Sustained B cell depletion by CD19-targeted CAR T cells is a highly effective treatment for murine lupus

Rita Kansal, Noah Richardson, Indira Neeli, Saleem Khawaja, Damian Chamberlain, Marium Ghani, Qurat-Ul-Ain Ghani, Louisa Balazs, Sarka Beranova-Giorgianni, Francesco Giorgianni, James N Kochenderfer, Tony Marion, Lorraine M Albritton, Marko Radic, Rita Kansal, Noah Richardson, Indira Neeli, Saleem Khawaja, Damian Chamberlain, Marium Ghani, Qurat-Ul-Ain Ghani, Louisa Balazs, Sarka Beranova-Giorgianni, Francesco Giorgianni, James N Kochenderfer, Tony Marion, Lorraine M Albritton, Marko Radic

Abstract

The failure of anti-CD20 antibody (Rituximab) as therapy for lupus may be attributed to the transient and incomplete B cell depletion achieved in clinical trials. Here, using an alternative approach, we report that complete and sustained CD19+ B cell depletion is a highly effective therapy in lupus models. CD8+ T cells expressing CD19-targeted chimeric antigen receptors (CARs) persistently depleted CD19+ B cells, eliminated autoantibody production, reversed disease manifestations in target organs, and extended life spans well beyond normal in the (NZB × NZW) F1 and MRL fas/fas mouse models of lupus. CAR T cells were active for 1 year in vivo and were enriched in the CD44+CD62L+ T cell subset. Adoptively transferred splenic T cells from CAR T cell-treated mice depleted CD19+ B cells and reduced disease in naive autoimmune mice, indicating that disease control was cell-mediated. Sustained B cell depletion with CD19-targeted CAR T cell immunotherapy is a stable and effective strategy to treat murine lupus, and its effectiveness should be explored in clinical trials for lupus.

Conflict of interest statement

Competing interests: UTHSC filed a patent application, no. 65022–274929, titled “Use of Anti-CD19 CAR T Cells in Lupus.” (M.R., inventor). J.N.K. has patent applications for CARs targeting human CD19.

Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.

Figures

Fig. 1.. CAR design and transduction.
Fig. 1.. CAR design and transduction.
(A) The structural gene for 1D3–28Z.1–3 is shown downstream of the MSGV 5′ long terminal repeat (LTR) and splice acceptor (Ψ). The asterisk indicates that the first and third ITAMs in CD3ζ have tyrosine to alanine replacement mutations. (B) Diagram of CAR protein in the cell membrane showing the extracellular single-chain Fv (scFv) domain and the transmembrane (TM) and signaling domains that combine CD28 and CD3ζ C termini. (C) Steps involved in the isolation, activation, and transduction of CD8+ splenocytes with virus before infusion into a recipient mouse that was irradiated ahead of T cell transfer to achieve transient myeloablation. (D) Timeline of CAR T cell treatment and sample collection in NZB/W and MRL-lpr mice. CFSE, carboxyfluorescein diacetate succinimidyl ester.
Fig. 2.. Flow cytometry of blood lymphocytes…
Fig. 2.. Flow cytometry of blood lymphocytes from CAR T cell–treated mice and controls.
Mice were administered CD19-targeted CAR CD8+ T cells transduced by one of three different viruses, as indicated above the plots. CD19+ B cell depletion was assessed 2 months after treatment. Pseudocolor plots indicate CD19 and CD3 expression on lymphocytes, as defined by forward and side scatter in blood from individual mice. Frequencies of CD19+ B cells are shown in the top left quadrant. (A) Fourteen NZB/W mice in the three transduction groups that had less than 1% CD19+ lymphocytes were designated as CD19-d, whereas control mice and 15 of the CAR T cell–treated mice that had 12 to 60% CD19+ B cells were combined into the CD19-sufficient (CD19-s) experimental group. (B) MRL-lpr mice were injected with A-MLV–CAR–transduced CD8+ T cells or with nontransduced CD8+ T cells (control), and frequencies of CD19+ lymphocytes were determined. (C) Total RNA from the spleens of four control and four CAR T cell–treated mice was purified to measure transcripts coding for CD19 and TATA binding protein (TBP). CD19 message, as measured by reverse transcription polymerase chain reaction (RT-PCR) at two exon-intron boundaries (CD19-A or CD19-B; see table S2 for details), was determined in CAR T cell–treated mice and controls. n.d., nondetectable.
Fig. 3.. Serologic analysis of IgM and…
Fig. 3.. Serologic analysis of IgM and IgG concentrations and anti-DNA reactivities at different times after CAR T cell infusion.
(A) NZB/W and (B) MRL-lpr plasma were tested using ELISA for total IgM and IgG concentration and IgM and IgG anti-DNA titers. Values for CD19-d and CD19-s and control mice are indicated. IgM and IgG standards were used to generate standard curves for estimating IgM and IgG concentrations. Times of plasma collection for the relevant groups are indicated. Horizontal lines are means. Differences between measurements taken at the indicated time points were compared by one-tailed t test, and significance is indicated by *P < 0.05, **P < 0.01, and ***P < 0.001.
Fig. 4.. Impact of CD19-targeted CAR T…
Fig. 4.. Impact of CD19-targeted CAR T cell treatment on lupus pathogenesis and survival.
(A) Survival curves of NZB/W mice after CAR T cell infusion into 7-month-old recipients. Survival of CD19-d mice (n = 12) was compared relative to CD19-s mice (n = 12), and significance was determined by log rank (Mantel-Cox; P = 0.0019). For comparison, the survival of control mice (n = 8) is plotted. (B) Survival curves of CAR T cell-treated and control MRL-lpr mice. CAR T cell–treated mice (n = 11) were compared to control MRL-lpr mice (n = 11), and survival of mice in the two groups was evaluated by long rank (Mantel-Cox; P = 0.0001). (C and D) High-grade proteinuria (1 mg/ml and higher) was determined before treatment (start) and at 5 months after CAR T cell treatment. (C) Proteinuria in CD19-s (n = 12) and CD19-d (n = 11) NZB/W mice was analyzed by single-factor analysis of variance (ANOVA) (P = 0.038). (D) High-grade proteinuria in CAR T cell–treated MRL-lpr mice (n = 11) and controls (n = 9) was similarly compared (P = 0.0017). (E) Lengths of spleens in CD19-d and CD19-s NZB/W mice were measured at euthanasia (P < 0.005). Bars are means, and horizontal lines are SEMs. (F) Kidneys from NZB/W mice were sectioned and analyzed for cellularity and morphology by H&E staining and for mouse IgG deposits by immunofluorescence (IF). (G) MRL-lpr skin sections were stained by H&E to compare the epidermis of control and CAR T cell–treated mice. (H) Pathology scores of MRL-lpr kidney and skin sections were determined as described in Materials and Methods.
Fig. 5.. Effect of CD19-targeted CAR T…
Fig. 5.. Effect of CD19-targeted CAR T cell treatment on lymphocyte phenotype and plasma proteome.
(A) The expression of surface IgM on peripheral blood B lymphocytes from control MRL-lpr mice (left) was used to define an intermediate and a high IgM+ population (indicated by stacked gates). These gates were used to assess expression of IgM on B cells from CAR T cell–treated MRL-lpr mice (right). (B) The ratio of CD4 to CD8 T cells was determined in 18-week-old MRL-lpr mice treated with CAR T cells and controls. Each symbol represents data from a single mouse, bars are means, and horizontal lines are SDs. Differences between CAR T cell–treated and control mice were examined by two-tailed t test and noted by *P < 0.05 and ***P < 0.001. (C) Proportions of CD4−CD8− DN T cells as the percentage of CD3+ cells from CAR T cell–treated and control MRL-lpr mice were plotted (n. s., no significant differences). (D) Effector memory (CD62LloCD44hi), central memory (CD62LhiCD44hi), and naïve (CD62LhiCD44lo) CD4+ T cells from representative control (top) and CAR T cell–treated MRL-lpr mice (bottom) were determined (for complete CD4+ T cell phenotype assessment, see table S3). (E) The CD62LhiCD44hi population of CD8+ T cells from a representative control mouse (left) and CAR T cell–treated MRL-lpr mouse (right) are shown, and the percentage of CD8+ T cells in the CD62LhiCD44hi gates was displayed. (F) Overall proportions of CD62LhiCD44hi cells among CD8+ or CD4+ T cells in CAR T cell–treated and control mice were determined and plotted. Each symbol represents data from a single mouse, horizontal lines are means, and offset lines are SDs. Differences between CAR T cell–treated and control mice were examined by two-tailed t test and marked by ***P < 0.001. (G) Relative RNA expression in spleens, kidneys, and bone marrow from four CAR T cell–treated and four control NZB/W mice were measured by RT-PCR for transcripts originating from TBP, a house keeping gene, two regions of CD19, TACI, BCMA, CXCR4, and the light-chain constant regions of Igκ and λ1 (see Materials and Methods and table S2). Values were plotted using ln2. Means and SE are displayed. (H) Plasma was prepared from CAR T cell–treated and control MRL-lpr mice, and plasma proteins were analyzed by mass spectrometry. Horizontal lines are means, and offset lines are SDs. Concentrations were compared by two-tailed t test, and significance was indicated by *P < 0.05, **P < 0.01.
Fig. 6.. Tests of CAR T cell…
Fig. 6.. Tests of CAR T cell function in recipient mice.
(A) Injections of CFSE-labeled B cells into control or CAR T cell–treated MRL-lpr mice were performed 4 months after CAR T cell administration. Six days after injection into control or CAR T cell–treated mice, peripheral blood was assayed for labeled B cells by flow cytometry. (B and C) Adoptive transfer (AT) of purified splenic CD8+ T cells from an MRL-lpr mouse that was treated 7 months earlier with CD19-targeted CAR T cells into a second group of previously untreated, 2-month-old MRL-lpr mice. The secondary MRL-lpr recipients (MRL-lpr AT) and control MRL-lpr mice were assayed for CD19+ B cells (B) in peripheral blood 4 months after transfer and CD44hiCD62Lhi CD8+ T cells (C). The data in (B) and (C) are representative of four MRL-lpr AT mice observed in two experiments.

References

    1. Rahman A, Isenberg DA, Systemic lupus erythematosus. N. Engl. J. Med 358, 929–939 (2008).
    1. Shlomchik MJ, Activating systemic autoimmunity: B’s, T’s, and tolls. Curr. Opin. Immunol 21, 626–633 (2009).
    1. Sang A, Zheng Y-Y, Morel L, Contributions of B cells to lupus pathogenesis. Mol. Immunol 62, 329–338 (2014).
    1. Li Y, Chen F, Putt M, Koo YK, Madaio M, Cambier JC, Cohen PL, Eisenberg RA, B cell depletion with anti-CD79 mAbs ameliorates autoimmune disease in MRL/lpr mice. J. Immunol 181, 2961–2972 (2008).
    1. Gallagher S, Yusuf I, McCaughtry TM, Turman S, Sun H, Kolbeck R, Herbst R, Wang Y, MEDI-551 treatment effectively depletes B cells and reduces serum titers of autoantibodies in mice transgenic for Sle1 and human CD19. Arthritis Rheumatol 68, 965–976 (2016).
    1. Touma Z, Gladman DD, Current and future therapies for SLE: Obstacles and recommendations for the development of novel treatments. Lupus Sci. Med 4, e000239 (2017).
    1. Cohen SB, Emery P, Greenwald MW, Dougados M, Furie RA, Genovese MC, Keystone EC, Loveless JE, Burmester GR, Cravets MW, Hessey EW, Shaw T, Totoritis MC; REFLEX Trial Group, Rituximab for rheumatoid arthritis refractory to anti–tumor necrosis factor therapy: Results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks. Arthritis Rheum 54, 2793–2806 (2006).
    1. Calabresi PA, B-cell depletion — A frontier in monoclonal antibodies for multiple sclerosis. N. Engl. J. Med 376, 280–282 (2017).
    1. Merrill JT, Neuwelt CM, Wallace DJ, Shanahan JC, Latinis KM, Oates JC, Utset TO, Gordon C, Isenberg DA, Hsieh H-J, Zhang D, Brunetta PG, Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: The randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum 62, 222–233 (2010).
    1. Gomez Mendez LM, Cascino MD, Garg J, Katsumoto TR, Brakeman P, Dall’Era M, Looney RJ, Rovin B, Dragone L, Brunetta P, Peripheral blood B cell depletion after rituximab and complete response in lupus nephritis. Clin. J. Am. Soc. Nephrol 13, 1502–1509 (2018).
    1. Bekar KW, Owen T, Dunn R, Ichikawa T, Wang W, Wang R, Barnard J, Brady S, Nevarez S, Goldman BI, Kehry M, Anolik JH, Prolonged effects of short-term anti-CD20 B cell depletion therapy in murine systemic lupus erythematosus. Arthritis Rheum 62, 2443–2457 (2010).
    1. Ahuja A, Teichmann LL, Wang H, Dunn R, Kehry MR, Shlomchik MJ, An acquired defect in IgG-dependent phagocytosis explains the impairment in antibody-mediated cellular depletion in lupus. J. Immunol 187, 3888–3894 (2011).
    1. Ahuja A, Shupe J, Dunn R, Kashgarian M, Kehry MR, Shlomchik MJ, Depletion of B cells in murine lupus: Efficacy and resistance. J. Immunol 179, 3351–3361 (2007).
    1. Reddy V, Cambridge G, Isenberg DA, Glennie MJ, Cragg MS, Leandro M, Internalization of rituximab and the efficiency of B cell depletion in rheumatoid arthritis and systemic lupus erythematosus. Arthritis Rheumatol 67, 2046–2055 (2015).
    1. Looney RJ, Anolik JH, Campbell D, Felgar RE, Young F, Arend LJ, Sloand JA, Rosenblatt J, Sanz I, B cell depletion as a novel treatment for systemic lupus erythematosus: A phase I/II dose-escalation trial of rituximab. Arthritis Rheum 50, 2580–2589 (2004).
    1. Cooper LJN, Topp MS, Serrano LM, Gonzalez S, Chang W-C, Naranjo A, Wright C, Popplewell L, Raubitschek A, Forman SJ, Jensen MC, T-cell clones can be rendered specific for CD19: Toward the selective augmentation of the graft-versus-B–lineage leukemia effect. Blood 101, 1637–1644 (2003).
    1. Brentjens RJ, Latouche J-B, Santos E, Marti F, Gong MC, Lyddane C, King PD, Larson S, Weiss M, Rivière I, Sadelain M, Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat. Med 9, 279–286 (2003).
    1. Lim WA, June CH, The principles of engineering immune cells to treat cancer. Cell 168, 724–740 (2017).
    1. Theofilopoulos AN, Dixon FJ, Murine models of systemic lupus erythematosus. Adv. Immunol 37, 269–390 (1985).
    1. Koh D-R, Ho A, Rahemtulla A, Fung-Leung W-P, Griesser H, Mak T-W, Murine lupus in MRL/lpr mice lacking CD4 or CD8 T cells. Eur. J. Immunol 25, 2558–2562 (1995).
    1. Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA, Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood 116, 3875–3886 (2010).
    1. Zhao Y, Wang QJ, Yang S, Kochenderfer JN, Zheng Z, Zhong X, Sadelain M, Eshhar Z, Rosenberg SA, Morgan RA, A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. J. Immunol 183, 5563–5574 (2009).
    1. Bribes E, Galiegue S, Bourrie B, Casellas P, Involvement of the peripheral benzodiazepine receptor in the development of cutaneous pathology in Mrl/Lpr mice. Immunol. Lett 85, 13–18 (2003).
    1. Jevnikar AM, Grusby MJ, Glimcher LH, Prevention of nephritis in major histocompatibility complex class II-deficient MRL-lpr mice. J. Exp. Med 179, 1137–1143 (1994).
    1. Salle V, Cordonnier C, Schmidt J, Mazière C, Smail A, Attencourt C, Mabille MP, Mazière JC, Makdassi R, Choukroun G, Diouf M, Duhaut P, Ducroix JP, Vascular expression of annexin A2 in lupus nephritis. J. Clin. Pathol 69, 533–536 (2016).
    1. Phi NC, Chien DK, Binh VV, Gergely P, Cathepsin D-like activity in serum of patients with systemic lupus erythematosus. J. Clin. Lab. Immunol 29, 185–188 (1989).
    1. Roldan V, Marco P, Fernandez C, Pascual E, Levels of tissue factor pathway inhibitor in lupus patients correlate with lupus activity and endothelial damage markers. Haematologica 87, 1231–1232 (2002).
    1. Bao L, Haas M, Quigg RJ, Complement factor H deficiency accelerates development of lupus nephritis. J. Am. Soc. Nephrol 22, 285–295 (2011).
    1. You Y, Zhao H, Wang Y, Carter RH, Cutting edge: Primary and secondary effects of CD19 deficiency on cells of the marginal zone. J. Immunol 182, 7343–7347 (2009).
    1. Sato S, Miller AS, Howard MC, Tedder TF, Regulation of B lymphocyte development and activation by the CD19/CD21/CD81/Leu 13 complex requires the cytoplasmic domain of CD19. J. Immunol 159, 3278–3287 (1997).
    1. Wang Y, Brooks SR, Li X, Anzelon AN, Rickert RC, Carter RH, The physiologic role of CD19 cytoplasmic tyrosines. Immunity 17, 501–514 (2002).
    1. Wang Y, Carter RH, CD19 regulates B cell maturation, proliferation, and positive selection in the FDC zone of murine splenic germinal centers. Immunity 22, 749–761 (2005).
    1. Pracht K, Meinzinger J, Daum P, Schulz SR, Reimer D, Hauke M, Roth E, Mielenz D, Berek C, Côrte-Real J, Jäck HM, Schuh W, A new staining protocol for detection of murine antibody-secreting plasma cell subsets by flow cytometry. Eur. J. Immunol 47, 1389–1392 (2017).
    1. Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak Ö, Brogdon JL, Pruteanu-Malinici, Bhoj V, Landsburg D, Wasik M, Levine BL, Lacey SF, Melenhorst JJ, Porter DL, June CH, Chimeric antigen receptor T cells in refractory B-cell lymphomas. N. Engl. J. Med 377, 2545–2554 (2017).
    1. Bhoj VG, Arhontoulis D, Wertheim G, Capobianchi J, Callahan CA, Ellebrecht CT, Obstfeld AE, Lacey SF, Melenhorst JJ, Nazimuddin F, Hwang W-T, Maude SL, Wasik MA, Bagg A, Schuster S, Feldman MD, Porter DL, Grupp SA, June CH, Milone MC, Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood 128, 360–370 (2016).
    1. Hayakawa K, Hardy RR, Honda M, Herzenberg LA, Steinberg AD, Herzenberg LA, Ly-1 B cells: Functionally distinct lymphocytes that secrete IgM autoantibodies. Proc. Natl. Acad. Sci. U.S.A 81, 2494–2498 (1984).
    1. Marion TN, Bothwell AL, Briles DE, Janeway CA Jr., IgG anti-DNA autoantibodies within an individual autoimmune mouse are the products of clonal selection. J. Immunol 142, 4269–4274 (1989).
    1. Shlomchik M, Mascelli M, Shan H, Radic MZ, Pisetsky D, Marshak-Rothstein A, Weigert M, Anti-DNA antibodies from autoimmune mice arise by clonal expansion and somatic mutation. J. Exp. Med 171, 265–292 (1990).
    1. Hiepe F, Alexander T, Voll RE, Plasma cells. Z. Rheumatol 74, 20–25 (2015).
    1. Hale M, Rawlings DJ, Jackson SW, The long and the short of it: Insights into the cellular source of autoantibodies as revealed by B cell depletion therapy. Curr. Opin. Immunol 55, 81–88 (2018).
    1. Mahévas M, Michel M, Weill J-C, Reynaud C-A, Long-lived plasma cells in autoimmunity: Lessons from B-cell depleting therapy. Front. Immunol 4, 494 (2013).
    1. Espeli M, Bokers S, Giannico G, Dickinson HA, Bardsley V, Fogo AB, Smith KGC, Local renal autoantibody production in lupus nephritis. J. Am. Soc. Nephrol 22, 296–305 (2011).
    1. Davila ML, Kloss CC, Gunset G, Sadelain M, CD19 CAR-targeted T cells induce long-term remission and B cell aplasia in an immunocompetent mouse model of B cell acute lymphoblastic leukemia. PLOS ONE 8, e61338 (2013).
    1. Christensen SR, Shupe J, Nickerson K, Kashgarian M, Flavell RA, Shlomchik MJ, Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus. Immunity 25, 417–428 (2006).
    1. Krishnan MR, Wang C, Marion TN, Anti-DNA autoantibodies initiate experimental lupus nephritis by binding directly to the glomerular basement membrane in mice. Kidney Int 82, 184–192 (2012).
    1. Wu F, Zhang W, Shao H, Bo H, Shen H, Li J, Liu Y, Wang T, Ma W, Huang S, Human effector T cells derived from central memory cells rather than CD8+ T cells modified by tumor-specific TCR gene transfer possess superior traits for adoptive immunotherapy. Cancer Lett 339, 195–207 (2013).
    1. Gattinoni L, Speiser DE, Lichterfeld M, Bonini C, T memory stem cells in health and disease. Nat. Med 23, 18–27 (2017).
    1. Ellebrecht CT, Bhoj VG, Nace A, Choi EJ, Mao X, Cho MJ, Di Zenzo G, Lanzavecchia A, Seykora JT, Cotsarelis G, Milone MC, Payne AS, Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science 353, 179–184 (2016).
    1. Pegram HJ, Lee JC, Hayman EG, Imperato GH, Tedder TF, Sadelain M, Brentjens RJ, Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 119, 4133–4141 (2012).
    1. Le RQ, Li L, Yuan W, Shord SS, Nie L, Habtemariam BA, Przepiorka D, Farrell AT, Pazdur R, FDA approval summary: Tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome. Oncologist 23, 943–947 (2018).
    1. Paszkiewicz PJ, Fräßle SP, Srivastava S, Sommermeyer D, Hudecek M, Drexler I, Sadelain M, Liu L, Jensen MC, Riddell SR, Busch DH, Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. J. Clin. Invest 126, 4262–4272 (2016).
    1. Distler U, Kuharev J, Navarro P, Levin Y, Schild H, Tenzer S, Drift time-specific collision energies enable deep-coverage data-independent acquisition proteomics. Nat. Methods 11, 167–170 (2014).
    1. Sedic M, Gethings LA, Vissers JPC, Shockcor JP, McDonald S, Vasieva O, Lemac M, Langridge JI, Batinić D, Pavelić SK, Label-free mass spectrometric profiling of urinary proteins and metabolites from paediatric idiopathic nephrotic syndrome. Biochem. Biophys. Res. Commun 452, 21–26 (2014).
    1. Silva JC, Gorenstein MV, Li G-Z, Vissers JPC, Geromanos SJ, Absolute quantification of proteins by LCMSE: A virtue of parallel MS acquisition. Mol. Cell. Proteomics 5, 144–156 (2006).
    1. Singh RR, Saxena V, Zang S, Li L, Finkelman FD, Witte DP, Jacob CO, Differential contribution of IL-4 and STAT6 vs STAT4 to the development of lupus nephritis. J. Immunol 170, 4818–4825 (2003).
    1. Adelman NE, Watling DL, McDevitt HO, Treatment of (NZB x NZW)F1 disease with anti-I-A monoclonal antibodies. J. Exp. Med 158, 1350–1355 (1983).

Source: PubMed

3
Iratkozz fel