulin (RATG) or RATG alone, and were maintained on low-dose ciclosporin (CsA)/mycophenolate mofetil (MMF). All MSC-treated patients had stable graft function during the 5- to 7-year follow-up, without increased susceptibility to infections or neoplasm. In three MSC recipients, but not historical control patients, circulating memory CD8+ T cell percentages remained lower than basal, coupled with persistent reduction of ex vivo donor-specific cytotoxicity. Two patients showed a long-lasting increase in the regulatory T cell/memory CD8+ T cell ratio, paralleled by high circulating levels of naïve and transitional B cells. In one of these two patients, CsA was successfully discontinued, and currently the low-dose MMF monotherapy is on the tapering phase. The study shows that MSC therapy is safe in the long term and could promote a pro-tolerogenic environment in selected patients. Extensive immunomonitoring of MSC-treated kidney transplant recipients could help selection of patients for safe withdrawal of maintenance immunosuppressive drugs (NCT00752479 and NCT02012153).

Keywords: B cells; kidney transplantation; memory CD8+ T cells; mesenchymal stromal cells; regulatory T cells; tolerance.

Figures

Figure 1
Figure 1
Clinical protocols of MSC-treated kidney transplant recipients implemented at our center. The first two living-related donor kidney transplant recipients adopted the initial clinical protocol (Clinical protocol #1), where autologous bone marrow-derived MSC were infused 7 days after transplantation and induction therapy comprised basiliximab and low-dose RATG. Since in both patients transient acute renal insufficiency occurred due to engraftment syndrome, the protocol was revised (Clinical protocol #2), and two subsequent patients received pretransplant MSC infusion and induction therapy with low-dose RATG alone. Abbreviations: CsA, ciclosporin; MMF, mycophenolate mofetil; MSC, mesenchymal stromal cell; RATG, rabbit anti-thymocyte globulin.
Figure 2
Figure 2
The rate of glomerular filtration rate (GFR) decline in the long term after transplantation. Panel (A) represents median slope of GFR in the first four patients given mesenchymal stromal cells (MSC) along with their respective follow-up (open circles) and in control kidney transplant recipients given either basiliximab/low-dose RATG or low-dose rabbit anti-thymocyte globulin (RATG) alone (closed circles) over 7 and 5 years posttransplantation, respectively. Measured GFR decreased to a significantly lower rate in the first four MSC-treated patients compared with living-related donor kidney transplant recipients in the basiliximab/low-dose RATG control group (n = 3) (−0.278 vs −1.834 mL/min/1.73 m2/year, respectively, P < 0.001). Panel (B) shows individual slopes of measured GFR in the first four patients given MSC along their respective follow-up.
Figure 3
Figure 3
Profile of circulating memory CD8+ T cells and regulatory T cells. Percentages of CD45RO+RA− memory CD8+ T cells (on CD3+CD8+ T cells) in mesenchymal stromal cell (MSC)-treated patients #1 (blue circles), #2 (orange circles) and in control kidney transplant recipients given basiliximab/low-dose rabbit anti-thymocyte globulin (RATG) induction therapy but not MSC (white histograms) during the follow-up (A) and in MSC-treated patients #3 (green circles) and # 4 (violet circles) and in control kidney transplant recipients given induction therapy with low-dose RATG alone but not MSC (gray histograms) during the follow-up (B). Panels (C,D) represent profile of percentages of regulatory T cells (Tregs) (Foxp3+CD127− on CD4+CD25high T cells) during the follow-up in MSC-treated patients #1 and #2 and in control patients given basiliximab/low-dose RATG (white histograms) and in MSC-patients #3 and #4 and in control patients given induction therapy with low-dose RATG alone (gray histograms), respectively. Ratio of Treg/memory CD8+ T cell percentages in MSC-treated patients and in control groups are shown in panels (E,F). Data from controls are mean ± SEM, *P < 0.05 vs pretransplant values.
Figure 4
Figure 4
Profile of naïve and transitional B cell counts in the peripheral blood. Number (A) and percentages [on total CD3−CD19+ B cells (B)] of naïve IgD+CD27− B cells in the peripheral blood during the follow-up in mesenchymal stromal cell (MSC)-treated patients #1 (blue circles) and #2 (orange circles) and in control kidney transplant recipients given basiliximab/low-dose rabbit anti-thymocyte globulin (RATG) induction therapy but not MSC (white histograms) during the follow-up (left panels) and in MSC-treated patients #3 (green circles) and #4 (violet circles) and in control kidney transplant recipients given induction therapy with low-dose RATG alone but not MSC (gray histograms) (right panels). Number (C) and percentages [on total CD3−CD19+ B cells (D)] of CD24highCD38high transitional B cells in MSC-treated patients #1 and #2 and in control patients given basiliximab/low-dose RATG (white histograms) (left panels) and in MSC-patients #3 and #4 and in control patients given induction therapy with low-dose RATG alone (gray histograms) (right panels), during the follow-up. Data from controls are mean ± SEM, P = NS.
Figure 5
Figure 5
IFNγ ELISpot assay against donor or third-party cells. Frequency of IFNγ-producing T cells after overnight incubation with donor (A) or third-party (B) cells of peripheral blood mononuclear cells isolated from mesenchymal stromal cell-treated patient #1 (blue circles), patient #2 (orange circles), patient #3 (green circles), and patient #4 (violet circles) and in control kidney transplant patients [white histograms; n = 6 given basiliximab/low-dose rabbit anti-thymocyte globulin (RATG) and n = 1 given only low-dose RATG induction therapy] during the follow-up. The dotted horizontal line represents the limit of test positivity (25 spots/300.000 responder cells). Data from controls are mean ± SEM; *P < 0.05 vs time 0.
Figure 6
Figure 6
Cell-mediated lympholysis against donor or third-party cells. Percentage of specific lysis of donor (A) or third-party (B) cells after 4 h incubation of 51Cr-labeled donor cells with peripheral blood mononuclear cells isolated from mesenchymal stromal cell-treated patient #1 (blue circles), patient #2 (orange circles), patient #3 (green circles), and patient #4 (violet circles) and in control kidney transplant patients [white histograms; n = 6 given basiliximab/low-dose rabbit anti-thymocyte globulin (RATG) and n = 1 given only low-dose RATG induction therapy] during the follow-up. Data from controls are mean ± SEM.
Figure 7
Figure 7
Profile of circulating NK cell subsets. (A) Profile of NK cell counts in mesenchymal stromal cell (MSC)-treated patients (n = 4), and kidney transplant patients undergoing induction therapy with bas/low dose rabbit anti-thymocyte globulin (RATG) (n = 6) or with low-dose RATG alone (n = 5) from 1 to 5 years posttransplant. Data are mean ± SEM. P = NS. (B) Gating strategy for identification of natural killer cell subsets and natural killer T cells. In the CD45+ live singlet cells, the population of CD3− lymphocytes was gated on the morphology based on SSC and plotted for CD16 and CD56 expression. CD16+CD56dim cells positive for CD11b expression and negative for CD27 were identified as cytotoxic NK cells. The CD56bright CD16−/+ NK cells were divided into CD27+ and CD27− subpopulations. NKT cells were defined as CD3+CD56+ cells. The frequency (% on the gated CD3− population) of CD56dim NK cells, CD56bright NK cells negative or positive for CD27 expression, and of NKT cells (% on CD3+ cells) in patients given MSC infusion and in kidney transplant patients undergoing induction therapy with bas/low dose RATG or with low-dose RATG alone from 1 to 5 years posttransplant are reported in panels (C–F), respectively. Data are mean ± SEM, *P < 0.05 vs MSC-treated patients and control patients given low-dose RATG at the same time point.
Figure 8
Figure 8
Profile of circulating monocytes. (A) Gating strategy for identification of classical, non-classical, and intermediate monocytes in peripheral blood mononuclear cells. In the CD45+ live singlet cells, the population of CD3− leukocytes was gated on the morphology based on SSC and plotted for CD14 expression. Classical monocytes were identified as CD64+ cells on gated CD33+HLADR+ cells in the CD14+CD16− cell population, intermediate monocytes as CD64+ cells on gated CD33+HLADR+ in the CD14+CD16+ cell population, and non-classical monocytes as CD64− cells on gated CD33+HLADR+ cells in the CD14−CD16+ cell population. Percentages of classical monocytes (% on the gated CD3− population) and their HLA-DR expression in patients given mesenchymal stromal cell (MSC) infusion (n = 4) and in kidney transplant patients undergoing induction therapy with bas/low dose rabbit anti-thymocyte globulin (RATG) (n = 6) or with low-dose RATG alone (n = 5) from 1 to 5 years posttransplant are reported in panels (B,C), respectively. Data are mean ± SEM, P = NS. Table in panel (D) reported mean ± SEM of intermediate and non classical monocytes; P = NS.
Figure 9
Figure 9
Histologic analysis of protocol kidney graft biopsy, profile of circulating B and T cell subsets, and immunologic functional assays of memory T cell response in patient #5, who received pretransplant infusion of mesenchymal stromal cell (MSC) in the setting of induction therapy with basiliximab/low-dose rabbit anti-thymocyte globulin (RATG). Patient #5, a 42-year-old woman with end-stage renal disease secondary to tubular interstitial nephropathy, received a kidney transplant from her sister, mismatched for two HLA alleles. She was given MSC before surgery (day −1) and induction therapy with the combination of basiliximab and low-dose RATG. A representative image of Gomori’s trichrome staining on 1-year protocol kidney graft biopsy (original magnification 100×) of MSC-treated patient #5 is shown (A). Percentages of circulating CD45RO+RA− memory CD8+ T cells (on CD3+CD8+ T cells) (B), percentages of circulating regulatory T cells (Tregs) (Foxp3+CD127− on CD4+CD25high T cells) (C), and ratio of the percentages of Tregs to CD45RO+RA− memory CD8+ T cells (D) in patient #5 (yellow circles) and in control kidney transplant recipients given basiliximab/low-dose RATG induction therapy but not MSC (white histograms) from baseline (pretransplant) to 1-year posttransplant. Number of circulating naïve IgD+CD27− B cells (E) and CD24highCD38high transitional B cells (F) in patient #5 (yellow circles) and in control kidney transplant recipients given basiliximab/low-dose RATG induction therapy (white histograms) from baseline to 1-year posttransplant. ELISpot for IFNγ (G) and CD8+ T cell function by T cell-mediated lympholysis [as percentage of specific lysis at 50:1 effector-target ratio (H)] toward donor or third-party antigens in patient #5 (yellow circles) and in control kidney transplant recipients given basiliximab/low-dose RATG induction therapy (white histograms) on peripheral blood mononuclear cells collected pretransplant and at 1 year after transplantation. Data are mean ± SEM.

References

    1. Lamb KE, Lodhi S, Meier-Kriesche HU. Long-term renal allograft survival in the United States: a critical reappraisal. Am J Transplant (2011) 11(3):450–62.10.1111/j.1600-6143.2010.03283.x
    1. Stoumpos S, Jardine AG, Mark PB. Cardiovascular morbidity and mortality after kidney transplantation. Transpl Int (2015) 28(1):10–21.10.1111/tri.12413
    1. Tufton N, Ahmad S, Rolfe C, Rajkariar R, Byrne C, Chowdhury TA. New-onset diabetes after renal transplantation. Diabet Med (2014) 31(11):1284–92.10.1111/dme.12534
    1. Salama AD, Womer KL, Sayegh MH. Clinical transplantation tolerance: many rivers to cross. J Immunol (2007) 178(9):5419–23.10.4049/jimmunol.178.9.5419
    1. Leventhal J, Abecassis M, Miller J, Gallon L, Ravindra K, Tollerud DJ, et al. Chimerism and tolerance without GVHD or engraftment syndrome in HLA-mismatched combined kidney and hematopoietic stem cell transplantation. Sci Transl Med (2012) 4(124):124ra28.10.1126/scitranslmed.3003509
    1. Leventhal JR, Mathew JM, Salomon DR, Kurian SM, Friedewald JJ, Gallon L, et al. Nonchimeric HLA-identical renal transplant tolerance: regulatory immunophenotypic/genomic biomarkers. Am J Transplant (2016) 16(1):221–34.10.1111/ajt.13416
    1. Scandling JD, Busque S, Shizuru JA, Lowsky R, Hoppe R, Dejbakhsh-Jones S, et al. Chimerism, graft survival, and withdrawal of immunosuppressive drugs in HLA matched and mismatched patients after living donor kidney and hematopoietic cell transplantation. Am J Transplant (2015) 15(3):695–704.10.1111/ajt.13091
    1. Kawai T, Sachs DH, Sprangers B, Spitzer TR, Saidman SL, Zorn E, et al. Long-term results in recipients of combined HLA-mismatched kidney and bone marrow transplantation without maintenance immunosuppression. Am J Transplant (2014) 14(7):1599–611.10.1111/ajt.12731
    1. Wood KJ, Bushell A, Hester J. Regulatory immune cells in transplantation. Nat Rev Immunol (2012) 12(6):417–30.10.1038/nri3227
    1. Casiraghi F, Perico N, Cortinovis M, Remuzzi G. Mesenchymal stromal cells in renal transplantation: opportunities and challenges. Nat Rev Nephrol (2016) 12(4):241–53.10.1038/nrneph.2016.7
    1. Perico N, Casiraghi F, Introna M, Gotti E, Todeschini M, Cavinato RA, et al. Autologous mesenchymal stromal cells and kidney transplantation: a pilot study of safety and clinical feasibility. Clin J Am Soc Nephrol (2011) 6(2):412–22.10.2215/CJN.04950610
    1. Perico N, Casiraghi F, Gotti E, Introna M, Todeschini M, Cavinato RA, et al. Mesenchymal stromal cells and kidney transplantation: pretransplant infusion protects from graft dysfunction while fostering immunoregulation. Transpl Int (2013) 26(9):867–78.10.1111/tri.12132
    1. Tan J, Wu W, Xu X, Liao L, Zheng F, Messinger S, et al. Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: a randomized controlled trial. JAMA (2012) 307(11):1169–77.10.1001/jama.2012.316
    1. Reinders ME, de Fijter JW, Roelofs H, Bajema IM, de Vries DK, Schaapherder AF, et al. Autologous bone marrow-derived mesenchymal stromal cells for the treatment of allograft rejection after renal transplantation: results of a phase I study. Stem Cells Transl Med (2013) 2(2):107–11.10.5966/sctm.2012-0114
    1. Peng Y, Ke M, Xu L, Liu L, Chen X, Xia W, et al. Donor-derived mesenchymal stem cells combined with low-dose tacrolimus prevent acute rejection after renal transplantation: a clinical pilot study. Transplantation (2013) 95(1):161–8.10.1097/TP.0b013e3182754c53
    1. Mudrabettu C, Kumar V, Rakha A, Yadav AK, Ramachandran R, Kanwar DB, et al. Safety and efficacy of autologous mesenchymal stromal cells transplantation in patients undergoing living donor kidney transplantation: a pilot study. Nephrology (Carlton) (2015) 20(1):25–33.10.1111/nep.12338
    1. Pan GH, Chen Z, Xu L, Zhu JH, Xiang P, Ma JJ, et al. Low-dose tacrolimus combined with donor-derived mesenchymal stem cells after renal transplantation: a prospective, non-randomized study. Oncotarget (2016) 7(11):12089–101.10.18632/oncotarget.7725
    1. Capelli C, Domenghini M, Borleri G, Bellavita P, Poma R, Carobbio A, et al. Human platelet lysate allows expansion and clinical grade production of mesenchymal stromal cells from small samples of bone marrow aspirates or marrow filter washouts. Bone Marrow Transplant (2007) 40(8):785–91.10.1038/sj.bmt.1705798
    1. Capelli C, Salvade A, Pedrini O, Barbui V, Gotti E, Borleri G, et al. The washouts of discarded bone marrow collection bags and filters are a very abundant source of hMSCs. Cytotherapy (2009) 11(4):403–13.10.1080/14653240902960437
    1. Ruggenenti P, Codreanu I, Cravedi P, Perna A, Gotti E, Remuzzi G. Basiliximab combined with low-dose rabbit anti-human thymocyte globulin: a possible further step toward effective and minimally toxic T cell-targeted therapy in kidney transplantation. Clin J Am Soc Nephrol (2006) 1(3):546–54.10.2215/CJN.01841105
    1. Gaspari F, Perico N, Matalone M, Signorini O, Azzollini N, Mister M, et al. Precision of plasma clearance of iohexol for estimation of GFR in patients with renal disease. J Am Soc Nephrol (1998) 9(2):310–3.
    1. Delanaye P, Melsom T, Ebert N, Back SE, Mariat C, Cavalier E, et al. Iohexol plasma clearance for measuring glomerular filtration rate in clinical practice and research: a review. Part 2: why to measure glomerular filtration rate with iohexol? Clin Kidney J (2016) 9(5):700–4.10.1093/ckj/sfw071
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy (2006) 8(4):315–7.10.1080/14653240600855905
    1. Noris M, Casiraghi F, Todeschini M, Cravedi P, Cugini D, Monteferrante G, et al. Regulatory T cells and T cell depletion: role of immunosuppressive drugs. J Am Soc Nephrol (2007) 18(3):1007–18.10.1681/ASN.2006101143
    1. Todeschini M, Cortinovis M, Perico N, Poli F, Innocente A, Cavinato RA, et al. In kidney transplant patients, alemtuzumab but not basiliximab/low-dose rabbit anti-thymocyte globulin induces B cell depletion and regeneration, which associates with a high incidence of de novo donor-specific anti-HLA antibody development. J Immunol (2013) 191(5):2818–28.10.4049/jimmunol.1203261
    1. Rakha A, Todeschini M, Casiraghi F. Assessment of anti-donor T cell proliferation and cytotoxic T lymphocyte-mediated lympholysis in living donor kidney transplant patients. Methods Mol Biol (2014) 1213:355–64.10.1007/978-1-4939-1453-1_29
    1. Pei R, Lee JH, Shih NJ, Chen M, Terasaki PI. Single human leukocyte antigen flow cytometry beads for accurate identification of human leukocyte antigen antibody specificities. Transplantation (2003) 75(1):43–9.10.1097/00007890-200301150-00008
    1. Klein U, Rajewsky K, Kuppers R. Human immunoglobulin (Ig)M+IgD+ peripheral blood B cells expressing the CD27 cell surface antigen carry somatically mutated variable region genes: CD27 as a general marker for somatically mutated (memory) B cells. J Exp Med (1998) 188(9):1679–89.10.1084/jem.188.9.1679
    1. Wei C, Anolik J, Cappione A, Zheng B, Pugh-Bernard A, Brooks J, et al. A new population of cells lacking expression of CD27 represents a notable component of the B cell memory compartment in systemic lupus erythematosus. J Immunol (2007) 178(10):6624–33.10.4049/jimmunol.178.10.6624
    1. Crespo M, Heidt S, Redondo D, Pascual J. Monitoring B cell subsets and alloreactivity in kidney transplantation. Transplant Rev (Orlando) (2015) 29(2):45–52.10.1016/j.trre.2015.02.001
    1. Augustine JJ, Siu DS, Clemente MJ, Schulak JA, Heeger PS, Hricik DE. Pre-transplant IFN-gamma ELISPOTs are associated with post-transplant renal function in African American renal transplant recipients. Am J Transplant (2005) 5(8):1971–5.10.1111/j.1600-6143.2005.00958.x
    1. Bestard O, Cruzado JM, Lucia M, Crespo E, Casis L, Sawitzki B, et al. Prospective assessment of antidonor cellular alloreactivity is a tool for guidance of immunosuppression in kidney transplantation. Kidney Int (2013) 84(6):1226–36.10.1038/ki.2013.236
    1. Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells (2006) 24(1):74–85.10.1634/stemcells.2004-0359
    1. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2, 3-dioxygenase and prostaglandin E2. Blood (2008) 111(3):1327–33.10.1182/blood-2007-02-074997
    1. Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells (2008) 26(1):212–22.10.1634/stemcells.2007-0554
    1. Freud AG, Mundy-Bosse BL, Yu J, Caligiuri MA. The broad spectrum of human natural killer cell diversity. Immunity (2017) 47(5):820–33.10.1016/j.immuni.2017.10.008
    1. Vossen MT, Matmati M, Hertoghs KM, Baars PA, Gent MR, Leclercq G, et al. CD27 defines phenotypically and functionally different human NK cell subsets. J Immunol (2008) 180(6):3739–45.10.4049/jimmunol.180.6.3739
    1. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, et al. Nomenclature of monocytes and dendritic cells in blood. Blood (2010) 116(16):e74–80.10.1182/blood-2010-02-258558
    1. Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: phenotypical vs. functional differentiation. Front Immunol (2014) 5:514.10.3389/fimmu.2014.00514
    1. Hudig D, Hunter KW, Diamond WJ, Redelman D. Properties of human blood monocytes. II. Monocytes from healthy adults are highly heterogeneous within and among individuals. Cytometry B Clin Cytom (2014) 86(2):121–34.10.1002/cyto.b.21141
    1. Du Rocher B, Mencalha AL, Gomes BE, Abdelhay E. Mesenchymal stromal cells impair the differentiation of CD14(++) CD16(-) CD64(+) classical monocytes into CD14(++) CD16(+) CD64(++) activate monocytes. Cytotherapy (2012) 14(1):12–25.10.3109/14653249.2011.594792
    1. de Witte SFH, Luk F, Sierra Parraga JM, Gargesha M, Merino A, Korevaar SS, et al. Immunomodulation by therapeutic mesenchymal stromal cells (MSC) is triggered through phagocytosis of MSC by monocytic cells. Stem Cells (2018) 36(4):602–15.10.1002/stem.2779
    1. Cortinovis M, Casiraghi F, Remuzzi G, Perico N. Mesenchymal stromal cells to control donor-specific memory T cells in solid organ transplantation. Curr Opin Organ Transplant (2015) 20(1):79–85.10.1097/MOT.0000000000000145
    1. Salama AD, Remuzzi G, Harmon WE, Sayegh MH. Challenges to achieving clinical transplantation tolerance. J Clin Invest (2001) 108(7):943–8.10.1172/JCI200114142
    1. Newell KA, Asare A, Kirk AD, Gisler TD, Bourcier K, Suthanthiran M, et al. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest (2010) 120(6):1836–47.10.1172/JCI39933
    1. Sagoo P, Perucha E, Sawitzki B, Tomiuk S, Stephens DA, Miqueu P, et al. Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans. J Clin Invest (2010) 120(6):1848–61.10.1172/JCI39922
    1. Chesneau M, Pallier A, Braza F, Lacombe G, Le Gallou S, Baron D, et al. Unique B cell differentiation profile in tolerant kidney transplant patients. Am J Transplant (2014) 14(1):144–55.10.1111/ajt.12508
    1. Newell KA, Asare A, Sanz I, Wei C, Rosenberg A, Gao Z, et al. Longitudinal studies of a B cell-derived signature of tolerance in renal transplant recipients. Am J Transplant (2015) 15(11):2908–20.10.1111/ajt.13480
    1. Chesneau M, Michel L, Dugast E, Chenouard A, Baron D, Pallier A, et al. Tolerant kidney transplant patients produce B cells with regulatory properties. J Am Soc Nephrol (2015) 26(10):2588–98.10.1681/ASN.2014040404
    1. Hidalgo LG, Campbell PM, Sis B, Einecke G, Mengel M, Chang J, et al. De novo donor-specific antibody at the time of kidney transplant biopsy associates with microvascular pathology and late graft failure. Am J Transplant (2009) 9(11):2532–41.10.1111/j.1600-6143.2009.02800.x
    1. Wiebe C, Gibson IW, Blydt-Hansen TD, Karpinski M, Ho J, Storsley LJ, et al. Evolution and clinical pathologic correlations of de novo donor-specific HLA antibody post kidney transplant. Am J Transplant (2012) 12(5):1157–67.10.1111/j.1600-6143.2012.04013.x
    1. Everly MJ, Rebellato LM, Haisch CE, Ozawa M, Parker K, Briley KP, et al. Incidence and impact of de novo donor-specific alloantibody in primary renal allografts. Transplantation (2013) 95(3):410–7.10.1097/TP.0b013e31827d62e3
    1. Ge W, Jiang J, Baroja ML, Arp J, Zassoko R, Liu W, et al. Infusion of mesenchymal stem cells and rapamycin synergize to attenuate alloimmune responses and promote cardiac allograft tolerance. Am J Transplant (2009) 9(8):1760–72.10.1111/j.1600-6143.2009.02721.x
    1. Franquesa M, Herrero E, Torras J, Ripoll E, Flaquer M, Goma M, et al. Mesenchymal stem cell therapy prevents interstitial fibrosis and tubular atrophy in a rat kidney allograft model. Stem Cells Dev (2012) 21(17):3125–35.10.1089/scd.2012.0096
    1. Comoli P, Ginevri F, Maccario R, Avanzini MA, Marconi M, Groff A, et al. Human mesenchymal stem cells inhibit antibody production induced in vitro by allostimulation. Nephrol Dial Transplant (2008) 23(4):1196–202.10.1093/ndt/gfm740
    1. Rostaing L, Hertig A, Albano L, Anglicheau D, Durrbach A, Vuiblet V, et al. Fibrosis progression according to epithelial-mesenchymal transition profile: a randomized trial of everolimus versus CsA. Am J Transplant (2015) 15(5):1303–12.10.1111/ajt.13132
    1. Liefeldt L, Brakemeier S, Glander P, Waiser J, Lachmann N, Schonemann C, et al. Donor-specific HLA antibodies in a cohort comparing everolimus with cyclosporine after kidney transplantation. Am J Transplant (2012) 12(5):1192–8.10.1111/j.1600-6143.2011.03961.x
    1. Abramowicz D, Del Carmen Rial M, Vitko S, del Castillo D, Manas D, Lao M, et al. Cyclosporine withdrawal from a mycophenolate mofetil-containing immunosuppressive regimen: results of a five-year, prospective, randomized study. J Am Soc Nephrol (2005) 16(7):2234–40.10.1681/ASN.2004100844
    1. Ekberg H, Grinyo J, Nashan B, Vanrenterghem Y, Vincenti F, Voulgari A, et al. Cyclosporine sparing with mycophenolate mofetil, daclizumab and corticosteroids in renal allograft recipients: the CAESAR Study. Am J Transplant (2007) 7(3):560–70.10.1111/j.1600-6143.2006.01645.x
    1. Ekberg H, Tedesco-Silva H, Demirbas A, Vitko S, Nashan B, Gurkan A, et al. Reduced exposure to calcineurin inhibitors in renal transplantation. N Engl J Med (2007) 357(25):2562–75.10.1056/NEJMoa067411
    1. Roodnat JI, Hilbrands LB, Hene RJ, de Sevaux RG, Smak Gregoor PJ, Kal-van Gestel JA, et al. 15-Year follow-up of a multicenter, randomized, calcineurin inhibitor withdrawal study in kidney transplantation. Transplantation (2014) 98(1):47–53.10.1097/01.TP.0000442774.46133.71
    1. Dugast E, Soulillou JP, Foucher Y, Papuchon E, Guerif P, Paul C, et al. Failure of calcineurin inhibitor (tacrolimus) weaning randomized trial in long-term stable kidney transplant recipients. Am J Transplant (2016) 16(11):3255–61.10.1111/ajt.13946
    1. Hricik DE, Formica RN, Nickerson P, Rush D, Fairchild RL, Poggio ED, et al. Adverse outcomes of tacrolimus withdrawal in immune-quiescent kidney transplant recipients. J Am Soc Nephrol (2015) 26(12):3114–22.10.1681/ASN.2014121234
    1. Ketterl N, Brachtl G, Schuh C, Bieback K, Schallmoser K, Reinisch A, et al. A robust potency assay highlights significant donor variation of human mesenchymal stem/progenitor cell immune modulatory capacity and extended radio-resistance. Stem Cell Res Ther (2015) 6:236.10.1186/s13287-015-0233-8
    1. Casiraghi F, Azzollini N, Todeschini M, Cavinato RA, Cassis P, Solini S, et al. Localization of mesenchymal stromal cells dictates their immune or proinflammatory effects in kidney transplantation. Am J Transplant (2012) 12(9):2373–83.10.1111/j.1600-6143.2012.04115.x
    1. Galleu A, Riffo-Vasquez Y, Trento C, Lomas C, Dolcetti L, Cheung TS, et al. Apoptosis in mesenchymal stromal cells induces in vivo recipient-mediated immunomodulation. Sci Transl Med (2017) 9:eaam7828.10.1126/scitranslmed.aam7828
    1. Detry O, Vandermeulen M, Delbouille MH, Somja J, Bletard N, Briquet A, et al. Infusion of mesenchymal stromal cells after deceased liver transplantation: a phase I-II, open-label, clinical study. J Hepatol (2017) 67(1):47–55.10.1016/j.jhep.2017.03.001

Source: PubMed

3
Abonneren