51
|
Tan HP, Donaldson J, Basu A, Unruh M, Randhawa P, Sharma V, Morgan C, McCauley J, Wu C, Shah N, Zeevi A, Shapiro R. Two hundred living donor kidney transplantations under alemtuzumab induction and tacrolimus monotherapy: 3-year follow-up. Am J Transplant 2009; 9:355-66. [PMID: 19120078 DOI: 10.1111/j.1600-6143.2008.02492.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alemtuzumab has been used in off-label studies of solid organ transplantation. We extend our report of the first 200 consecutive living donor solitary kidney transplantations under alemtuzumab pretreatment with tacrolimus monotherapy and subsequent spaced weaning to 3 years of follow-up. We focused especially on the causes of recipient death and graft loss, and the characteristics of rejection. The actuarial 1-, 2- and 3-year patient and graft survivals were 99.0% and 98.0%, 96.4% and 90.8% and 93.3% and 86.3%, respectively. The cumulative incidence of acute cellular rejection (ACR) at the following months was 2%</=6, 9.0%</=12, 16.5%</=18, 19.5%</=24, 23.5%</=30, 24.0%</=36 and 25%</=42. The mean serum creatinine (mg/dL) and glomerular filtration rate (mL/min/1.73 m(2)) at 1 and 3 years were 1.4 +/- 0.6 and 58.7 +/- 21.6 and 1.5 +/- 0.7 and 54.9 +/- 20.9, respectively. Fifty (25%) recipients had a total of 89 episodes of ACR. About 88.7% of ACR episodes were Banff 1, and of those, 82% were steroid-sensitive. Nine (4.5%) recipients had antibody-mediated rejection (AMR). About 76.5% were weaned but only 46% are currently on spaced dose (qod or less) tacrolimus monotherapy, and 94.4% remained steroid-free from the time of transplantation. Infectious complications were uncommon. This experience suggests the 3-year efficacy of this approach.
Collapse
Affiliation(s)
- H P Tan
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Pediatric Living Donor Kidney Transplantation Under Alemtuzumab Pretreatment and Tacrolimus Monotherapy: 4-Year Experience. Transplantation 2008; 86:1725-31. [DOI: 10.1097/tp.0b013e3181903da7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
53
|
Ochi H, Abraham M, Ishikawa H, Frenkel D, Yang K, Basso A, Wu H, Chen ML, Gandhi R, Miller A, Maron R, Weiner HL. New immunosuppressive approaches: oral administration of CD3-specific antibody to treat autoimmunity. J Neurol Sci 2008; 274:9-12. [PMID: 18804221 DOI: 10.1016/j.jns.2008.07.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 07/10/2008] [Accepted: 07/24/2008] [Indexed: 01/12/2023]
Abstract
One of the major goals for the immunotherapy of autoimmune diseases is the induction of regulatory T cells that mediate immunologic tolerance. Parenteral administration of anti-CD3 monoclonal antibody is an approved therapy for transplantation in humans and is effective in autoimmune diabetes. We have found that oral administration of anti-CD3 monoclonal antibody is biologically active in the gut and suppresses experimental autoimmune encephalomyelitis both prior to disease induction and at the height of disease. Oral anti-CD3 antibody acts by inducing a unique type of regulatory T cell characterized by latency-associated peptide (LAP) on its cell surface that functions in vivo and in vitro via TGF-beta dependent mechanism. Orally delivered antibody would not have side effects including cytokine release syndromes, thus oral anti-CD3 antibody is clinically applicable for chronic therapy. These findings identify a novel and powerful immunologic approach that is widely applicable for the treatment of human autoimmune conditions.
Collapse
Affiliation(s)
- Hirofumi Ochi
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Isaacs JD. Therapeutic T-cell manipulation in rheumatoid arthritis: past, present and future. Rheumatology (Oxford) 2008; 47:1461-8. [PMID: 18503092 DOI: 10.1093/rheumatology/ken163] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Accumulating evidence suggests that RA is a T-cell-mediated autoimmune disease. Early attempts at disease modulation using strategies such as CD4 mAbs were severely hampered by a lack of biomarkers of autoreactivity. Recently, however, co-stimulation blockade has emerged as an effective treatment for RA. Alongside a greatly improved mechanistic understanding of immune regulation, this has rekindled hopes for authentic and robust immune programming. The final pieces of the jigsaw are not yet in place for RA but, in other disciplines, emerging treatment paradigms such as non-mitogenic anti-CD3 mAbs, autoantigenic peptides and even cellular therapies are providing hope for a future in which immunopathology can be specifically and vigorously curtailed.
Collapse
Affiliation(s)
- J D Isaacs
- Musculoskeletal Research Group and Wilson Horne Immunotherapy Centre, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, UK.
| |
Collapse
|
55
|
Essa S, Pacsa A, Said T, Nampoory MRN, Raghupathy R, Johny KV, Al-Nakib W, Al-Mosawy M. Is combined pretransplantation seropositivity of kidney transplant recipients for cytomegalovirus antigens (pp150 and pp28) a predictor for protection against infection? Med Princ Pract 2008; 17:66-70. [PMID: 18059104 DOI: 10.1159/000109593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study was aimed at detecting antibodies to the antigens which may contribute to protection against cytomegalovirus (CMV) infection after organ transplantation. MATERIALS AND METHODS A total of 203 kidney transplant patients were enrolled in the study. Based on CMV antigenemia assay, 23 patients were antigen-positive and of the remaining 180 antigen-negative patients, 46 were selected as controls matched for age, gender and source of kidney. The 69 kidney recipients (KR) had CMV antibody due to previous infection and were followed up for a period of 6 months after transplantation for the development of active CMV infections by the antigenemia assay. Antibody responses to five CMV-related peptide antigens (pp65, gB, pp150, pp28 and pp38) were investigated by enzyme immunoassay and their presence was correlated with the results of the CMV antigenemia assay. RESULTS Of the five CMV-related peptide antigens, only gB antigen showed response to the antibody in 10/23 (43.5%) antigen-positive patients and 9/46 antigen-negative patients and the difference was statistically significant (p = 0.048). On the other hand, there was no significant difference in antibody responses between the antigen-positive and antigen-negative KR to the other four CMV peptide antigens (p > 0.05). However, among the antigen-positive KR there was only 1 patient who had antibodies to both pp150 and pp28 antigen, while among the antigen-negative KR, 22 of 46 (47.8%) had the antibodies (p < 0.001). CONCLUSION The findings suggest that the combined presence of antibodies against the pp150 and pp28 antigens may indicate a lower risk of CMV reactivation after kidney transplantation.
Collapse
Affiliation(s)
- S Essa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait.
| | | | | | | | | | | | | | | |
Collapse
|
56
|
Chatenoud L. The use of CD3-specific antibodies in autoimmune diabetes: a step toward the induction of immune tolerance in the clinic. Handb Exp Pharmacol 2008:221-36. [PMID: 18071948 DOI: 10.1007/978-3-540-73259-4_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD3-specific monoclonal antibodies were the first rodent monoclonals introduced in clinical practice in the mid 1980s as approved immunosuppressants to prevent and treat organ allograft rejection. Since then compelling evidence has been accumulated to suggest that in addition to their immunosuppressive properties, CD3-specific antibodies can also afford inducing immune tolerance especially in the context of ongoing immune responses. Thus, they are highly effective at restoring self-tolerance in overt autoimmunity, a capacity first demonstrated in the experimental setting, which was recently transferred to the clinic with success.
Collapse
|
57
|
You S, Candon S, Kuhn C, Bach JF, Chatenoud L. Chapter 2 CD3 Antibodies as Unique Tools to Restore Self-Tolerance in Established Autoimmunity. Adv Immunol 2008; 100:13-37. [DOI: 10.1016/s0065-2776(08)00802-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
58
|
History of Clinical Transplantation. Surgery 2008. [DOI: 10.1007/978-0-387-68113-9_80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
59
|
Czechowicz A, Kraft D, Weissman IL, Bhattacharya D. Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches. Science 2007; 318:1296-9. [PMID: 18033883 DOI: 10.1126/science.1149726] [Citation(s) in RCA: 307] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Upon intravenous transplantation, hematopoietic stem cells (HSCs) can home to specialized niches, yet most HSCs fail to engraft unless recipients are subjected to toxic preconditioning. We provide evidence that, aside from immune barriers, donor HSC engraftment is restricted by occupancy of appropriate niches by host HSCs. Administration of ACK2, an antibody that blocks c-kit function, led to the transient removal of >98% of endogenous HSCs in immunodeficient mice. Subsequent transplantation of these mice with donor HSCs led to chimerism levels of up to 90%. Extrapolation of these methods to humans may enable mild but effective conditioning regimens for transplantation.
Collapse
Affiliation(s)
- Agnieszka Czechowicz
- Institute of Stem Cell Biology and Regenerative Medicine, Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
60
|
Chatenoud L. Une étape vers la restauration de la tolérance immunitaire au soi dans les maladies auto-immunes humaines. Med Sci (Paris) 2007; 23:167-71. [PMID: 17291426 DOI: 10.1051/medsci/2007232167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In developed countries the incidence of autoimmune insulin-dependent or type 1 diabetes as the one of all autoimmune diseases has steadily increased over the last decades. Conventional therapy of type 1 diabetes is essentially palliative namely, chronic delivery of exogenous insulin that is associated with major constraints (multiple daily parenteral administration, serious risks linked to hypoglycemic episodes) and incomplete effectiveness in preventing severe degenerative complications. This explains the growing attention on modern therapeutic strategies using biological agents such as CD3 monoclonal antibodies that allow 'reprogramming' the immune system to restore self-tolerance to pancreatic beta cell antigens. This strategy which proved successful in the experimental setting has recently been translated to the clinic with very encouraging results. CD3 antibodies may represent a new category of drugs affording a real cure for autoimmunity namely, inhibiting the pathogenic immune response while preserving the host reactivity to unrelated antigens.
Collapse
MESH Headings
- Adult
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Autoantibodies/biosynthesis
- Autoantibodies/immunology
- Autoantigens/immunology
- Autoantigens/therapeutic use
- Autoimmune Diseases/immunology
- Autoimmune Diseases/therapy
- Autoimmunity/immunology
- CD3 Complex/immunology
- Chaperonin 60/immunology
- Chaperonin 60/therapeutic use
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Clonal Deletion
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/therapy
- Glutamate Decarboxylase/immunology
- Glutamate Decarboxylase/therapeutic use
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/therapeutic use
- Immunotherapy/methods
- Infant, Newborn
- Insulin/immunology
- Insulin/therapeutic use
- Islets of Langerhans/immunology
- Mice
- Mice, Inbred NOD
- Muromonab-CD3/immunology
- Muromonab-CD3/therapeutic use
- Randomized Controlled Trials as Topic
- Self Tolerance/immunology
- T-Lymphocyte Subsets/immunology
- Thymectomy/adverse effects
Collapse
Affiliation(s)
- Lucienne Chatenoud
- Inserm U580, Faculté René-Descartes Paris 5, Hôpital Necker- Enfants-Malades, 161, rue de Sèvres 75015, Paris, France.
| |
Collapse
|
61
|
Liu EH, Siegel RM, Harlan DM, O'Shea JJ. T cell–directed therapies: lessons learned and future prospects. Nat Immunol 2007; 8:25-30. [PMID: 17179969 DOI: 10.1038/ni1429] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Agents interfering with T cell function are therapeutic mainstays for various autoimmune diseases and for transplant approaches to organ failure. The understanding of T cell biology has blossomed since the development of most agents now in use. Here we discuss T cell-specific agents now in use, others recently added to the therapeutic armamentarium and promising agents being investigated in clinical and preclinical studies. In addition, we reflect on the risks and benefits involved in the testing of such agents clinically, with examples of agents that have successfully been used in the clinic and agents that failed to reach therapeutic use.
Collapse
Affiliation(s)
- Eric H Liu
- Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
62
|
Abstract
Less than ten years after the discovery of hybridomas by Kohler and Milstein in 1975, the first monoclonal antibody was injected in humans to prevent graft rejection. The antibodies used were targeted against the CD3 molecule involved in the transduction of T cell signals after recognition of the antigen. Paradoxically, because of pharmaceutical hesitations later found to be unwarranted, the therapeutic class developed slowly. It was not until the 1990s that other formulations emerged. In parallel, work was undertaken to modify the antibodies using genetic engineering techniques to humanize the molecules by changing the majority of the species determinants from the murine producing animals to the human recipients. It was even found possible to produce totally human antibodies using several different strategies based on in vitro phage display. This led to rapid development of new antibodies used for a wide range of indications: organ transplantation (anti-CD2, anti-CD4, antiCD25, antiCD40L), as well as anti-respirator syncytial virus (RSV) for respiratory infections, and anti cancer agents (Anti-CD20 antibodies and anti-tumor antibodies), as well as autoimmune diseases (with significant success with anti-TNF antibodies). It was found the monoclonal antibodies offer an exceptionally effective method in domains where conventional compounds have reached their limit. This specific action of monoclonal antibodies, with fine specificity for the targets (which can be varied to infinity), as well as their capacity to provide positive signals to cells whose functions are not often inhibited by small molecules. An illustration of this mode of action is the effect recently demonstrated by our team on restored self tolerance using anti-CD3 antibodies in newly diagnosed diabetics in whom sustained remission can be injected after less than one week's treatment.
Collapse
Affiliation(s)
- J-F Bach
- Inserm U580, Hôpital Necker-Enfants Malades, 161, rue de Sèvres, F 75015 Paris.
| |
Collapse
|
63
|
Steffens S, Burger F, Pelli G, Dean Y, Elson G, Kosco-Vilbois M, Chatenoud L, Mach F. Short-Term Treatment With Anti-CD3 Antibody Reduces the Development and Progression of Atherosclerosis in Mice. Circulation 2006; 114:1977-84. [PMID: 17043169 DOI: 10.1161/circulationaha.106.627430] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Atherosclerosis is a chronic inflammatory disease of the large arteries that is the primary cause of heart disease and stroke. Anti-CD3–specific antibodies suppress immune responses by antigenic modulation of the CD3 antibody/T-cell receptor complex. Their unique capacity to restore self-tolerance in a mouse model of diabetes and, importantly, in patients with recent-onset type 1 diabetes involves transforming growth factor-β–dependent mechanisms via expansion and/or activation of regulatory T cells. We hypothesized that treatment with anti-CD3–specific antibodies might inhibit atherosclerosis development and progression in mice.
Methods and Results—
Low-density lipoprotein receptor–deficient mice were fed a high-cholesterol diet for 13 or 24 weeks. Anti-CD3 antibody was administered on 5 consecutive days beginning 1 week before or 13 weeks after the high-cholesterol diet was initiated, respectively. Control mice were injected in parallel with phosphate-buffered saline. Anti-CD3 antibody therapy reduced plaque development when administered before a high-cholesterol diet and markedly decreased lesion progression in mice with already established atherosclerosis. We found increased production of the antiinflammatory cytokine transforming growth factor-β in concanavalin A–stimulated lymph node cells and enhanced expression of the regulatory T-cell marker Foxp3 in spleens of anti-CD3 antibody–treated mice. A higher percentage of apoptotic cells within the plaques of anti-CD3 antibody–treated mice was also observed.
Conclusions—
Altered disease progression, combined with the emergence of this particular cytokine pattern, indicates that short-term treatment with an anti-CD3 antibody induces a regulatory T-cell phenotype that restores self-tolerance in a mouse model of atherosclerosis.
Collapse
Affiliation(s)
- Sabine Steffens
- Division of Cardiology, Department of Medicine, University Hospital, Foundation for Medical Researches, 64 Avenue Roseraie, 1211 Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Chatenoud L. CD3-specific antibodies as promising tools to aim at immune tolerance in the clinic. Int Rev Immunol 2006; 25:215-33. [PMID: 16818372 DOI: 10.1080/08830180600743032] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Currently, therapies applied in transplantation and autoimmunity are essentially based on the use of immunosuppressants. These agents depress all immune responses and expose individuals to the recurrence of the pathogenic immune process once they are withdrawn, thus necessitating a chronic administration leading to the risk of recurrent infections and increased frequency of tumors. At variance, CD3 monoclonal antibodies appear unique in their capacity to induce immunological tolerance that is an antigen-specific unresponsiveness in the absence of chronic immunosuppression. This has been well-established in experimental models, and recent data show successful clinical translation using humanized anti-CD3 antibodies. The aim of this brief review is to discuss the main characteristics of these very promising tools and to present the experimental and clinical results arguing for their unique tolerogenic ability.
Collapse
Affiliation(s)
- Lucienne Chatenoud
- Université René Descartes Paris 5, Hôpital Necker Enfants Malades, Paris, France.
| |
Collapse
|
65
|
Weir N, Athwal D, Brown D, Foulkes R, Kollias G, Nesbitt A, Popplewell A, Spitali M, Stephens S. A new generation of high-affinity humanized PEGylated Fab´ fragment anti-tumor necrosis factor-α monoclonal antibodies. ACTA ACUST UNITED AC 2006. [DOI: 10.2217/14750708.3.4.535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
66
|
Weir N, Athwal D, Brown D, Foulkes R, Kollias G, Nesbitt A, Popplewell A, Spitali M, Stephens S. A new generation of high-affinity humanized PEGylated Fab� fragment anti-tumor necrosis factor-? monoclonal antibodies. ACTA ACUST UNITED AC 2006. [DOI: 10.1586/14750708.3.4.535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
67
|
Kanai T, Watanabe M. Clinical application of human CD4+ CD25+ regulatory T cells for the treatment of inflammatory bowel diseases. Expert Opin Biol Ther 2006; 5:451-62. [PMID: 15934825 DOI: 10.1517/14712598.5.4.451] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As our understanding of the immunological and genetic basis of inflammatory bowel disease (IBD) grows, potential therapeutic options are being developed at a rapid pace. Nevertheless, new drugs for IBD are needed because about half of all patients with severe ulcerative colitis (UC) eventually undergo colectomy, and a significant part of Crohn's disease (CD) patients do not respond to standard medical therapies, including immunosuppressants and TNF-alpha neutralising antibodies, or suffer from significant side effects. Finally, recurrence of disease activity following remission is frequent in both UC and CD, and there is an unmet need for effective maintenance strategies. It is well-known that immune responses in the intestine remain in a state of controlled inflammation, suggesting that not only does active suppression by regulatory T (TR) cells play an important role in the normal intestinal homeostasis, but also its dysregulation might lead to the development of IBD. TR cells are functional subsets of T cells that downregulate adaptive immune responses by interfering with the activation of dendritic cells and proliferation of T cells. From experimental work it is now clear that TR cells play a critical role in maintaining immune homeostasis, and several therapeutic approaches have been targeted at the induction of TR cells in order to control mucosal inflammation. Before using TR cells clinically as living immunosuppressants for the treatment of IBD, however, we have to pass many critical checkpoints, such as the in vitro expansion of TR cells and the confirmation of their safety. This paper will discuss recently gained knowledge of human TR cells and the possibility of their clinical usages as a new strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Takanori Kanai
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Graduate School of Medicine, 1-5-45 Yushima, Tokyo 113-8519, Japan.
| | | |
Collapse
|
68
|
Chatenoud L. CD3-specific antibodies restore self-tolerance: mechanisms and clinical applications. Curr Opin Immunol 2005; 17:632-7. [PMID: 16214320 DOI: 10.1016/j.coi.2005.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Accepted: 09/21/2005] [Indexed: 01/06/2023]
Abstract
The treatment of autoimmune diseases using conventional chemical immunosuppressants has short-term effects, imposing the need for chronic treatment with its risks of over-immunosuppression. CD3-specific monoclonal antibodies can restore self-tolerance in a durable fashion after a single short-term treatment, as demonstrated in several experimental models and clinically in recent-onset insulin-dependent diabetes. Disease remission involves first an immediate 'freezing' of the autoimmune response, which is linked to CD3-specific antibody-induced antigenic modulation of CD3-TCR complex at the T lymphocyte surface, followed by 'resetting' of TGF-beta-dependent T-cell mediated immunoregulation. Tolerance induction is demonstrated by persisting disease protection in spite of recovery of full immunocompetence to unrelated antigens.
Collapse
Affiliation(s)
- Lucienne Chatenoud
- Université René Descartes Paris 5, INSERM U580, Hôpital Necker Enfants Malades, 161 Rue de Sèvres, 75015 Paris, France.
| |
Collapse
|
69
|
Yang W, Hussain S, Mi QS, Santamaria P, Delovitch TL. Perturbed Homeostasis of Peripheral T Cells Elicits Decreased Susceptibility to Anti-CD3-Induced Apoptosis in Prediabetic Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 2004; 173:4407-16. [PMID: 15383571 DOI: 10.4049/jimmunol.173.7.4407] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation-induced cell death (AICD) plays a key role in the homeostasis of the immune system. Autoreactive T cells are eliminated through AICD both from the thymus and periphery. In this study, we show that NOD peripheral T cells, especially CD8(+) T cells, display a decreased susceptibility to anti-CD3-induced AICD in vivo compared with T cells from diabetes-resistant B6, nonobese diabetes-resistant, and NOD.B6Idd4 mice. The susceptibility of NOD CD8(+) T cells to AICD varies in an age- and dose-dependent manner upon stimulation in vivo with either a mitogenic or nonmitogenic anti-CD3. NOD T cells preactivated by anti-CD3 in vivo are less susceptible than B6 T cells to TCR-induced AICD. Treatment of NOD mice with a mitogenic anti-CD3 depletes CD4(+)CD25(-)CD62L(+) but not CD4(+)CD25(+)CD62L(+) T cells, thereby resulting in an increase of the latter subset in the spleen. Treatment with a nonmitogenic anti-CD3 mAb delays the onset of T1D in 8.3 TCR transgenic NOD mice. These results demonstrate that the capacity of anti-CD3 to protect NOD mice from T1D correlates with its ability to perturb T cell homeostasis by inducing CD8(+) T cell AICD and increasing the number of CD4(+)CD25(+)CD62L(+) T cells in the periphery.
Collapse
MESH Headings
- Aging/genetics
- Aging/immunology
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Apoptosis/genetics
- Apoptosis/immunology
- CD3 Complex/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Death/genetics
- Cell Death/immunology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Genetic Predisposition to Disease
- Homeostasis/genetics
- Homeostasis/immunology
- Humans
- Interferon-gamma/antagonists & inhibitors
- Interferon-gamma/metabolism
- Interleukin-10/antagonists & inhibitors
- Interleukin-10/metabolism
- Interleukin-4/antagonists & inhibitors
- Interleukin-4/metabolism
- L-Selectin/biosynthesis
- Lymphocyte Activation/genetics
- Lymphocyte Depletion
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Transgenic
- Prediabetic State/genetics
- Prediabetic State/immunology
- Prediabetic State/pathology
- Prediabetic State/prevention & control
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Interleukin-2/biosynthesis
- Spleen/cytology
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/metabolism
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Wen Yang
- Autoimmunity/Diabetes Group, Robarts Research Institute, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
70
|
't Hart BA, Amor S, Jonker M. Evaluating the validity of animal models for research into therapies for immune-based disorders. Drug Discov Today 2004; 9:517-24. [PMID: 15183159 DOI: 10.1016/s1359-6446(04)03112-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The last few decades of the 20th century have shown an intensified search for safer and more effective medications against chronic diseases that burden ageing societies of the western world. The impressive development of biotechnological production techniques has greatly facilitated the pharmaceutical development of relatively non-toxic biological molecules. However, despite the huge investments, only a few effective therapies for immune-based diseases have reached the clinic. In this article we use examples from monoclonal antibody trials to discuss the validity and predictive strength of the animal models currently used for the development of effective therapies.
Collapse
MESH Headings
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibodies, Monoclonal/therapeutic use
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Disease Models, Animal
- Drug Evaluation, Preclinical/methods
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Immune System Diseases/immunology
- Immune System Diseases/therapy
- Immunotherapy/methods
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Organ Transplantation
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | |
Collapse
|
71
|
Tjelle TE, Corthay A, Lunde E, Sandlie I, Michaelsen TE, Mathiesen I, Bogen B. Monoclonal Antibodies Produced by Muscle after Plasmid Injection and Electroporation. Mol Ther 2004; 9:328-36. [PMID: 15006599 DOI: 10.1016/j.ymthe.2003.12.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Accepted: 12/16/2003] [Indexed: 01/20/2023] Open
Abstract
Antibodies are useful for the treatment of a variety of diseases. We here demonstrate that mouse muscle produced monoclonal antibodies (mAb) after a single injection of immunoglobulin genes as plasmid DNA. In vivo electroporation of muscle greatly enhanced antibody production. For chimeric antibodies, levels of 50-200 ng mAb/ml serum were obtained but levels declined after 7-14 days due to an immune response against the xenogeneic parts of the antibody. By contrast, fully mouse antibodies persisted in serum for at least 7 months. mAb produced by the muscle had correct structure, specificity, and biological effector functions. The findings were extended to a larger animal, the sheep, in which mAb serum levels of 30-50 ng/ml were obtained. Sustained levels of serum mAb, induced by single injection of Ig genes and electroporation of muscle cells, may offer significant advantages in the treatment of human diseases.
Collapse
|
72
|
Abstract
Immunosuppressive therapy aims to protect transplanted organs from host responses. Individuals have unique repertoires of responses to foreign antigens and toxic reactions to immunosuppressants; the former determining the type or intensity of rejection reactions and the latter influencing the severity of iatrogenic effects. Because existing agents target molecules that are widely distributed in tissues, new strategies must selectively block lymphoid cells only, disrupt alloresponses but not innate immune responses, interact synergistically with other agents, facilitate the homeostatic process that naturally leads to graft acceptance and ideally only interrupt donor-specific responses. Approaches presently under investigation aim to alter cell trafficking, or selectively deviate the maturation of antigen-presenting cells or inhibit lymphocyte-activation cascades - events that are crucial to rejection responses.
Collapse
Affiliation(s)
- Barry D Kahan
- Division of Immunology and Organ Transplantation, Department of Surgery, University of Texas Medical School at Houston, Suite 6.240, 6431 Fannin, Houston, Texas 77030, USA.
| |
Collapse
|
73
|
Simon T, Opelz G, Wiesel M, Ott RC, Süsal C. Serial peripheral blood perforin and granzyme B gene expression measurements for prediction of acute rejection in kidney graft recipients. Am J Transplant 2003; 3:1121-7. [PMID: 12919092 DOI: 10.1034/j.1600-6143.2003.00187.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In the present study we investigated whether peripheral blood gene expression measurements may serve as an early and non-invasive tool to predict renal allograft rejection. Peripheral blood was collected twice weekly after transplantation and gene expression was measured using real-time polymerase chain reaction (PCR). Recipients with acute rejection (n = 17) had higher levels of perforin and granzyme B transcript on days 5-7, 8-10, 11-13, 17-19, 20-22, and 26-29, as compared to patients without rejection (n = 50, p < 0.05 in all cases). Rejection diagnosis using gene expression criteria, determined with receiver operating characteristic (ROC) curves, was possible 2-30 days before traditional diagnosis (median 11 days). The best diagnostic result was obtained from samples taken on days 8-10, with a specificity of 90% and a sensitivity of 82% for perforin, and a specificity of 87% and sensitivity of 72% for granzyme B. Decreases in perforin (p < 0.01) and granzyme B expression (p < 0.05) were observed after initiation of anti-rejection therapy. Our data indicate that gene expression measurement is a useful tool for the recognition of graft rejection in its earliest stages. Serial measurements could be implemented as a monitoring system to highlight patients at higher risk of rejection, making them candidates for biopsy or pre-emptive anti-rejection therapy.
Collapse
Affiliation(s)
- Tania Simon
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
74
|
Mazzoccoli G, Grilli M, Carughi S, Puzzolante F, De Cata A, La Viola M, Giuliani A, Urbano N, Tarquini R, Perfetto F. Immune system alterations in lung cancer patients. Int J Immunopathol Pharmacol 2003; 16:167-74. [PMID: 12797908 DOI: 10.1177/039463200301600211] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The immune system plays an important role in the defense against neoplastic disease and immune responses show temporal changes related to circadian variations of antibodies, total lymphocytes in the peripheral blood and cell mediated immune responses. In this study we evaluate. lymphocyte subpopulations and interleukin-2 (IL-2) serum levels in peripheral blood samples collected at four-hour intervals for 24-hours starting at 06.00 h from ten healthy subjects aged 65-79 years (mean age +/- s.e. 67.28 +/- 3.11) and from ten subjects suffering from untreated non small cell lung cancer aged 65-78 years (mean age +/- s.e. 68.57 +/- 1.81). Areas under the curve, mean diurnal levels (mean of 06.00-10.00-14.00 h) and mean nocturnal levels (mean of 18.00-22.00-02.00 h) were calculated, and the presence of circadian rhythmicity was evaluate. When we compared AUC values there was a decrease in CD8bright (T suppressor subset) and an increase in CD16 (natural killer cells) and of IL-2 serum levels in cancer patients. When we compared mean diurnal levels, CD8 (T suppressor/cytotoxic subset) and CD8bright levels were lower, and CD16 levels were higher in cancer patients. When we compared mean nocturnal levels, CD16 and CD25 (T and B activated lymphocytes with expression of the a chain of IL-2 receptor) levels were higher, while CD8, CD8bright, CD20 (total B-cells), TcRd1 (epitope of the constant domain of d chain of T-cell receptor 1) and dTcS1 (epitope of the variable domain of d chain of T-cell receptor1) levels were lower in cancer patients. A clear circadian rhythm was validated for the time-qualified changes in CD4, CD20, HLA-DR with acrophase at night, and CD8, CD8 bright, CD8 dim, CD16, TcRd1 and dTcS1 with acrophase in the morning in the control group. A clear circadian rhythm was validated for the time-qualified changes in CD4 with acrophase at night, in the group of cancer patients. Results obtained in our study show that lung cancer is associated with anomalies of proportion and circadian variations of lymphocyte subsets that must be considered when adoptive immunotherapy has to be planned.
Collapse
Affiliation(s)
- G Mazzoccoli
- Department of Internal Medicine, Regional General Hospital, Casa Sollievo della Sofferenza, S.Giovanni Rotondo (FG), Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
Although they were used initially as non-specific immunosuppressants in transplantation, CD3-specific monoclonal antibodies have elicited renewed interest owing to their capacity to induce immune tolerance. In mouse models of autoimmune diabetes, CD3-specific antibodies induce stable disease remission by restoring tolerance to pancreatic beta-cells. This phenomenon was extended recently to the clinic--preservation of beta-cell function in recently diagnosed patients with diabetes was achieved by short-term administration of a CD3-specific antibody. CD3-specific antibodies arrest ongoing disease by rapidly clearing pathogenic T cells from the target. Subsequently, they promote long-term T-cell-mediated active tolerance. Recent data indicate that transforming growth factor-beta-dependent CD4+CD25+ regulatory T cells might have a central role in this effect.
Collapse
Affiliation(s)
- Lucienne Chatenoud
- Centre de l'Association Claude Bernard sur les Maladies Autoimmunes and Hôpital Necker Enfants Malades IRNEM, 161 Rue de Sèvres, 75015 Paris, France.
| |
Collapse
|
76
|
Abstract
The discovery of the human MHC in 1967 launched the field of organ and tissue transplantation. More than 800,000 such transplants have been performed during this time. Although matching of donor and recipient for MHC antigens was shown to be of great importance and continues to be so, the development of pharmacologic agents and antilymphocyte antibodies that interfere with the process of graft rejection has had a crucial role in the success of organ transplantation during the past 2 decades. Enormous progress has been made in understanding the immunologic mechanisms of graft rejection and of graft-versus-host disease. The roles of antibodies, antigen-presenting cells, helper and cytotoxic T cells, immune cell surface molecules, and signaling mechanisms and the cytokines they release have been clarified. This understanding is leading to the development of newer immunosuppressive agents targeting various components of the rejection process. Combinations of these agents work synergistically, leading to lower doses and reduced toxicity. Similarly, the development of effective T-cell depletion techniques has been of great importance for bone marrow transplantation when an HLA-identical sibling is not available. The major obstacle to the performance of solid organ transplantation currently is the shortage of donor organs.
Collapse
Affiliation(s)
- Rebecca H Buckley
- Department of Pediatrics, Allergy/Immunology, Duke University Medical Center, 362 Jones Building (Campus Box 2898), Durham, NC 27710-0001, USA
| |
Collapse
|
77
|
Yokota N, Daniels F, Crosson J, Rabb H. Protective effect of T cell depletion in murine renal ischemia-reperfusion injury. Transplantation 2002; 74:759-63. [PMID: 12364852 DOI: 10.1097/00007890-200209270-00005] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is the main cause of acute renal failure in both allograft and native kidney. Studies using T cell knockout mice have established an important role for T cells in renal IRI. T cell depletion strategies are effective in human allograft rejection. However, it is not known whether those are effective in renal IRI. Therefore, the effect of T cell depletion in a murine model of renal IRI using well-characterized antibodies (Abs) that have been effective in preventing experimental allograft rejection was studied. METHODS T cell depleting Abs to CD4 (GK1.5), CD8 (2.43) or pan-T cells (30.H12) were purified from hybridoma culture supernatant. Thymectomized C57BL/6 mice, treated with different combinations of T cell depleting Abs, underwent 30 min of bilateral renal IRI, followed by assessment of renal function, structure, and degree of T cell depletion in spleen, lymph nodes, and peripheral blood by flow cytometry. RESULTS Mice given both GK1.5 and 2.43 had considerable CD4 and CD8 cell depletion but no protection of renal function after ischemia-reperfusion (I/R) as measured by the rise in serum creatinine. However, when GK1.5 and 2.43 was administered combined with 30.H12, which more effectively depleted CD4 T cell numbers, a significant protection of renal function and structure was observed after I/R. Antibody combinations did not significantly alter other leukocyte populations. CONCLUSIONS These data demonstrate that T cell depletion can improve the course of experimental renal IRI. However, more aggressive T cell depletion strategies were required compared with that needed to prevent experimental allograft rejection.
Collapse
Affiliation(s)
- Naoko Yokota
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
78
|
Chatenoud L. The use of monoclonal antibodies to restore self-tolerance in established autoimmunity. Endocrinol Metab Clin North Am 2002; 31:457-75, ix. [PMID: 12092461 DOI: 10.1016/s0889-8529(01)00018-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The author hopes to convince the reader that the data presented argue for a stage during the development of IDDM when beta-cell destruction can be counteracted and tolerance to beta cells restored, provided the immune aggression is arrested. This argument constitutes a solid rationale for immunointervention in established IDDM, especially by using potent agents such as CD3. The future for the application of monoclonal antibodies not only in autoimmunity but also in transplantation is exiting. With the development of humanized monoclonal antibodies, therapeutic uses for them are likely to expand. Enormous progress has been made in the last 15 years, and it is likely that before a similar time period has elapsed, monoclonal antibodies will have become standard tools that will dispense the need for long-term immunosuppression and its inherent dangers in various clinical arenas.
Collapse
|
79
|
Niemeyer G, Koch M, Light S, Kuse ER, Nashan B. Long-term safety, tolerability and efficacy of daclizumab (Zenapax) in a two-dose regimen in liver transplant recipients. Am J Transplant 2002; 2:454-60. [PMID: 12123212 DOI: 10.1034/j.1600-6143.2002.20510.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A major thrust of transplantation research is to find more effective and less broadly toxic immunosuppressive agents. One potential way is the use of monoclonal antibodies directed to IL-2R alpha. Immunoprophylaxis with daclizumab, a humanized anti-IL-2R alpha monoclonal antibody, has been shown to be effective in the prevention of acute rejection in kidney transplant patients. These results encouraged us to initiate a pilot study in 28 liver transplant patients in 1997. Daclizumab was administered intravenously approximately 6 h after reperfusion (1 mg/kg) and on day 4 post-transplant (0.5 mg/kg). Additional immunosuppression consisted of cyclosporine A as well as of corticosteroids. Administration of daclizumab was not associated with any side-effects. We only experienced one acute rejection in a patient on day 17 post-transplant. It resolved immediately under therapy with prednisolone. The rate of opportunistic infections did not differ from results with conventional immunosuppressive regimens. At 4 years post-transplant no lymphoproliferative disease was observed. Patient survival at 12, 24, 36 and 48 months post-transplant was 88.5, 84.6, 80.8 and 73.1%, respectively. Immunoprophylaxis with a two-dose daclizumab regimen is safe, effective and well tolerated, and does not lead to increased opportunistic infections.
Collapse
Affiliation(s)
- Gerit Niemeyer
- Medizinische Hochschule Hannover, Klinik für Viszeral- und Transplantationschirurgie, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | | | | | | | | |
Collapse
|
80
|
Pascual M, Theruvath T, Kawai T, Tolkoff-Rubin N, Cosimi AB. Strategies to improve long-term outcomes after renal transplantation. N Engl J Med 2002; 346:580-90. [PMID: 11856798 DOI: 10.1056/nejmra011295] [Citation(s) in RCA: 622] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Manuel Pascual
- Renal Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | | | | | | | |
Collapse
|
81
|
von Herrath MG, Coon B, Wolfe T, Chatenoud L. Nonmitogenic CD3 antibody reverses virally induced (rat insulin promoter-lymphocytic choriomeningitis virus) autoimmune diabetes without impeding viral clearance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:933-41. [PMID: 11777992 DOI: 10.4049/jimmunol.168.2.933] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Treatment with nonmitogenic CD3 Ab reverses established autoimmune diabetes in nonobese diabetic mice by restoring self-tolerance, and is currently under clinical evaluation in patients presenting recent onset type I diabetes. Due to the immunosuppressive potential of this strategy, it was relevant to explore how this treatment would influence the outcome of concomitant viral infections. In this study, we used a transgenic model of virally induced autoimmune diabetes (rat insulin promoter-lymphocytic choriomeningitis virus) that allows for more precise tracking of the autoaggressive response and choice of the time point for initiation of autoimmunity. CD3 was most effective during a clearly defined prediabetic phase and prevented up to 100% of diabetes by drastically lowering activation of autoaggressive CD8 lymphocytes and their production of inflammatory cytokines. Interestingly, reversion of established disease could be achieved as well, when nonmitogenic CD3 was administered late during pathogenesis to overtly diabetic recipients. Most importantly, competence to clear viral infections was maintained. Thus, administration of nonmitogenic CD3 prevents diabetes by sufficient systemic reduction of (auto)aggressive lymphocytes, but without compromising antiviral immune competence.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/therapeutic use
- Arenaviridae Infections/immunology
- Arenaviridae Infections/prevention & control
- Arenaviridae Infections/virology
- Binding Sites, Antibody
- CD3 Complex/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Division/immunology
- Cell Movement/immunology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Diabetes Mellitus, Type 1/virology
- Immunoglobulin Fab Fragments/metabolism
- Immunoglobulin Fab Fragments/pharmacology
- Insulin/genetics
- Insulin/immunology
- Interleukin-4/biosynthesis
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Lymphocytes/cytology
- Lymphocytes/immunology
- Lymphocytic choriomeningitis virus/genetics
- Lymphocytic choriomeningitis virus/immunology
- Lymphotoxin-alpha/antagonists & inhibitors
- Lymphotoxin-beta
- Membrane Proteins/antagonists & inhibitors
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitogens/pharmacology
- Promoter Regions, Genetic/immunology
- Rats
- Receptors, Fc/metabolism
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- Spleen/transplantation
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Viral Proteins/genetics
Collapse
Affiliation(s)
- Matthias G von Herrath
- Department of Immune Regulation, La Jolla Institute for Allergy and Immunology, San Diego, CA 92121, USA.
| | | | | | | |
Collapse
|
82
|
Abstract
Over the past 3 decades, renal allograft survival has improved significantly as a result of the development of powerful immunosuppressive agents. Nevertheless, the overall half-life of renal allografts has increased marginally during that time period, owing to drug-related nephrotoxicity and chronic rejection. New immunosuppressive agents are being evaluated because of the need for a reduction in the dose of nephrotoxic calcineurin inhibitors and corticosteroids. Additional agents have demonstrated the ability to retard the onset of chronic rejection in preclinical transplant models. In concert with these efforts, approaches are in development to alleviate the ever increasing shortage of donor organs, including the as yet unrealized goals of successful and practical xenotransplantation and the bioartificial kidney. Further identification and development of novel agents that target the specific components of the allograft response will provide the key to the achievement of donor-specific tolerance, the "Holy Grail" of solid organ transplantation.
Collapse
Affiliation(s)
- P P Luke
- Departments of Surgery and Urology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
83
|
Nishikido M, Kiyohara T, Koga S, Shindo K, Matsuya F, Saito Y, Kanetake H. OK432-induced killer cell activity: potential method for monitoring immunological complications after renal transplantation. Nephrol Dial Transplant 2001; 16:2067-71. [PMID: 11572898 DOI: 10.1093/ndt/16.10.2067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Various clinical and biochemical parameters are currently in use for monitoring allograft rejection. However, the mechanism of allograft rejection is complex and it is frequently difficult to obtain a prompt and accurate diagnosis. We examined the usefulness of OK432-induced killer cell activity as an immunological monitoring system for acute renal rejection after renal transplantation. METHODS Twenty-four renal transplant recipients, seven patients on haemodialysis, and 10 normal volunteers were enrolled in our study. The killer cell activity of peripheral blood mononuclear cells was induced by culturing these cells with the immunopotentiator, OK432, a heat and penicillin-treated lyophilized powder of the Su-strain of Streptococcus pyogenes. RESULTS The OK432-induced killer cell activity of renal transplant recipients without acute rejection (stable recipients) was significantly lower than in normal volunteers. In four renal transplant recipients with acute rejection, the killer cell activity was significantly higher than in stable recipients. In three recipients suffering from opportunistic infections, killer cell activity was significantly suppressed compared with stable recipients. CONCLUSIONS Our new test utilizing OK432-induced killer cell activity is potentially useful for monitoring the immunological state and complications after renal transplantation.
Collapse
Affiliation(s)
- M Nishikido
- Department of Urology, Nagasaki University School of Medicine, Nagasaki University Hospital, Nagasaki City, Japan
| | | | | | | | | | | | | |
Collapse
|
84
|
Chung DH, Belyakov IM, Derby MA, Wang J, Boyd LF, Berzofsky JA, Margulies DH. Competitive inhibition in vivo and skewing of the T cell repertoire of antigen-specific CTL priming by an anti-peptide-MHC monoclonal antibody. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:699-707. [PMID: 11441073 DOI: 10.4049/jimmunol.167.2.699] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have recently described a mAb, KP15, directed against the MHC-I/peptide molecular complex consisting of H-2D(d) and a decamer peptide corresponding to residues 311-320 of the HIV IIIB envelope glycoprotein gp160. When administered at the time of primary immunization with a vaccinia virus vector encoding gp160, the mAb blocks the subsequent appearance of CD8(+) CTL with specificity for the immunodominant Ag, P18-I10, presented by H-2D(d). This inhibition is specific for this particular peptide Ag; another H-2D(d)-restricted gp160 encoded epitope from a different HIV strain is not affected, and an H-2L(d)-restricted epitope encoded by the viral vector is also not affected. Using functional assays and specific immunofluorescent staining with multivalent, labeled H-2D(d)/P18-I10 complexes (tetramers), we have enumerated the effects of blocking of priming on the subsequent appearance, avidity, and TCR Vbeta usage of Ag-specific CTL. Ab blocking skews the proportion of high avidity cells emerging from immunization. Surprisingly, Vbeta7-bearing Ag-specific TCR are predominantly inhibited, while TCR of several other families studied are not affected. The ability of a specific MHC/peptide mAb to inhibit and divert the CD8(+) T cell response holds implications for vaccine design and approaches to modulate the immune response in autoimmunity.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/administration & dosage
- Antibodies, Blocking/metabolism
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/metabolism
- Binding, Competitive/immunology
- Cytotoxicity, Immunologic/immunology
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- HIV Antigens/immunology
- HIV Antigens/metabolism
- HIV Envelope Protein gp160/immunology
- HIV Envelope Protein gp160/metabolism
- Histocompatibility Antigen H-2D
- Humans
- Injections, Intraperitoneal
- Injections, Intravenous
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred BALB C
- Oligopeptides/antagonists & inhibitors
- Oligopeptides/immunology
- Oligopeptides/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/antagonists & inhibitors
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Stem Cells/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- D H Chung
- Laboratory of Immunology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
85
|
Affiliation(s)
- S Lederman
- Laboratory of Molecular Immunology, Columbia University, New York, New York, USA
| |
Collapse
|
86
|
A History of Clinical Transplantation. Surgery 2001. [DOI: 10.1007/978-3-642-57282-1_61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
87
|
Affiliation(s)
- G M Danovitch
- UCLA School of Medicine, Los Angeles, California 90095-1796, USA.
| |
Collapse
|
88
|
Abstract
New drugs have recently been added that may eventually replace the two-decade dominance of cyclosporin in solid organ transplantation. This cornerstone of immunosuppression was introduced by Borel [1] and Calne [2] in the mid-70s. In 1989, Starzl et al., after 2 years of preclinical experimentation, introduced tacrolimus (originally designated as FK506 by the Fujisawa Pharmaceutical Company of Japan) as a potent immunosuppressant for liver transplants [3]. Also, in recent years, a variety of novel purine and pyrimidine biosynthesis inhibitors have been tested for transplantation therapy. The leading agent which appears to be replacing the 35-year position occupied by azathioprine is the semi-synthetic morpholinoethyl ester of mycophenolic acid (MPA), mycophenolate mofetil (MMF), introduced by Allison [4] and Sollinger [5], and developed by the Syntex Corporation (now Roche Pharmaceuticals). Others, affecting different intra- or intercellular messages amplifying immunity, are in the pipeline.
Collapse
Affiliation(s)
- G Ciancio
- Department of Surgery, Division of Transplantation, University of Miami School of Medicine, PO Box 012440, Miami, Fl. 33101, USA.
| | | | | |
Collapse
|
89
|
Abstract
The emergence of transplantation has seen the development of increasingly potent immunosuppressive agents, progressively better methods of tissue and organ preservation, refinements in histocompatibility matching, and numerous innovations in surgical techniques. Such efforts in combination ultimately made it possible to successfully engraft all of the organs and bone marrow cells in humans. At a more fundamental level, however, the transplantation enterprise hinged on two seminal turning points. The first was the recognition by Billingham, Brent, and Medawar in 1953 that it was possible to induce chimerism-associated neonatal tolerance deliberately. This discovery escalated over the next 15 years to the first successful bone marrow transplantations in humans in 1968. The second turning point was the demonstration during the early 1960s that canine and human organ allografts could self-induce tolerance with the aid of immunosuppression. By the end of 1962, however, it had been incorrectly concluded that turning points one and two involved different immune mechanisms. The error was not corrected until well into the 1990s. In this historical account, the vast literature that sprang up during the intervening 30 years has been summarized. Although admirably documenting empiric progress in clinical transplantation, its failure to explain organ allograft acceptance predestined organ recipients to lifetime immunosuppression and precluded fundamental changes in the treatment policies. After it was discovered in 1992 that long-surviving organ transplant recipients had persistent microchimerism, it was possible to see the mechanistic commonality of organ and bone marrow transplantation. A clarifying central principle of immunology could then be synthesized with which to guide efforts to induce tolerance systematically to human tissues and perhaps ultimately to xenografts.
Collapse
Affiliation(s)
- T E Starzl
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, PA 15213, USA
| |
Collapse
|
90
|
Moldenhauer A, Kohlhaus R, Salama A. An Extended Lymphocytotoxicity Test for Patients Treated with Lymphocytotoxic Antibodies. Vox Sang 2000. [DOI: 10.1046/j.1423-0410.2000.7840250.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
91
|
Gonin JM. Maintenance immunosuppression: new agents and persistent dilemmas. ADVANCES IN RENAL REPLACEMENT THERAPY 2000; 7:95-116. [PMID: 10782729 DOI: 10.1053/rr.2000.5271] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Since the approval of cyclosporine in 1983, only 3 drugs, mycophenolate mofetil, tacrolimus, and sirolimus, have been approved for maintenance immunosuppression in renal transplant recipients. All 3 agents decrease the incidence of early acute allograft rejection. An increase in intermediate and long-term graft survival has not been shown. However, survival data from these clinical trials should be interpreted with caution because the studies were not designed for this purpose. All 3 drugs have significant, albeit different, safety profiles. It remains to be seen whether, the lower incidence of hypertension and hyperlipidemia seen in tacrolimus-treated patients will reduce the incidence and severity of the cardiovascular disease experienced by renal transplant recipients. Sirolimus causes severe hyperlipidemia, and the long-term consequences both on the pathogenesis of cardiovascular disease and on lipid-associated renal injury have yet to be determined. Tacrolimus and mycophenolate mofetil appear to increase graft survival in pancreas-kidney recipients but their efficacy in another high-risk group, African-American recipients, has not yet been clearly shown. However, the trend toward improved graft survival in African-American recipients treated with tacrolimus is encouraging. Steroid-withdrawal remains a goal in the posttransplant period. The available data from steroid-withdrawal and steroid-avoidance clinical trials are mixed. Steroid withdrawal can be achieved in about 50% of patients on a cyclosporine-based immunosuppression regimen. Steroid-withdrawal under coverage of tacrolimus, mycophenolate mofetil or Neoral (Novartis Pharmaceuticals, East Hanover, NJ) may be more successful than that achieved in patients receiving Sandimmune (Novartis Pharmaceuticals). Further studies are needed in this area.
Collapse
Affiliation(s)
- J M Gonin
- Division of Nephrology and Hypertension, Georgetown University Medical Center, Washington, DC 20007, USA.
| |
Collapse
|
92
|
Cook DJ, Bishay ES, Yamani M. The use and misuse of immunologic monitoring after transplantation: approaches that have proved useful. Curr Opin Cardiol 2000; 15:104-7. [PMID: 10963147 DOI: 10.1097/00001573-200003000-00007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Current practice in the monitoring of cardiac transplants revolves around the use of the endomyocardial biopsy. While this is effective for the identification of an ongoing immune response in the graft, for years investigators have explored less invasive approaches in the hope of achieving the same goal by examining the patient's immune response. For a number of years, lymphocytes, their subsets, and their level of activation in the periphery were investigated. To a large degree, it was a lack of specificity in these approaches that led to their falling out of favor. Examination of donor-specific reactivity by means of lymphocyte proliferation assays has also been used; however, these approaches have been impeded by the time and effort required to accomplish them. During the last few years, flow cytometric cross-matching during the posttransplant period has been used at our institution. While this cross-matching focuses on the humoral immune responses, we have found it to be of value in identifying patients at risk of rejection and in allowing the assessment of treatment modalities used to treat ongoing rejection. While the perfect approach remains to be found, the potential advantages of immunologic monitoring would seem to justify continued study.
Collapse
Affiliation(s)
- D J Cook
- The Transplant Center, Cleveland Clinic Foundation, Ohio 44195, USA
| | | | | |
Collapse
|
93
|
Laroche-Traineau J, Clofent-Sanchez G, Santarelli X. Three-step purification of bacterially expressed human single-chain Fv antibodies for clinical applications. JOURNAL OF CHROMATOGRAPHY. B, BIOMEDICAL SCIENCES AND APPLICATIONS 2000; 737:107-17. [PMID: 10681047 DOI: 10.1016/s0378-4347(99)00441-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We have obtained a cell line which secretes a human monoclonal IgM (B7) reacting with the myosin heavy chain of human heart. We have constructed single-chain fragments (scFv) of B7. The scFv may be useful for the imaging of myocardial necrosis after myocarditis, cardiac drug toxicosis or graft rejection. The aim of our work was to purify the scFv for immunoscintigraphy. We describe several purification steps including immobilized metal affinity chromatography (IMAC), anti-c-myc monoclonal antibody affinity chromatography, size-exclusion chromatography with Superdex 75 HR 10/30 and ion-exchange chromatography (mini Q TM 30Q).
Collapse
Affiliation(s)
- J Laroche-Traineau
- Laboratoire de Pathologie Cellulaire de l'Hémostase, CNRS, UMR 5533, Hôpital du Haut-Lévêque, Pessac, France
| | | | | |
Collapse
|
94
|
Noda H, Fujimura Y, Gohra H, Hamano K, Katoh T, Esato K. Coronary bypass surgery after renal transplantation. THE JAPANESE JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY : OFFICIAL PUBLICATION OF THE JAPANESE ASSOCIATION FOR THORACIC SURGERY = NIHON KYOBU GEKA GAKKAI ZASSHI 1999; 47:135-40. [PMID: 10226414 DOI: 10.1007/bf03217958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
We report, herein, cases of two renal transplantation patients who underwent coronary artery bypass grafting and discuss the perioperative management of this clinical situation. The first case was a 43-year-old male who underwent coronary artery bypass grafting 50 days after renal transplantation, and the second was a chronic case of a 49-year-old male who underwent coronary artery bypass grafting 17 years after renal transplantation. Prior to the operation, the first patient was continuously administered 2 mg/kg/day of cyclosporin A with the dosage regulated according to the plasma level. The second patient was administered 50 mg/day of cyclophosphamide intravenously instead of an oral dosage of 50 mg/day of azathioprine just prior to the operation. In both patients, perfusion pressure during cardiopulmonary bypass was maintained at approximately 80 mmHg in order to obtain optimal urine output. The CD4/CD8 ratio was monitored for indication of graft rejection, but no remarkable changes were observed perioperatively in either patient. Both patients followed a good clinical course and their postoperative renal function was well maintained. The urine output during cardiopulmonary bypass was 300 ml and 650 ml, respectively. The patients were discharged 15 and 27 days after their operation, respectively. In conclusion, coronary artery bypass grafting can be safely performed in patients who have undergone renal transplantation, if there is appropriate perioperative usage of immunosuppressive agents and maintenance of perfusion pressure during cardiopulmonary bypass.
Collapse
Affiliation(s)
- H Noda
- First Department of Surgery, Yamaguchi University School of Medicine, Ube, Japan
| | | | | | | | | | | |
Collapse
|
95
|
High-dose/activation-associated tolerance model for allografts: lessons from spontaneous tolerance of transplanted livers. Curr Opin Organ Transplant 1999. [DOI: 10.1097/00075200-199903000-00011] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
96
|
Rubin RH, Ikonen T, Gummert JF, Morris RE. The therapeutic prescription for the organ transplant recipient: the linkage of immunosuppression and antimicrobial strategies. Transpl Infect Dis 1999; 1:29-39. [PMID: 11428969 DOI: 10.1034/j.1399-3062.1999.10104.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Infection and rejection, the two major barriers to successful organ transplantation, are closely linked, with immunosuppressive therapy being central to the pathogenesis of both. After almost two decades when azathioprine and prednisone, supplemented by antilymphocyte antibody therapy, were the cornerstones of post-transplant immunosuppressive programs, there has been a major increase in the therapeutic armamentarium available to treat rejection: cyclosporine, tacrolimus, mycophenolate mofetil, rapamycin, and antibodies directed against the interleukin-2 receptor. These agents are potent inhibitors of microbial specific T cell function, thus potentiating opportunistic infection with herpes group viruses, fungal and mycobacterial species, Strongyloides stercoralis, and a variety of intracellular pathogens. The mechanisms by which each of these drugs exerts its effects are an important determinant of the antimicrobial strategies that will be necessary to combat infection. Indeed, strategies to limit these infections are being linked to the nature of the immunosuppressive therapy required in a particular patient. Thus, the therapeutic prescription for the transplant patient is said to have two components: an immunosuppressive component to prevent and treat rejection, and an antimicrobial one to make it safe. In addition to using antimicrobial agents therapeutically, in the transplant patient prevention is stressed in which antibiotics are deployed prophylactically or preemptively.
Collapse
Affiliation(s)
- R H Rubin
- Massachusetts General Hospital, Boston, Massachusetts 02114-2686, USA.
| | | | | | | |
Collapse
|
97
|
Abstract
Although several new immunosuppressive medications have been developed in the past decade, many possible avenues are yet to be explored. Although the newer agents have not reflected any clear benefit in patient or graft survival over CsA or tacrolimus, they have been useful in reducing the incidence and severity of rejection, reducing the concomitant use of steroids, and decreasing the doses of CsA or tacrolimus to minimize their toxicity profile. The appearance of these new agents has given more options to clinicians, who can select the one with the least toxicity and most efficacy for individual patients. In the future, combinations of these agents, in conjunction with a strategy to induce tolerance of the donor organ without drug toxicity, will be the goal.
Collapse
Affiliation(s)
- A Jain
- Thomas E. Starzl Transplantation Institute, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
98
|
Vallhonrat H, Williams WW, Cosimi AB, Tolkoff-Rubin N, Ginns LC, Wain JC, Preffer F, Olszak I, Wee S, Delmonico FL, Pascual M. In vivo generation of C4d, Bb, iC3b, and SC5b-9 after OKT3 administration in kidney and lung transplant recipients. Transplantation 1999; 67:253-8. [PMID: 10075589 DOI: 10.1097/00007890-199901270-00011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND OKT3 monoclonal antibody therapy results in an acute clinical syndrome (ACS) associated with the release of tumor necrosis factor and sequestration of neutrophils in the lungs. We have previously shown that inhibition of tumor necrosis factor does not completely eliminate OKT3-ACS, suggesting that other factors also contribute to the ACS. The current studies analyzed complement activation in vivo during the first hour after OKT3 administration. METHODS Renal (n=4) and lung (n=4) transplant recipients received OKT3 as treatment for rejection and induction therapy, respectively. Complement activation products C4d, Bb, iC3b, and SC5b-9 were measured by ELISA. Hemodynamic parameters were also monitored in the lung transplant recipients. Neutrophil expression of CD11a, CD11b, and CD18 was monitored by flow cytometry. Controls included patients receiving methylprednisolone for rejection (n=4), two adults with adult respiratory distress syndrome who received extracorporeal membrane oxygenation, and normal volunteers (n=5). P values less than 0.05 (*) were considered significant. RESULTS Increases in the plasma levels of C4d, Bb, iC3b, and SC5b-9 were observed in seven of eight patients after OKT3 administration. Mean values (n=8) at 0, 15, and 60 min (in microg/ml) were as follows-C4d: 1.865, 2.644*, and 2.607*; Bb: 0.245, 0.411, and 0.385; iC3b: 10.881, 17.242*, and 15.145*; and SC5b-9: 0.232, 0.269, and 0.302*. An increase in CD11b and CD18 and a decrease of CD11a on neutrophils in parallel with complement activation was observed. In lung transplant recipients, C3 activation correlated with increases in mean pulmonary and central venous pressures (P<0.05). As compared with extracorporeal membrane oxygenation, which activated classical and alternative pathways, OKT3 predominantly activated complement by the classical pathway. Methylprednisolone pulses did not activate complement. CONCLUSIONS Complement activation is an early event after OKT3 administration and is associated with the increased expression of adhesion molecules on neutrophils and with pulmonary hemodynamic changes. Effective therapeutic approaches to the control of early monoclonal antibody side effects may require measures that limit complement activation in addition to reducing cytokine activity.
Collapse
Affiliation(s)
- H Vallhonrat
- Massachusetts General Hospital, and Department of Surgery, Harvard Medical School, Boston 02114, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Nashan B, Light S, Hardie IR, Lin A, Johnson JR. Reduction of acute renal allograft rejection by daclizumab. Daclizumab Double Therapy Study Group. Transplantation 1999; 67:110-5. [PMID: 9921806 DOI: 10.1097/00007890-199901150-00019] [Citation(s) in RCA: 252] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Acute rejection is still a major problem in renal transplantation and is one of the most important causes of chronic graft dysfunction and late graft loss. Selective immunosuppression with a humanized antibody against the alpha-chain of the interleukin (IL)-2 receptor (CD25) was evaluated to demonstrate the efficacy of this type of immunoprophylaxis in combination with dual immunosuppression. METHODS We studied the effect of daclizumab, a humanized monoclonal antibody against the alpha-chain of the IL-2 receptor, in a randomized double-blind, prospective phase III clinical trial in 275 patients receiving a first cadaveric renal allograft. Among them 111 (83%) in the placebo arm and 116 (82%) in the daclizumab arm received the full regimen of five doses (1.0 mg/kg) every other week. Baseline immunosuppression consisted of cyclosporine and corticosteroids. RESULTS At 6 months, 39 (28%) of the patients in the daclizumab group had biopsy-proven rejections, as compared with 63 (47%) in the placebo group (P=0.001). The need for additional antilymphocyte therapy, antithymocyte globulin, antilymphocyte globulin (ATG, ALG, OKT3) was also lower in the daclizumab group (8% vs. 16%, P=0.02), and they required significantly lower mean (+/- SD) cumulative doses of prednisone (3750+/-1981 mg vs. 4438+/-2667 mg in the placebo group, P=0.01). Graft function was significantly better (P=0.02) in the daclizumab group (graft function rate: 58 vs. 51 ml/min, mean) as was patient survival (P=0.01, 99% vs. 94%). No specific adverse events were observed in daclizumab-treated patients. Patients receiving daclizumab experienced fewer cytomegalovirus infections (18% vs. 25%), and none died from severe infectious complications, compared to four patients in the placebo arm. No patient in the daclizumab group had a lymphoproliferative disorder or any other form of immunosuppression-related tumor during the first year after transplant. CONCLUSIONS Administration of daclizumab in addition to dual immunosuppression therapy significantly reduced biopsy-proven acute rejection after renal transplantation, improved patient survival, and did not add to the toxicity of the immunosuppressive regimen.
Collapse
Affiliation(s)
- B Nashan
- Klinik für Abdominal- und Transplantationschirurgie, Medizinische Hochschule Hannover, Germany.
| | | | | | | | | |
Collapse
|
100
|
Jindal RM, Greer G, Popescu I, Sidner RA. Lymphocyte Subset Analysis for the Diagnosis of Rejection and Infection in Recipients of Liver Transplants. Am Surg 1999. [DOI: 10.1177/000313489906500118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We retrospectively investigated the predictive value of lymphocyte subset changes in the peripheral blood of 37 consecutive liver transplant recipients for rejection and infection, both bacterial and cytomegalovirus, during a period of 2 years. Total lymphocyte count correlated with biopsy-proven rejection. There was moderate elevation of CD16+ count during the course of bacterial infection.
Collapse
Affiliation(s)
- R. M. Jindal
- Departments of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Gerald Greer
- Departments of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Irinel Popescu
- Departments of Surgery, Mount Sinai Medical Center, New York, New York
| | - R. A. Sidner
- Departments of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|