Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-t6hkb Total loading time: 0 Render date: 2024-07-08T11:50:47.227Z Has data issue: false hasContentIssue false

23 - Immune Privilege and Tolerance – Therapeutic Antibody Approaches

from PART IX - INNOVATIVE IMMUNOTHERAPEUTIC APPROACHES

Published online by Cambridge University Press:  15 December 2009

Get access

Summary

The discovery of monoclonal antibodies by Kohler and Milstein in 1975 sparked the generation of novel drugs that could be used to antagonize functional receptors of the immune system. The anti-CD3 antibody, OKT3, was the first of these drugs to be exploited clinically in the treatment of acute allograft rejection. Although the antibody was efficacious, neutralizing immunogenicity and, in particular, the often severe “flu-like” cytokine-release syndrome associated with initial doses of the antibody limited its application to other indications. As a consequence, the emergence of other immune-modulating CD3 or T cell-directed antibodies as therapeutics took a surprisingly long time. Three scientific developments rekindled interest in immune-modulating therapeutic antibodies resulting in many more antibody candidates entering clinical trials. The first development was the discovery that co-receptor CD4 antibodies could be used to tolerize to other proteins, thus establishing tolerance as a therapeutic paradigm. The second development was the discovery that rodent antibodies could be reengineered or reshaped to minimize their immunogenicity. Finally, the third development was the discovery that transplantation tolerance induced by co-receptor blockade was “dominant” and dependent on the induction of CD4+ regulatory T cells through so-called infectious tolerance. These findings together suggested that antibodies might be used sparingly to recruit the host's own tolerance mechanisms without evoking neutralizing responses.

Further studies in transplant models indicated that anti-CD4 therapeutic antibodies alone were insufficient when CD8+ T cells were also involved. In those circumstances, antagonism of CD8 function was also required.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Cosimi, A.B., et al., Treatment of acute renal allograft rejection with OKT3 monoclonal antibody. Transplantation, 1981. 32(6): pp. 535–9.CrossRefGoogle ScholarPubMed
Cosimi, A.B., et al., Use of monoclonal antibodies to T-cell subsets for immunologic monitoring and treatment in recipients of renal allografts. N Engl J Med, 1981. 305(6): pp. 308–14.CrossRefGoogle ScholarPubMed
Chatenoud, L., et al., Restriction of the human in vivo immune response against the mouse monoclonal antibody OKT3. J Immunol, 1986. 137(3): pp. 830–8.Google ScholarPubMed
Jaffers, G.J., et al., Monoclonal antibody therapy. Anti-idiotypic and non-anti-idiotypic antibodies to OKT3 arising despite intense immunosuppression. Transplantation, 1986. 41(5): pp. 572–8.CrossRefGoogle ScholarPubMed
Abramowicz, D., et al., Release of tumor necrosis factor, interleukin-2, and gamma-interferon in serum after injection of OKT3 monoclonal antibody in kidney transplant recipients. Transplantation, 1989. 47(4): pp. 606–8.CrossRefGoogle ScholarPubMed
Chatenoud, L., et al., Systemic reaction to the anti-T-cell monoclonal antibody OKT3 in relation to serum levels of tumor necrosis factor and interferon-gamma [corrected]. N Engl J Med, 1989. 320(21): pp. 1420–1.Google Scholar
Wofsy, D., et al., Inhibition of humoral immunity in vivo by monoclonal antibody to L3T4: studies with soluble antigens in intact mice. J Immunol, 1985. 135(3): pp. 1698–701.Google ScholarPubMed
Benjamin, R.J., et al., Tolerance to rat monoclonal antibodies. Implications for serotherapy. J Exp Med, 1986. 163(6): pp. 1539–52.CrossRefGoogle ScholarPubMed
Riechmann, L., et al., Reshaping human antibodies for therapy. Nature, 1988. 332(6162): pp. 323–7.CrossRefGoogle ScholarPubMed
Qin, S., et al., “Infectious” transplantation tolerance. Science, 1993. 259(5097): pp. 974–7.CrossRefGoogle ScholarPubMed
Cobbold, S.P., et al., Reprogramming the immune system for peripheral tolerance with CD4 and CD8 monoclonal antibodies. Immunol Rev, 1992. 129: pp. 165–201.CrossRefGoogle ScholarPubMed
Waldmann, H., et al., Infectious tolerance and the long-term acceptance of transplanted tissue. Immunol Rev, 2006. 212: pp. 301–13.CrossRefGoogle ScholarPubMed
Hutchings, P., et al., The use of a non-depleting anti-CD4 monoclonal antibody to re-establish tolerance to beta cells in NOD mice. Eur J Immunol, 1992. 22(7): pp. 1913–8.CrossRefGoogle ScholarPubMed
Cooke, A., Phillips, J.M., and Parish, N.M., Tolerogenic strategies to halt or prevent type 1 diabetes. Nat Immunol, 2001. 2(9): pp. 810–5.CrossRefGoogle ScholarPubMed
Chatenoud, L., et al., Anti-CD3 antibody induces long-term remission of overt autoimmunity in nonobese diabetic mice. Proc Natl Acad Sci USA, 1994. 91(1): pp. 123–7.CrossRefGoogle ScholarPubMed
Abbs, I.C., et al., Sparing of first dose effect of monovalent anti-CD3 antibody used in allograft rejection is associated with diminished release of pro-inflammatory cytokines. Ther Immunol, 1994. 1(6): pp. 325–31.Google ScholarPubMed
Routledge, E.G., et al., A humanized monovalent CD3 antibody which can activate homologous complement. Eur J Immunol, 1991. 21(11): pp. 2717–25.CrossRefGoogle ScholarPubMed
Routledge, E.G., et al., The effect of aglycosylation on the immunogenicity of a humanized therapeutic CD3 monoclonal antibody. Transplantation, 1995. 60(8): pp. 847–53.CrossRefGoogle ScholarPubMed
Friend, P.J., et al., Phase I study of an engineered aglycosylated humanized CD3 antibody in renal transplant rejection. Transplantation, 1999. 68(11): pp. 1632–7.CrossRefGoogle ScholarPubMed
Keymeulen, B., et al., Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N Engl J Med, 2005. 352(25): pp. 2598–608.CrossRefGoogle ScholarPubMed
Clark, M., et al., The improved lytic function and in vivo efficacy of monovalent monoclonal CD3 antibodies. Eur J Immunol, 1989. 19(2): pp. 381–8.CrossRefGoogle ScholarPubMed
Bolt, S., et al., The generation of a humanized, non-mitogenic CD3 monoclonal antibody which retains in vitro immunosuppressive properties. Eur J Immunol, 1993. 23(2): pp. 403–11.CrossRefGoogle ScholarPubMed
McMichael, A.J., Beverly, P.C.L., Cobbold, S., Crumpton, M.J., Gilks, W., Gotch, F.M., Hogg, N., Horton, M., Ling, N., MacLennan, I.C.M., Mason, D.Y., Milstein, C., Spiegelhalter, D., Waldmann, H., eds., Leukocyte Typing III: White Cell Differentiation Antigens, 1987, Oxford, UK: Oxford University Press.Google Scholar
Knapp, W., Dorken, B., Gilks, W.R., Rieber, E.P., Schmidt, R.E., Stein, H., dem Borne, A.E.G. Kr., eds., Leucocyte Typing IV: White Cell Differentiation Antigens, 1989, Oxford, UK: Oxford University Press.Google Scholar
Cobbold, S.P. and Waldmann, H., Therapeutic potential of monovalent monoclonal antibodies. Nature, 1984. 308(5958): pp. 460–2.CrossRefGoogle ScholarPubMed
Orlandi, R., et al., Cloning immunoglobulin variable domains for expression by the polymerase chain reaction. Proc Natl Acad Sci USA, 1989. 86(10): pp. 3833–7.CrossRefGoogle ScholarPubMed
Steen, M.L., Hellman, L., and Pettersson, U., The immunoglobulin lambda locus in rat consists of two C lambda genes and a single V lambda gene. Gene, 1987. 55(1): pp. 75–84.CrossRefGoogle Scholar
Kabat, E., Sequences of proteins of immunological interest. 4th ed, ed. N.I.o.H.U.D.o.R. Resources. 1987, Bethesda, MD: US Department of Health and Human Services, Public Health Service, National Institutes of Health.Google Scholar
Dersimonian, H., et al., Relationship of human variable region heavy chain germ-line genes to genes encoding anti-DNA autoantibodies. J Immunol, 1987. 139(7): pp. 2496–501.Google ScholarPubMed
Takahashi, N., et al., Structure of human immunoglobulin gamma genes: implications for evolution of a gene family. Cell, 1982. 29(2): pp. 671–9.CrossRefGoogle ScholarPubMed
Rabbitts, T.H., Forster, A., and Matthews, J.G., The breakpoint of the Philadelphia chromosome 22 in chronic myeloid leukaemia is distal to the immunoglobulin lambda light chain constant region genes. Mol Biol Med, 1983. 1(1): pp. 11–9.Google ScholarPubMed
Smith, J.A., et al., Nonmitogenic anti-CD3 monoclonal antibodies deliver a partial T cell receptor signal and induce clonal anergy. J Exp Med, 1997. 185(8): pp. 1413–22.CrossRefGoogle ScholarPubMed
Yu, X.Z., et al., Lck is required for activation-induced T cell death after TCR ligation with partial agonists. J Immunol, 2004. 172(3): pp. 1437–43.CrossRefGoogle ScholarPubMed
Tax, W.J., et al., Fc receptors for mouse IgG1 on human monocytes: polymorphism and role in antibody-induced T cell proliferation. J Immunol, 1984. 133(3): pp. 1185–9.Google ScholarPubMed
Clement, L.T., Tilden, A.B., and Dunlap, N.E., Analysis of the monocyte Fc receptors and antibody-mediated cellular interactions required for the induction of T cell proliferation by anti-T3 antibodies. J Immunol, 1985. 135(1): pp. 165–71.Google ScholarPubMed
Anasetti, C., et al., Induction of specific nonresponsiveness in unprimed human T cells by anti-CD3 antibody and alloantigen. J Exp Med, 1990. 172(6): pp. 1691–700.CrossRefGoogle Scholar
Baecher-Allan, C. and Hafler, D.A., Human regulatory T cells and their role in autoimmune disease. Immunol Rev, 2006. 212: pp. 203–16.CrossRefGoogle ScholarPubMed
Takahashi, T., et al., Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int Immunol, 1998. 10(12): pp. 1969–80.CrossRefGoogle ScholarPubMed
Thornton, A.M. and Shevach, E.M., CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med, 1998. 188(2): pp. 287–96.CrossRefGoogle ScholarPubMed
Chen, W., et al., Conversion of peripheral CD4+CD25-naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med, 2003. 198(12): pp. 1875–86.CrossRefGoogle ScholarPubMed
Cobbold, S.P., et al., Induction of foxP3+ regulatory T cells in the periphery of T cell receptor transgenic mice tolerized to transplants. J Immunol, 2004. 172(10): pp. 6003–10.CrossRefGoogle ScholarPubMed
Lindley, S., et al., Defective suppressor function in CD4(+)CD25(+) T-cells from patients with type 1 diabetes. Diabetes, 2005. 54(1): pp. 92–9.CrossRefGoogle ScholarPubMed
Belghith, M., et al., TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med, 2003. 9(9): pp. 1202–8.CrossRefGoogle ScholarPubMed
Woodberry, T., et al., Differential targeting and shifts in the immunodominance of Epstein-Barr virus–specific CD8 and CD4 T cell responses during acute and persistent infection. J Infect Dis, 2005. 192(9): pp. 1513–24.CrossRefGoogle ScholarPubMed
Ferran, C., et al., Cytokine-related syndrome following injection of anti-CD3 monoclonal antibody: further evidence for transient in vivo T cell activation. Eur J Immunol, 1990. 20(3): pp. 509–15.CrossRefGoogle ScholarPubMed
Alegre, M., et al., Hypothermia and hypoglycemia induced by anti-CD3 monoclonal antibody in mice: role of tumor necrosis factor. Eur J Immunol, 1990. 20(3): pp. 707–10.CrossRefGoogle ScholarPubMed
Ferran, C., et al., Cascade modulation by anti-tumor necrosis factor monoclonal antibody of interferon-gamma, interleukin 3 and interleukin 6 release after triggering of the CD3/T cell receptor activation pathway. Eur J Immunol, 1991. 21(10): pp. 2349–53.CrossRefGoogle ScholarPubMed
Chamberlain, P., Immunogenicity of therapeutic proteins. Regulatory Review, 2002. 5(5): pp. 4–9.Google Scholar
Amin, T.C., , G., Immunogenicity issues with therapeutic proteins. Current Drug Discovery, 2004: pp. 20–24.Google Scholar
Alegre, M.L., et al., A non-activating “humanized” anti-CD3 monoclonal antibody retains immunosuppressive properties in vivo. Transplantation, 1994. 57(11): pp. 1537–43.CrossRefGoogle ScholarPubMed
Alegre, M.L., et al., Effect of a single amino acid mutation on the activating and immunosuppressive properties of a “humanized” OKT3 monoclonal antibody. J Immunol, 1992. 148(11): pp. 3461–8.Google ScholarPubMed
Cole, M.S., Anasetti, C., and Tso, J.Y., Human IgG2 variants of chimeric anti-CD3 are nonmitogenic to T cells. J Immunol, 1997. 159(7): pp. 3613–21.Google ScholarPubMed
Cole, M.S., et al., HuM291, a humanized anti-CD3 antibody, is immunosuppressive to T cells while exhibiting reduced mitogenicity in vitro. Transplantation, 1999. 68(4): pp. 563–71.CrossRefGoogle Scholar
Alegre, M.L., et al., An anti-murine CD3 monoclonal antibody with a low affinity for Fc gamma receptors suppresses transplantation responses while minimizing acute toxicity and immunogenicity. J Immunol, 1995. 155(3): pp. 1544–55.Google ScholarPubMed
Hughes, C., et al., Induction of T helper cell hyporesponsiveness in an experimental model of autoimmunity by using nonmitogenic anti-CD3 monoclonal antibody. J Immunol, 1994. 153(7): pp. 3319–25.Google Scholar
Herold, K.C., et al., Prevention of autoimmune diabetes with nonactivating anti-CD3 monoclonal antibody. Diabetes, 1992. 41(3): pp. 385–91.CrossRefGoogle ScholarPubMed
Parlevliet, K.J., et al., In vivo effects of IgA and IgG2a anti-CD3 isotype switch variants. J Clin Invest, 1994. 93(6): pp. 2519–25.CrossRefGoogle ScholarPubMed
Hirsch, R., et al., Anti-CD3 F(ab′)2 fragments are immunosuppressive in vivo without evoking either the strong humoral response or morbidity associated with whole mAb. Transplantation, 1990. 49(6): pp. 1117–23.CrossRefGoogle ScholarPubMed
Vossen, A.C., et al., Fc receptor binding of anti-CD3 monoclonal antibodies is not essential for immunosuppression, but triggers cytokine-related side effects. Eur J Immunol, 1995. 25(6): pp. 1492–6.CrossRefGoogle Scholar
Carpenter, P.A., et al., Non-Fc receptor-binding humanized anti-CD3 antibodies induce apoptosis of activated human T cells. J Immunol, 2000. 165(11): pp. 6205–13.CrossRefGoogle ScholarPubMed
Smith, J.A., Tang, Q., and Bluestone, J.A., Partial TCR signals delivered by FcR-nonbinding anti-CD3 monoclonal antibodies differentially regulate individual Th subsets. J Immunol, 1998. 160(10): pp. 4841–9.Google ScholarPubMed
Woodle, E.S., Hussein, S., and Bluestone, J.A., In vivo administration of anti-murine CD3 monoclonal antibody induces selective, long-term anergy in CD8+ T cells. Transplantation, 1996. 61(5): pp. 798–803.CrossRefGoogle ScholarPubMed
Hirsch, R., Archibald, J., and Gress, R.E., Differential T cell hyporesponsiveness induced by in vivo administration of intact or F(ab′)2 fragments of anti-CD3 monoclonal antibody. F(ab′)2 fragments induce a selective T helper dysfunction. J Immunol, 1991. 147(7): pp. 2088–93.Google ScholarPubMed
Yu, X.Z., et al., Induction of apoptosis by anti-CD3 epsilon F(ab′)2 in antigen receptor transgenic murine T cells activated by specific peptide. J Immunol, 1996. 157(8): pp. 3420–9.Google ScholarPubMed
Yu, X.Z. and Anasetti, C., Enhancement of susceptibility to Fas-mediated apoptosis of TH1 cells by nonmitogenic anti-CD3epsilon F(ab′)2. Transplantation, 2000. 69(1): pp. 104–12.CrossRefGoogle ScholarPubMed
Popma, S.H., Griswold, D.E., and Li, L., Anti-CD3 antibodies OKT3 and hOKT3gamma1(Ala-Ala) induce proliferation of T cells but impair expansion of alloreactive T cells; aspecifc T cell proliferation induced by anti-CD3 antibodies correlates with impaired expansion of alloreactive T cells. Int Immunopharmacol, 2005. 5(1): pp. 155–62.CrossRefGoogle ScholarPubMed
Yu, X.Z., et al., Anti-CD3 epsilon F(ab′)2 prevents graft-versus-host disease by selectively depleting donor T cells activated by recipient alloantigens. J Immunol, 2001. 166(9): pp. 5835–9.CrossRefGoogle ScholarPubMed
Cao, O., et al., Induction and role of regulatory CD4+CD25+ T cells in tolerance to the transgene product following hepatic in vivo gene transfer. Blood, 2007. 110(4): pp. 1132–40.CrossRefGoogle ScholarPubMed
Chatenoud, L., CD3-specific antibody-induced active tolerance: from bench to bedside. Nat Rev Immunol, 2003. 3(2): pp. 123–32.CrossRefGoogle ScholarPubMed
Peng, Y., et al., TGF-beta regulates in vivo expansion of Foxp3-expressing CD4+CD25+ regulatory T cells responsible for protection against diabetes. Proc Natl Acad Sci USA, 2004. 101(13): pp. 4572–7.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure [email protected] is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×