51
|
Villa NY, Rahman MM, McFadden G, Cogle CR. Therapeutics for Graft-versus-Host Disease: From Conventional Therapies to Novel Virotherapeutic Strategies. Viruses 2016; 8:85. [PMID: 27011200 PMCID: PMC4810275 DOI: 10.3390/v8030085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has a curative potential for many hematologic malignancies and blood diseases. However, the success of allo-HSCT is limited by graft-versus-host disease (GVHD), an immunological syndrome that involves inflammation and tissue damage mediated by donor lymphocytes. Despite immune suppression, GVHD is highly incident even after allo-HSCT using human leukocyte antigen (HLA)-matched donors. Therefore, alternative and more effective therapies are needed to prevent or control GVHD while preserving the beneficial graft-versus-cancer (GVC) effects against residual disease. Among novel therapeutics for GVHD, oncolytic viruses such as myxoma virus (MYXV) are receiving increased attention due to their dual role in controlling GVHD while preserving or augmenting GVC. This review focuses on the molecular basis of GVHD, as well as state-of-the-art advances in developing novel therapies to prevent or control GVHD while minimizing impact on GVC. Recent literature regarding conventional and the emerging therapies are summarized, with special emphasis on virotherapy to prevent GVHD. Recent advances using preclinical models with oncolytic viruses such as MYXV to ameliorate the deleterious consequences of GVHD, while maintaining or improving the anti-cancer benefits of GVC will be reviewed.
Collapse
Affiliation(s)
- Nancy Y Villa
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
52
|
Abstract
Antigen-experienced T cells, also known as memory T cells, are functionally and phenotypically distinct from naive T cells. Their enhanced expression of adhesion molecules and reduced requirement for co-stimulation enables them to mount potent and rapid recall responses to subsequent antigen encounters. Memory T cells generated in response to prior antigen exposures can cross-react with other nonidentical, but similar, antigens. This heterologous cross-reactivity not only enhances protective immune responses, but also engenders de novo alloimmunity. This latter characteristic is increasingly recognized as a potential barrier to allograft acceptance that is worthy of immunotherapeutic intervention, and several approaches have been investigated. Calcineurin inhibition effectively controls memory T-cell responses to allografts, but this benefit comes at the expense of increased infectious morbidity. Lymphocyte depletion eliminates allospecific T cells but spares memory T cells to some extent, such that patients do not completely lose protective immunity. Co-stimulation blockade is associated with reduced adverse-effect profiles and improved graft function relative to calcineurin inhibition, but lacks efficacy in controlling memory T-cell responses. Targeting the adhesion molecules that are upregulated on memory T cells might offer additional means to control co-stimulation-blockade-resistant memory T-cell responses.
Collapse
|
53
|
Abstract
Memory cells are the products of immune responses but also exert significant impact on subsequent immunity and immune tolerance, thus placing them in a unique position in transplant research. Memory cells are heterogeneous, including not only memory T cells but also memory B cells and innate memory cells. Memory cells are a critical component of protective immunity against invading pathogens, especially in immunosuppressed patients, but they also mediate graft loss and tolerance resistance. Recent studies suggest that some memory cells unexpectedly act as regulatory cells, promoting rather than hindering transplant survival. This functional diversity makes therapeutic targeting of memory cells a challenging task in transplantation. In this article, we highlight recent advances in our understanding of memory cells, focusing on diversity of memory cells and mechanisms involved in their induction and functions. We also provide a broad overview on the challenges and opportunities in targeting memory cells in the induction of transplant tolerance.
Collapse
|
54
|
Wang Y, Zheng Z, Zhu X, Han J, Dong M, Tao K, Wang H, Wang Y, Hu D. The amelioration of composite tissue allograft rejection by TIM-3-modified dendritic cell: Regulation of the balance of regulatory and effector T cells. Immunol Lett 2016; 169:15-22. [DOI: 10.1016/j.imlet.2015.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/19/2015] [Accepted: 11/04/2015] [Indexed: 11/29/2022]
|
55
|
Alegre ML, Chong AS. Costimulatory molecules. Transpl Immunol 2015. [DOI: 10.1002/9781119072997.ch4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
56
|
Maltzman JS, Thomson A, Rothstein DM. T cells and the principles of immune responses. Transpl Immunol 2015. [DOI: 10.1002/9781119072997.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
57
|
Abstract
Generation of an effective immune response against foreign antigens requires two distinct molecular signals: a primary signal provided by the binding of antigen-specific T-cell receptor to peptide-MHC on antigen-presenting cells and a secondary signal delivered via the engagement of costimulatory molecules. Among various costimulatory signaling pathways, the interactions between CD40 and its ligand CD154 have been extensively investigated given their essential roles in the modulation of adaptive immunity. Here, we review current understanding of the role CD40/CD154 costimulation pathway has in alloimmunity, and summarize recent mechanistic and preclinical advances in the evaluation of candidate therapeutic approaches to target this receptor-ligand pair in transplantation.
Collapse
Affiliation(s)
- Tianshu Zhang
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard N Pierson
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Baltimore VA Medical Center, Baltimore, MD, USA
| | - Agnes M Azimzadeh
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
58
|
You S. Differential sensitivity of regulatory and effector T cells to cell death: a prerequisite for transplant tolerance. Front Immunol 2015; 6:242. [PMID: 26042125 PMCID: PMC4437185 DOI: 10.3389/fimmu.2015.00242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/06/2015] [Indexed: 12/13/2022] Open
Abstract
Despite significant progress achieved in transplantation, immunosuppressive therapies currently used to prevent graft rejection are still endowed with severe side effects impairing their efficiency over the long term. Thus, the development of graft-specific, non-toxic innovative therapeutic strategies has become a major challenge, the goal being to selectively target alloreactive effector T cells while sparing CD4+Foxp3+ regulatory T cells (Tregs) to promote operational tolerance. Various approaches, notably the one based on monoclonal antibodies or fusion proteins directed against the TCR/CD3 complex, TCR coreceptors, or costimulatory molecules, have been proposed to reduce the alloreactive T cell pool, which is an essential prerequisite to create a therapeutic window allowing Tregs to induce and maintain allograft tolerance. In this mini review, we focus on the differential sensitivity of Tregs and effector T cells to the depleting and inhibitory effect of these immunotherapies, with a particular emphasis on CD3-specific antibodies that beyond their immunosuppressive effect, also express potent tolerogenic capacities.
Collapse
Affiliation(s)
- Sylvaine You
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; INSERM U1151, Institut Necker-Enfants Malades , Paris , France ; CNRS UMR 8253, Institut Necker-Enfants Malades , Paris , France
| |
Collapse
|
59
|
Khalifian S, Raimondi G, Lee WA, Brandacher G. Taming inflammation by targeting cytokine signaling: new perspectives in the induction of transplantation tolerance. Immunotherapy 2015; 6:637-53. [PMID: 24896631 DOI: 10.2217/imt.14.25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transplantation tolerance remains an elusive goal, partly due to limitations in our understanding of the interplay between inflammatory mediators and their role in the activation and regulation of T lymphocytes. Although multiple mechanisms acting both centrally and peripherally are responsible for tolerance induction, the signaling pathways leading to activation or regulation of adaptive immunity are often complex, branched, redundant and modulated by the microenvironment's inflammatory milieu. Accumulating evidence clearly indicates that inflammatory cytokines limit the tolerogenic potential of immunomodulatory protocols by supporting priming of the immune system and counteracting regulatory mechanisms, ultimately promoting rejection. In this review, we summarize recent progress in the development of novel therapeutics to manipulate this inflammatory environment and achievements in targeted inhibition of inflammatory cytokine signaling. Ultimately, robust transplant tolerance induction will probably require a multifaceted, holistic approach that integrates the various mechanisms of tolerance induction, incorporates the dynamic alterations in costimulatory requirements of alloreactive T cells, while maintaining endogenous mechanisms of immune regulation.
Collapse
Affiliation(s)
- Saami Khalifian
- Department of Plastic and Reconstructive Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
60
|
Egli A, Humar A, Widmer LA, Lisboa LF, Santer DM, Mueller T, Stelling J, Baluch A, O'Shea D, Houghton M, Kumar D. Effect of Immunosuppression on T-Helper 2 and B-Cell Responses to Influenza Vaccination. J Infect Dis 2015; 212:137-46. [DOI: 10.1093/infdis/jiv015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/23/2014] [Indexed: 12/23/2022] Open
|
61
|
Song Y, Buchwald P. TNF superfamily protein-protein interactions: feasibility of small- molecule modulation. Curr Drug Targets 2015; 16:393-408. [PMID: 25706111 PMCID: PMC4408546 DOI: 10.2174/1389450116666150223115628] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 01/09/2023]
Abstract
The tumor necrosis factor (TNF) superfamily (TNFSF) contains about thirty structurally related receptors (TNFSFRs) and about twenty protein ligands that bind to one or more of these receptors. Almost all of these cell surface protein-protein interactions (PPIs) represent high-value therapeutic targets for inflammatory or immune modulation in autoimmune diseases, transplant recipients, or cancers, and there are several biologics including antibodies and fusion proteins targeting them that are in various phases of clinical development. Small-molecule inhibitors or activators could represent possible alternatives if the difficulties related to the targeting of protein-protein interactions by small molecules can be addressed. Compounds proving the feasibility of such approaches have been identified through different drug discovery approaches for a number of these TNFSFR-TNFSF type PPIs including CD40-CD40L, BAFFR-BAFF, TRAIL-DR5, and OX40-OX40L. Corresponding structural, signaling, and medicinal chemistry aspects are briefly reviewed here. While none of these small-molecule modulators identified so far seems promising enough to be pursued for clinical development, they provide proof-of-principle evidence that these interactions are susceptible to small-molecule modulation and can serve as starting points toward the identification of more potent and selective candidates.
Collapse
Affiliation(s)
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, 1450 NW 10 Ave (R-134), Miami, FL 33136, USA.
| |
Collapse
|
62
|
Sivanathan KN, Gronthos S, Rojas-Canales D, Thierry B, Coates PT. Interferon-gamma modification of mesenchymal stem cells: implications of autologous and allogeneic mesenchymal stem cell therapy in allotransplantation. Stem Cell Rev Rep 2014; 10:351-75. [PMID: 24510581 DOI: 10.1007/s12015-014-9495-2] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSC) have unique immunomodulatory and reparative properties beneficial for allotransplantation cellular therapy. The clinical administration of autologous or allogeneic MSC with immunosuppressive drugs is able to prevent and treat allograft rejection in kidney transplant recipients, thus supporting the immunomodulatory role of MSC. Interferon-gamma (IFN-γ) is known to enhance the immunosuppressive properties of MSC. IFN-γ preactivated MSC (MSC-γ) directly or indirectly modulates T cell responses by enhancing or inducing MSC inhibitory factors. These factors are known to downregulate T cell activation, enhance T cell negative signalling, alter T cells from a proinflammatory to an anti-inflammatory phenotype, interact with antigen-presenting cells and increase or induce regulatory cells. Highly immunosuppressive MSC-γ with increased migratory and reparative capacities may aid tissue repair, prolong allograft survival and induce allotransplant tolerance in experimental models. Nevertheless, there are contradictory in vivo observations related to allogeneic MSC-γ therapy. Many studies report that allogeneic MSC are immunogenic due to their inherent expression of major histocompatibility (MHC) molecules. Enhanced expression of MHC in allogeneic MSC-γ may increase their immunogenicity and this can negatively impact allograft survival. Therefore, strategies to reduce MSC-γ immunogenicity would facilitate "off-the-shelf" MSC therapy to efficiently inhibit alloimmune rejection and promote tissue repair in allotransplantation. In this review, we examine the potential benefits of MSC therapy in the context of allotransplantation. We also discuss the use of autologous and allogeneic MSC and the issues associated with their immunogenicity in vivo, with particular focus on the use of enhanced MSC-γ cellular therapy.
Collapse
Affiliation(s)
- Kisha Nandini Sivanathan
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005, South Australia, Australia,
| | | | | | | | | |
Collapse
|
63
|
Thude H, Kramer K, Koch M, Peine S, Sterneck M, Nashan B. Lack of association between CD40 polymorphisms and acute rejection in German liver transplant recipients. Hum Immunol 2014; 75:1123-7. [PMID: 25305459 DOI: 10.1016/j.humimm.2014.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 01/15/2023]
Abstract
CD40 and its ligand, CD154, are major costimulatory molecules whose interactions are important in alloreactive transplant rejection. The aim of this study was to examine the association of CD40 polymorphisms with the susceptibility to acute rejection episodes in liver transplantation. In total, 112 liver transplant recipients with biopsy proven acute rejections (BPAR), 97 without BPAR (WBPAR), and 112 healthy control individuals were enrolled in the study. Two single nucleotide polymorphisms (SNPs) of CD40 gene (rs1883832 and rs4810485) were genotyped by polymerase chain reaction-allele specific restriction enzyme analysis (PCR-ASRA). Both SNPs has been tested for a recessive and a dominant model. No significant differences were found in the genotype and allele frequencies of the SNPs rs1883832 and rs4810485 between BPAR liver recipients and WBPAR recipients. Our results do not suggest an important role of tested CD40 SNPs in the susceptibility to acute liver transplant rejection in a Caucasian population.
Collapse
Affiliation(s)
- Hansjörg Thude
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| | - Kathrin Kramer
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| | - Martina Koch
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| | - Sven Peine
- University Medical Center Hamburg-Eppendorf, Institute for Transfusion Medicine, Martinistraße 52, 20246 Hamburg, Germany
| | - Martina Sterneck
- University Medical Center Hamburg-Eppendorf, Department of Medicine, Martinistraße 52, 20246 Hamburg, Germany
| | - Björn Nashan
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| |
Collapse
|
64
|
Overexpression of Jagged-1 combined with blockade of CD40 pathway prolongs allograft survival. Immunol Cell Biol 2014; 93:213-7. [PMID: 25287443 DOI: 10.1038/icb.2014.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 08/25/2014] [Accepted: 09/03/2014] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) have the tolerogenic potential to regulate adaptive immunity and induce allografts acceptance. Here we investigated whether blockade of the CD40 pathway could enhance the immune tolerance induced by DC2.4 cells modified to express Jagged-1 (JAG1-DC) in heart transplantation. Results showed that JAG1-DC treatment combined with anti-CD40L monoclonal antibody (mAb) administration significantly prolonged cardiac allograft survival in mice, with long-term survival (>110 days) of 50% of the allografts in the recipients. The therapy specifically inhibited the immune response, induced alloantigen-specific T-cell hyporesponsiveness, upregulated transforming growth factor-β synthesis and increased the population of regulatory T cells (Tregs) driven by Jagged-1-Notch activation. These results highlight the potential application of gene therapy to induce alloantigen-specific Tregs effectively by providing the Jagged-1 stimulation.
Collapse
|
65
|
Karimi MH, Ebadi P, Pourfathollah AA. Association of cytokine/costimulatory molecule polymorphism and allograft rejection: a comparative review. Expert Rev Clin Immunol 2014; 9:1099-112. [PMID: 24168415 DOI: 10.1586/1744666x.2013.844462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
One reason for genetic variations among human individuals is SNP which may confer diverse disease susceptibility or resistance in a population. Genetic variations in a key immunoregulatory agent can manifest various immunological responses, such as graft rejection. In fact, the outcome of organ transplantation can be impacted by several genetic causes including polymorphisms in genes encoding cytokines and costimulatory molecules in the donor or recipient. Thus, it can be helpful to contemplate the SNPs relating to these immunological determinants in order to achieve an improved transplantation therapy.
Collapse
Affiliation(s)
- Mohammad H Karimi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | |
Collapse
|
66
|
Esposito P, Grosjean F, Rampino T, Libetta C, Gregorini M, Fasoli G, Marchi G, Sileno G, Montagna F, Dal Canton A. Costimulatory pathways in kidney transplantation: pathogenetic role, clinical significance and new therapeutic opportunities. Int Rev Immunol 2014; 33:212-233. [PMID: 24127878 DOI: 10.3109/08830185.2013.829470] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2013] [Indexed: 02/05/2023]
Abstract
Costimulatory pathways play a key role in immunity, providing the second signal required for a full activation of adaptive immune response. Different costimulatory families (CD28, TNF-related, adhesion and TIM molecules), characterized by structural and functional analogies, have been described. Costimulatory molecules modulate T cell activation, B cell function, Ig production, cytokine release and many other processes, including atherosclerosis. Patients suffering from renal diseases present significant alterations of the costimulatory pathways, which might make them particularly liable to infections. These alterations are further pronounced in patients undergoing kidney transplantation. In these patients, different costimulatory patterns have been related to distinct clinical features. The importance that costimulation has gained during the last years has led to development of several pharmacological approaches to modulate this critical step in the immune activation. Different drugs, mainly monoclonal antibodies targeting various costimulatory molecules (i.e. anti-CD80, CTLA-4 fusion proteins, anti-CD154, anti-CD40, etc.) were designed and tested in both experimental and clinical studies. The results of these studies highlighted some criticisms, but also some promising findings and now costimulatory blockade is considered a suitable strategy, with belatacept (a CTLA-4 fusion protein) being approved as the first costimulatory blocker for use in renal transplantation. In this review, we summarize the current knowledge on costimulatory pathways in the setting of kidney transplantation. We describe the principal costimulatory molecule families, their role and clinical significance in patients undergoing renal transplantation and the new therapeutic approaches that have been developed to modulate the costimulatory pathways.
Collapse
Affiliation(s)
- Pasquale Esposito
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico S. Matteo and University of Pavia , Pavia , Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Vergani A, Gatti F, Lee KM, D'Addio F, Tezza S, Chin M, Bassi R, Tian Z, Wu E, Maffi P, Ben Nasr M, Kim JI, Secchi A, Markmann JF, Rothstein DM, Turka LA, Sayegh MH, Fiorina P. TIM4 Regulates the Anti-Islet Th2 Alloimmune Response. Cell Transplant 2014; 24:1599-1614. [PMID: 24612609 DOI: 10.3727/096368914x678571] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The role of the novel costimulatory molecule TIM4 in anti-islet response is unknown. We explored TIM4 expression and targeting in Th1 (BALB/c islets into C57BL/6 mice) and Th2 (BALB/c islets into Tbet(-/-) C57BL/6 mice) models of anti-islet alloimmune response and in a model of anti-islet autoimmune response (diabetes onset in NOD mice). The targeting of TIM4, using the monoclonal antibody RMT4-53, promotes islet graft survival in a Th1 model, with 30% of the graft surviving in the long term; islet graft protection appears to be mediated by a Th1 to Th2 skewing of the immune response. Differently, in the Th2 model, TIM4 targeting precipitates graft rejection by further enhancing the Th2 response. The effect of anti-TIM4 treatment in preventing autoimmune diabetes was marginal with only minor Th1 to Th2 skewing. B-Cell depletion abolished the effect of TIM4 targeting. TIM4 is expressed on human B-cells and is upregulated in diabetic and islet-transplanted patients. Our data suggest a model in which TIM4 targeting promotes Th2 response over Th1 via B-cells. The targeting of TIM4 could become a component of an immunoregulatory protocol in clinical islet transplantation, aiming at redirecting the immune system toward a Th2 response.
Collapse
Affiliation(s)
- Andrea Vergani
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA.,Transplant Medicine, Ospedale San Raffaele, Milan, 20132, Italy
| | - Francesca Gatti
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA.,University of Salento, Lecce, 73100, Italy
| | - Kang M Lee
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Francesca D'Addio
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA.,Transplant Medicine, Ospedale San Raffaele, Milan, 20132, Italy
| | - Sara Tezza
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Melissa Chin
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Roberto Bassi
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Ze Tian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, 58104, USA
| | - Paola Maffi
- Transplant Medicine, Ospedale San Raffaele, Milan, 20132, Italy
| | - Moufida Ben Nasr
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - James I Kim
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Antonio Secchi
- Transplant Medicine, Ospedale San Raffaele, Milan, 20132, Italy.,Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - James F Markmann
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - David M Rothstein
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, US
| | - Laurence A Turka
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Mohamed H Sayegh
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Paolo Fiorina
- Transplantation Research Center, Division of Nephrology, Boston Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA.,Transplant Medicine, Ospedale San Raffaele, Milan, 20132, Italy
| |
Collapse
|
68
|
Yeung MY, Najafian N, Sayegh MH. Targeting CD28 to prevent transplant rejection. Expert Opin Ther Targets 2013; 18:225-42. [PMID: 24329604 DOI: 10.1517/14728222.2014.863875] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The pivotal role of costimulatory pathways in regulating T-cell activation versus tolerance has stimulated tremendous interest in their manipulation for therapeutic purposes. Of these, the CD28-B7 pathway is arguably the most important and best studied. Therapeutic targets of CD28 are currently used in the treatment of melanoma, autoimmune diseases and in transplantation. AREAS COVERED In this review, we summarize our current knowledge of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) signaling, and review the current state and challenges of harnessing them to promote transplant tolerance. EXPERT OPINION Despite the success of belatacept, a first-in-class CTLA-4 fusion protein now clinically used in transplantation, it is apparent that we have only scratched the surface in understanding the complexities of how costimulatory pathways modulate the immune system. Our initial assumption that positive costimulators activate effector T cells and prevent tolerance, while negative costimulators inhibit effector T cells and promote tolerance, is clearly an oversimplified view. Indeed, belatacept is not only capable of blocking deleterious CD28-B7 interactions that promote effector T-cell responses but can also have undesired effects on tolerogenic regulatory T-cell populations.
Collapse
Affiliation(s)
- Melissa Y Yeung
- Brigham and Women's Hospital, Transplantation Research Center, Harvard Medical School, Renal Division , Boston, MA , USA +1 617 525 8005 ; +1 617 732 5254 ;
| | | | | |
Collapse
|
69
|
Riella LV, Sayegh MH. T-cell co-stimulatory blockade in transplantation: two steps forward one step back! Expert Opin Biol Ther 2013; 13:1557-68. [PMID: 24083381 DOI: 10.1517/14712598.2013.845661] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The concern about nephrotoxicity with calcineurin inhibitors led to the search of novel agents for immunosuppression. Based on the requirement of T-cell co-stimulatory signals to fully activated naïve T cells, it became clear that blocking these pathways could be an appealing therapeutic target. However, some unexpected findings were noticed in the recent clinical trials of belatacept, including a higher rate of rejection, which warranted further investigation with some interesting concepts emerging from the bench. AREAS COVERED This article aims to review the literature of the B7:CD28 co-stimulatory blockade in transplantation, including the basic immunology behind its development, clinical application and potential limitations. EXPERT OPINION Targeting co-stimulatory pathways were found to be much more complex than initially anticipated due to the interplay between not only various co-stimulatory pathways but also various co-inhibitory ones. In addition, co-stimulatory signals have different roles in diverse immune cell types. Therefore, targeting CD28 ligands with cytotoxic T lymphocyte antigen-4 (CTLA4)-Ig may have some deleterious effects, including the inhibition of regulatory T cells, blockade of co-inhibitory signals (CTLA4) and promotion of Th17 cells. Co-stimulatory independence of memory T cells was another unforeseen limitation. Learning how to better integrate co-stimulatory targeting with other immunosuppressive agents will be critical for the improvement of long-term graft survival.
Collapse
Affiliation(s)
- Leonardo V Riella
- Brigham & Women's Hospital, Boston Children's Hospital, Harvard Medical School, Transplantation Research Center, Renal Division , 221 Longwood Ave, Boston MA 02115 , USA +1 617 732 5259 ; +1 617 732 5254 ;
| | | |
Collapse
|
70
|
Abstract
PURPOSE OF REVIEW Memory T cells present a different set of challenges to transplant patients; they are needed for protection against invading pathogens, especially under conditions of immunosuppression. But their presence also threatens transplant survival, as some of them are alloreactive. Efforts to resolve this paradox will be critical in the induction of transplant tolerance. RECENT FINDINGS There has been significant progress made in the past few years in the areas of population diversity of memory T cells, metabolic control of their induction, and mechanisms and pathways involved in memory cell exhaustion. Multiple targets on memory T cells have been identified, some of which are under vigorous testing in various transplant models. SUMMARY Memory T cells are both friends and foes to transplant patients, and tolerance strategies should selectively target alloreactive memory T cells and leave other memory cells unaltered. This situation remains a major challenge in the clinic.
Collapse
|
71
|
Abstract
T(h)9 cells are a new subset of helper T cells, and the signature cytokine for T(h)9 cells is IL-9. Both T(h)9 cells and T(h)9 products are implicated in multiple disease settings. Thus, a clear understanding of how T(h)9 cells are induced and controlled is an important and clinically relevant issue. There are different molecular pathways identified thus far in the induction of T(h)9 cells, and activation of such diverse pathways requires integration of signals from TGF-β and IL-4 cytokine receptors as well as costimulatory molecules. These signals converge on the induction of multiple transcription factors that collectively drive the development of T(h)9 cells.
Collapse
Affiliation(s)
- Picheng Zhao
- Immunobiology and Transplant Research, Houston Methodist Hospital and Methodist Hospital Research Institute, Texas Medical Center, 6670 Bertner Avenue, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
72
|
Thude H, Schipler AD, Treszl A, Peine S, Koch M, Sterneck M, Nashan B. Lack of association between CTLA-4 and PDCD1 polymorphisms and acute rejection in German liver transplant recipients. Hum Immunol 2013; 74:1041-5. [DOI: 10.1016/j.humimm.2013.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/23/2013] [Accepted: 04/10/2013] [Indexed: 11/28/2022]
|
73
|
Ezzelarab M, Zahorchak A, Lu L, Morelli A, Chalasani G, Demetris A, Lakkis F, Wijkstrom M, Murase N, Humar A, Shapiro R, Cooper D, Thomson A. Regulatory dendritic cell infusion prolongs kidney allograft survival in nonhuman primates. Am J Transplant 2013; 13:1989-2005. [PMID: 23758811 PMCID: PMC4070451 DOI: 10.1111/ajt.12310] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/03/2013] [Accepted: 04/16/2013] [Indexed: 01/25/2023]
Abstract
We examined the influence of regulatory dendritic cells (DCreg), generated from cytokine-mobilized donor blood monocytes in vitamin D3 and IL-10, on renal allograft survival in a clinically relevant rhesus macaque model. DCreg expressed low MHC class II and costimulatory molecules, but comparatively high levels of programmed death ligand-1 (B7-H1), and were resistant to pro-inflammatory cytokine-induced maturation. They were infused intravenously (3.5-10 × 10(6) /kg), together with the B7-CD28 costimulation blocking agent CTLA4Ig, 7 days before renal transplantation. CTLA4Ig was given for up to 8 weeks and rapamycin, started on Day -2, was maintained with tapering of blood levels until full withdrawal at 6 months. Median graft survival time was 39.5 days in control monkeys (no DC infusion; n = 6) and 113.5 days (p < 0.05) in DCreg-treated animals (n = 6). No adverse events were associated with DCreg infusion, and there was no evidence of induction of host sensitization based on circulating donor-specific alloantibody levels. Immunologic monitoring also revealed regulation of donor-reactive memory CD95(+) T cells and reduced memory/regulatory T cell ratios in DCreg-treated monkeys compared with controls. Termination allograft histology showed moderate combined T cell- and Ab-mediated rejection in both groups. These findings justify further preclinical evaluation of DCreg therapy and their therapeutic potential in organ transplantation.
Collapse
Affiliation(s)
- M. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A.F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - L. Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A.E. Morelli
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - G. Chalasani
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.J. Demetris
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - F.G. Lakkis
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M. Wijkstrom
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - N. Murase
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A. Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - R. Shapiro
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - D.K.C. Cooper
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A.W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: Angus W. Thomson, PhD DSc, University of Pittsburgh School of Medicine, 200 Lothrop Street, W1540 BST, Pittsburgh, PA 15261, Phone: (412) 624-6392,
| |
Collapse
|
74
|
Krill KT, Csencsits-Smith K, Wood SC, Faust S, Lu G, Bishop DK. Glucocorticoid-induced TNFR-related protein stimulation reverses cardiac allograft acceptance induced by CD40-CD40L blockade. Clin Dev Immunol 2013; 2013:986859. [PMID: 23690829 PMCID: PMC3652106 DOI: 10.1155/2013/986859] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/14/2013] [Indexed: 12/16/2022]
Abstract
CD40-CD40L blockade has potent immunosuppressive effects in cardiac allograft rejection but is less effective in the presence of inflammatory signals. To better understand the factors that mediate CD40-CD40L blockade-resistant rejection, we studied the effects of stimulation through glucocorticoid-induced TNFR-related protein (GITR), a costimulatory protein expressed by regulatory and effector T cells. Stimulation of CD40-/- or wild-type recipient mice treated with anti-CD40L mAb (WT+anti-CD40L) and with agonistic anti-GITR mAb resulted in cardiac allograft rejection. GITR stimulation did not induce rejection once long-term graft acceptance was established. In vitro, GITR stimulation increased proliferation of effector T cells and decreased regulatory T cell (Treg) differentiation in both treatment groups. GITR-stimulated CD40-/- recipients rejected their allografts more rapidly compared to GITR-stimulated WT+anti-CD40L recipients, and this rejection, characterized by a robust Th2 response and significant eosinophilic infiltrate, could be mediated by CD4+ T cells alone. In contrast, both CD4+ and CD8+ T cells were required to induce rejection in GITR-stimulated WT+anti-CD40L-treated recipients, and the pathology of rejection was less severe. Hence, early GITR stimulation could initiate graft rejection despite CD40 deficiency or anti-CD40L mAb treatment, though the recipient response was dependent on the mechanism of CD40-CD40L disruption.
Collapse
Affiliation(s)
- Kenneth T. Krill
- Program in Cellular and Molecular Biology, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
| | - Keri Csencsits-Smith
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, TX 77030, USA
| | - Sherri C. Wood
- Section of Transplantation Surgery, Department of Surgery, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
| | - Susan Faust
- Program in Cellular and Molecular Biology, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
- Section of Transplantation Surgery, Department of Surgery, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
| | - Guanyi Lu
- Section of Transplantation Surgery, Department of Surgery, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
| | - D. Keith Bishop
- Program in Cellular and Molecular Biology, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
- Section of Transplantation Surgery, Department of Surgery, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
- Graduate Program in Immunology, University of Michigan, Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
75
|
Abstract
PURPOSE OF REVIEW In June 2011 the US Food and Drug Administration approved belatacept (Nulojix; Bristol-Myers Squibb, Princeton, New Jersey, USA) for the prophylaxis of organ rejection in adult kidney transplant recipients. This review will discuss the use of belatacept for the prevention of acute rejection as part of a maintenance immunosuppression regimen. RECENT FINDINGS Belatacept is a selective costimulation blocker designed to provide effective immunosuppression while avoiding the toxicities associated with calcineurin inhibitors. Phase 3 trial data have demonstrated that belatacept is noninferior to cyclosporine in 1-year patient and allograft survival. Three-year data demonstrate an ongoing improvement in mean measured glomerular filtration rate in belatacept-treated versus cyclosporine-treated patients. Overall, there seemed to be an improvement in cardiometabolic parameters in patients treated with belatacept compared with cyclosporine. There was a trend toward higher rates of early rejection episodes in patients treated with belatacept. One safety issue that must be considered when using belatacept is the potential for increased risk of posttransplant lymphoproliferative disease, especially in Epstein-Barr virus-seronegative recipients or patients treated with lymphocyte-depleting agents. SUMMARY Belatacept is the first new agent available in kidney transplant that may achieve the goal of improved long-term renal function.
Collapse
|
76
|
Memory T cells and their exhaustive differentiation in allograft tolerance and rejection. Curr Opin Organ Transplant 2013; 17:15-9. [PMID: 22186090 DOI: 10.1097/mot.0b013e32834ee443] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Memory T cells have emerged as a major threat to transplant survival; they are well equipped and well positioned to respond to antigens in an accelerated fashion. They participate in transplant rejection and resist interventions that usually contain naïve T cells. Thus, the means to prevent memory T cells from attacking allotransplants are an important issue in transplantation. RECENT FINDINGS Recent studies in other models suggest that effector T cells, which include both freshly activated T cells and memory T cells, can acquire 'an exhausted phenotype' in that they progressively lose their effector activities. This response is highly regulated, antigen specific, and driven primarily by antigen persistence. This exhausted phenotype has not been carefully explored in transplant models, and its role in transplant survival remains largely unknown. SUMMARY Studies of T-cell exhaustion may reveal additional facets of the fundamental mechanisms of transplant survival. T-cell exhaustion may be an alternative way of preventing memory development. Future studies are needed to further improve our understanding of T-cell exhaustion in transplantation.
Collapse
|
77
|
Abstract
PURPOSE OF REVIEW Achieving allograft tolerance is the holy grail of transplantation. However, tolerance and rejection are two extreme ends of a scale that can be tipped in either direction. We review the novel effector and regulatory mechanisms involved and factors that tip the balance in favor of rejection or regulation. RECENT FINDINGS It is increasingly recognized that established T-cell phenotypes could change their commitments. New data point to the plasticity of Th17 cells in vivo with a reciprocal balance of Th17 cells and regulatory T cells (Tregs) driven by the local cytokine environment. Treg-cell profiles have been linked to acute and chronic allograft outcomes, and emerging data also indicate a novel role of a regulatory B-cell population. Current research efforts are looking into factors that tip the balance toward allograft tolerance by targeting cytokines, novel costimulatory pathways such as T-cell immunoglobulin mucin molecules, and components of innate immunity, particularly dendritic cells. SUMMARY The balance of effector and regulatory mechanisms contributing to allograft outcome is very complex. It is likely that targeting multiple pathways will be required to achieve tolerance. Further studies are warranted to define this balance and identify optimal combination of therapeutic interventions.
Collapse
|
78
|
B7h (ICOS-L) maintains tolerance at the fetomaternal interface. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:2204-13. [PMID: 23578385 DOI: 10.1016/j.ajpath.2013.02.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 01/28/2013] [Accepted: 02/12/2013] [Indexed: 11/24/2022]
Abstract
In a successful pregnancy, the semiallogeneic fetus is not rejected by the maternal immune system, which implies tolerance mechanisms protecting fetal tissues from maternal immune attack. Here we report that the ICOS-B7h costimulatory pathway plays a critical role in maintaining the equilibrium at the fetomaternal interface. Blockade of this pathway increased fetal resorption and decreased fetal survival in an allogeneic pregnancy model (CBA female × B6 male). Locally in the placenta, levels of regulatory markers such as IDO and TGF-β1 were reduced after anti-B7h monoclonal antibody treatment, whereas levels of effector cytokines (eg, IFN-γ) were significantly increased. In secondary lymphoid organs, enhanced IFN-γ and granzyme B production (predominantly by CD8(+) T cells) was observed in the anti-B7h-treated group. The deleterious effect of B7h blockade in pregnancy was maintained only in CD4 knockout mice, not in CD8 knockout mice, which suggests a role for CD8(+) T cells in immune regulation by the ICOS-B7h pathway. In accord, regulatory CD8(+) T cells (in particular, CD8(+)CD103(+) cells) were significantly decreased after anti-B7h monoclonal antibody treatment, and adoptive transfer of this subset abrogated the deleterious effect of B7h blockade in fetomaternal tolerance. Taken together, these data support the hypothesis that B7h blockade abrogates tolerance at the fetomaternal interface by enhancing CD8(+) effector response and reducing local immunomodulation mediated by CD8(+) regulatory T cells.
Collapse
|
79
|
del Rio ML, Schneider P, Fernandez-Renedo C, Perez-Simon JA, Rodriguez-Barbosa JI. LIGHT/HVEM/LTβR interaction as a target for the modulation of the allogeneic immune response in transplantation. Am J Transplant 2013; 13:541-51. [PMID: 23356438 DOI: 10.1111/ajt.12089] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/12/2012] [Accepted: 11/30/2012] [Indexed: 01/25/2023]
Abstract
The exchange of information during interactions of T cells with dendritic cells, B cells or other T cells regulates the course of T, B and DC-cell activation and their differentiation into effector cells. The tumor necrosis factor superfamily member LIGHT (homologous to lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes) is transiently expressed upon T cell activation and modulates CD8 T cell-mediated alloreactive responses upon herpes virus entry mediator (HVEM) and lymphotoxin β receptor (LTβR) engagement. LIGHT-deficient mice, or WT mice treated with LIGHT-targeting decoy receptors HVEM-Ig, LTβR-Ig or sDcR3-Ig, exhibit prolonged graft survival compared to untreated controls, suggesting that LIGHT modulates the course and severity of graft rejection. Therefore, targeting the interaction of LIGHT with HVEM and/or LTβR using recombinant soluble decoy receptors or monoclonal antibodies represent an innovative therapeutic strategy for the prevention and treatment of allograft rejection and for the promotion of donor-specific tolerance.
Collapse
Affiliation(s)
- M-L del Rio
- Transplantation Immunobiology Section, Institute of Biomedicine, University of Leon, Leon, Spain
| | | | | | | | | |
Collapse
|
80
|
Abstract
T cells must be activated before they can elicit damage to allografts, through interaction of their T cell receptor (TCR) with peptide-MHC complex and through accessory molecules. Signaling through accessory molecules or costimulatory molecules is a critical way for the immune system to fine tune T cell activation. An emerging therapeutic strategy is to target selective molecules involved in the process of T cell activation using biologic agents, which do not impact TCR signaling, thus only manipulating the T cells, which recognize alloantigen. Costimulatory receptors and their ligands are attractive targets for this strategy and could be used both to prevent acute graft rejection as well as for maintenance immunosuppression. Therapeutic agents targeting costimulatory molecules, notably belatacept, have made the progression from the bench, through nonhuman primate studies and into the clinic. This overview describes some of the most common costimulatory molecules, their role in T cell activation, and the development of reagents, which target these pathways and their efficacy in transplantation.
Collapse
Affiliation(s)
| | | | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU UK
| |
Collapse
|
81
|
Li W, Li B, Li S. Adenovirus mediated CTLA4Ig transgene therapy alleviates abortion by inhibiting spleen lymphocyte proliferation and regulating apoptosis in the feto-placental unit. J Reprod Immunol 2013; 97:167-74. [PMID: 23433910 DOI: 10.1016/j.jri.2013.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/10/2013] [Accepted: 01/17/2013] [Indexed: 12/28/2022]
Abstract
Pregnancy is thought to be a state of immunological tolerance. The mechanisms underlying this phenomenon are still poorly understood. In our previous study, adenovirus mediated CTLA4Ig transgene (Ad-CTLA4Ig) therapy was demonstrated to improve pregnancy outcome in an abortion-prone mouse model by skewing the Th2/Th1 cytokine balance, expanding peripheral CD4(+) CD25(+) regulatory T cell populations and inducing indoleamine 2,3 dioxygenase (IDO) mRNA expression at the maternal-fetal interface. However, it is still not clear whether other mechanisms are involved in the protective effect of CTLA-4 on pregnancy outcome in abortion-prone matings. In this study, we focused on the effect of CTLA4Ig on spleen lymphocyte proliferation and apoptosis at the maternal-fetal interface. We demonstrated that Ad-CTLA4Ig therapy inhibited the proliferation of CBA/J splenocytes and IL-2 secretion in response to DBA/2 stimulator cells in the abortion-prone mice model. Ad-CTLA4Ig therapy also skewed cytokine production toward a Th2 bias and regulated the expression of anti-apoptosis factor Bcl-2 and pro-apoptosis factor Bax at the maternal-fetal interface. However, it did not influence the apoptosis and cell cycles of splenocytes in pregnant CBA/J mice. On the whole, these findings indicated that Ad-CTLA4Ig therapy could ameliorate the outcome of spontaneous abortion by inhibiting proliferation of maternal spleen lymphocytes and regulating apoptosis in the feto-placental unit.
Collapse
Affiliation(s)
- Weihong Li
- Assisted Reproductive Center, The First affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | |
Collapse
|
82
|
Ueno T, Yeung MY, McGrath M, Yang S, Zaman N, Snawder B, Padera RF, Magee CN, Gorbatov R, Hashiguchi M, Azuma M, Freeman GJ, Sayegh MH, Najafian N. Intact B7-H3 signaling promotes allograft prolongation through preferential suppression of Th1 effector responses. Eur J Immunol 2013; 42:2343-53. [PMID: 22733595 DOI: 10.1002/eji.201242501] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ligands of the B7 family provide both positive and negative costimulatory signals to the CD28 family of receptors on T lymphocytes, the balance of which determines the immune response. B7-H3 is a member of the B7 family whose function in T-cell activation has been the subject of some controversy: in autoimmunity and tumor immunity, it has been described as both costimulatory and coinhibitory, while in transplantation, B7-H3 signaling is thought to contribute to graft rejection. However, we now demonstrate results to the contrary. Signaling through a putative B7-H3 receptor prolonged allograft survival in a fully MHC-mismatched cardiac model and promoted a shift toward a Th2 milieu; conversely, B7-H3 blockade, achieved by use of a blocking antibody, resulted in accelerated rejection, an effect associated with enhanced IFN-γ production. Finally, graft prolongation achieved by CTLA4 Ig was shortened both by B7-H3 blockade and the absence of recipient B7-H3. These findings suggest a coinhibitory role for B7-H3. However, experience with other CD28/B7 family members suggests that immune redundancy plays a crucial role in determining the functions of various pathways. Given the abundance of conflicting data, it is plausible that, under differing conditions, B7-H3 may have both positive and negative costimulatory functions.
Collapse
Affiliation(s)
- Takuya Ueno
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Dave SD, Vanikar A, Trivedi HL, Gumber MR, Patel HV, Shah PR, Kute VB. Stem cells versus donor specific transfusions for tolerance induction in living donor renal transplantation: a single-center experience. Transplantation 2013; 95:155-160. [PMID: 23263505 DOI: 10.1097/tp.0b013e3182752bcc] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND We undertook this study to define the role of stem cell transplantation (SCT) versus donor-specific transfusion (DST) in tolerance induction and sustenance in living donor renal transplantation (LDRT). METHODS In this prospective three-armed trial in LDRT with 13 patients each in demographically well-balanced groups, tolerance induction protocol (TIP) was used with SCT in group 1, DST in group 2, and no induction in group 3. Tolerance induction protocol consisted of SCT/DST under conditioning with bortezomib, methylprednisone, rituximab, and rabbit antithymocyte globulin. Transplantation was performed with prednisone in groups 1 and 2 and with triple immunosuppression in group 3, if lymphocyte/flow crossmatch was negative; and if donor-specific antibodies (DSAs) were absent in the first 2 groups. Posttransplant monitoring included serum creatinine (SCr), peripheral T-regulatory cells (pTregs)(127/CD4+/25), and DSA for groups 1 and 2; DSA was eliminated in group 3. Rescue IS was started with rise of SCr/DSA/ rejection. RESULTS Tolerance induction protocol was safe. Over a mean follow-up of 2 years, no patient/graft was lost in groups 1 and 2. One patient of group 3 lost graft to noncompliance. Protocol biopsies were unremarkable. Rejections were noted in six patients of group 1, five of group 2, and seven of group 3. Donor-specific antibodies were elevated in three patients of both groups. Mean SCr of all groups was similar; however, pTregs were increased posttransplant in groups 1 and 2 versus group 3. Group 1 had sustained rise in pTregs. CONCLUSION Stem cell transplantation and DST are useful for immunosuppression minimization in LDRT with sustained generation of pTregs with SCT.
Collapse
Affiliation(s)
- Shruti D Dave
- Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre (IKDRC)-Dr. H.L. Trivedi Institute of Transplantation Sciences (ITS), Civil Hospital Campus, Asarwa, Ahmedabad, Gujarat, India.
| | | | | | | | | | | | | |
Collapse
|
84
|
D’Addio F, Boenisch O, Magee CN, Yeung MY, Yuan X, Mfarrej B, Vergani A, Ansari MJ, Fiorina P, Najafian N. Prolonged, low-dose anti-thymocyte globulin, combined with CTLA4-Ig, promotes engraftment in a stringent transplant model. PLoS One 2013; 8:e53797. [PMID: 23326509 PMCID: PMC3542267 DOI: 10.1371/journal.pone.0053797] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/03/2012] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Despite significant nephrotoxicity, calcineurin inhibitors (CNIs) remain the cornerstone of immunosuppression in solid organ transplantation. We, along with others, have reported tolerogenic properties of anti-thymocyte globulin (ATG, Thymoglobulin®), evinced by its ability both to spare Tregs from depletion in vivo and, when administered at low, non-depleting doses, to expand Tregs ex vivo. Clinical trials investigating B7/CD28 blockade (LEA29Y, Belatacept) in kidney transplant recipients have proven that the replacement of toxic CNI use is feasible in selected populations. METHODS Rabbit polyclonal anti-murine thymocyte globulin (mATG) was administered as induction and/or prolonged, low-dose therapy, in combination with CTLA4-Ig, in a stringent, fully MHC-mismatched murine skin transplant model to assess graft survival and mechanisms of action. RESULTS Prolonged, low-dose mATG, combined with CTLA4-Ig, effectively promotes engraftment in a stringent transplant model. Our data demonstrate that mATG achieves graft acceptance primarily by promoting Tregs, while CTLA4-Ig enhances mATG function by limiting activation of the effector T cell pool in the early stages of treatment, and by inhibiting production of anti-rabbit antibodies in the maintenance phase, thereby promoting regulation of alloreactivity. CONCLUSION These data provide the rationale for development of novel, CNI-free clinical protocols in human transplant recipients.
Collapse
Affiliation(s)
- Francesca D’Addio
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Transplantation Medicine Division, San Raffaele Hospital, Milan, Italy
| | - Olaf Boenisch
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ciara N. Magee
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Melissa Y. Yeung
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xueli Yuan
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Bechara Mfarrej
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Andrea Vergani
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Transplantation Medicine Division, San Raffaele Hospital, Milan, Italy
| | - Mohammed Javeed Ansari
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Divisions of Nephrology and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Paolo Fiorina
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- Transplantation Medicine Division, San Raffaele Hospital, Milan, Italy
| | - Nader Najafian
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
85
|
Abstract
Rejection is the major barrier to successful transplantation and usually results from the integration of multiple mechanisms. Activation of elements of the innate immune system, triggered as a consequence of tissue injury sustained during cell isolation or organ retrieval as well as ischemia-reperfusion, will initiate and amplify the adaptive response. For cell mediated rejection, T cells require multiple signals for activation, the minimum being two signals; antigen recognition and costimulation. The majority of B cells require help from T cells to initiate alloantibody production. Antibodies reactive to donor HLA molecules, minor histocompatibility antigens, endothelial cells, red blood cells, or autoantigens can trigger or contribute to rejection early as well as late after transplantation.
Collapse
Affiliation(s)
- Kathryn J Wood
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
86
|
Tumor dormancy and cancer stem cells: two sides of the same coin? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:145-79. [PMID: 23143979 DOI: 10.1007/978-1-4614-1445-2_8] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Increasing evidence suggests that tumor dormancy represents an important mechanism underlying the observed failure of existing therapeutic modalities to fully eradicate cancers. In addition to its more established role in maintaining minimal residual disease after treatment, dormancy might also critically contribute to early stages of tumor development and the formation of clinically undetectable micrometastatic foci. There are striking parallels between the concept of tumor dormancy and the cancer stem cell (CSC) theory of tumor propagation. For instance, the CSC hypothesis similarly predicts that a subset of self-renewing cancer cells-that is CSCs-is responsible for tumor initiation, bears the preferential ability to survive tumor therapy, and persists long term to ultimately cause delayed cancer recurrence and metastatic progression. Additionally, many of the biological mechanisms involved in controlling the dormant state of a tumor can also govern CSC behavior, including cell cycle modifications, alteration of angiogenic processes, and modulation of antitumor immune responses. In fact, quiescence and immune escape are emerging hallmark features of at least some CSCs, indicating significant overlap between dormant cancer populations and CSCs. Herein, we crucially dissect whether CSCs occupy specific roles in orchestrating the switch between dormancy and exuberant tumor growth. We elucidate how recently uncovered CSC biological features could enable these cells to evade immunologic clearance and regulate cancer expansion, relapse, and progression. We propose that the study of CSC immunobiological pathways holds the promise to critically advance our understanding of the processes mediating tumor dormancy. Ultimately, such research endeavors could unravel novel therapeutic avenues that efficiently target both proliferating and dormant CSCs to minimize the risk of tumor recurrence in cancer patients.
Collapse
|
87
|
Wojciechowski D, Vincenti F. Belatacept for prevention of acute rejection in adult patients who have had a kidney transplant: an update. Biologics 2012; 6:385-93. [PMID: 23152668 PMCID: PMC3496190 DOI: 10.2147/btt.s23561] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In June 2011, the US Food and Drug Administration approved belatacept for the prophylaxis of organ rejection in adult kidney transplant recipients. This review discusses the use of belatacept for the prevention of acute rejection as part of a maintenance immunosuppression regimen. Belatacept is a selective costimulation blocker designed to provide effective immunosuppression while avoiding the toxicities associated with calcineurin inhibitors. Phase III trial data have demonstrated that belatacept is noninferior to cyclosporine in 1-year patient and allograft survival. Three-year data demonstrate an ongoing improvement in mean measured glomerular filtration rate in belatacept-treated versus cyclosporine-treated patients. However, the rate of acute rejection was higher in belatacept-treated patients compared with cyclosporine. Specifically, there was a higher incidence of Banff type II rejections in patients treated with belatacept. Despite the higher Banff grade, rejections on belatacept were not associated with other factors associated with poor outcomes, such as the development of donor-specific antibodies or reduced estimated glomerular filtration rate. One safety issue that must be considered when using belatacept is the potential for increased risk of post-transplant lymphoproliferative disease. There were more cases of post-transplant lymphoproliferative disease in belatacept-treated patients, especially in recipients seronegative for Epstein–Barr virus or patients treated with lymphocyte-depleting agents. Therefore, belatacept can be recommended for use in Epstein–Barr virus antibody-positive recipients.
Collapse
|
88
|
Ippoliti G, D’Armini AM, Lucioni M, Marjieh M, Viganò M. Introduction to the use of belatacept: a fusion protein for the prevention of posttransplant kidney rejection. Biologics 2012; 6:355-62. [PMID: 23055693 PMCID: PMC3468025 DOI: 10.2147/btt.s27565] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The development of new immunosuppressive drugs for kidney transplantation resulted both in better short-term outcomes and in decreased metabolic, cardiovascular, and nephrotoxicity risk. Belatacept belongs to a new class of immunosuppressive drugs that selectively inhibits T-cell activation by preventing CD28 activation and by binding its ligands B7-1 and B7-2. The result is an inactivation of costimulatory pathways. A comparative analysis of the BENEFIT and BENEFIT-EXT datasets showed belatacept regimens resulted in better cardiovascular and metabolic risk profiles than did cyclosporin A (CsA) regimens: belatacept likewise outperformed CsA in terms of lower blood pressure and serum lipids and less new onset diabetes after transplantation. About 20% of belatacept-treated patients developed adverse effects which included anemia, pyrexia, neutropenia, diarrhea, urinary tract infection, headache, and peripheral edema. At present, belatacept does not seem to predispose patients to a higher rate of infection than CsA maintenance immunosuppression. The risk of posttransplant lymphoproliferative diseases was higher in Epstein-Barr virus (EBV)-seronegative patients than in EBV-seropositive patients, but the risk may be reduced by use of a less intensive regimen and avoidance of EBV-negative patients and of patients whose pretransplant EBV serology is unknown. Belatacept provides a new option for immunosuppressive therapy in kidney transplantation, but needs further evaluation in terms of the late effects that may derive from prolonged blockage of the costimulatory system and the induction of tolerance status.
Collapse
Affiliation(s)
- Giovanbattista Ippoliti
- UO Medicina Interna, Policlinico di Monza, Monza, Italy
- Department of Surgical Sciences, Charles Dubost Transplant Center, Fondazione IRCCS San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Andrea Maria D’Armini
- Department of Surgical Sciences, Charles Dubost Transplant Center, Fondazione IRCCS San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Marco Lucioni
- Anatomic Pathology, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Mazen Marjieh
- UO Medicina Interna, Policlinico di Monza, Monza, Italy
| | - Mario Viganò
- Department of Surgical Sciences, Charles Dubost Transplant Center, Fondazione IRCCS San Matteo Hospital, University of Pavia, Pavia, Italy
| |
Collapse
|
89
|
Xiao X, Balasubramanian S, Liu W, Chu X, Wang H, Taparowsky EJ, Fu YX, Choi Y, Walsh MC, Li XC. OX40 signaling favors the induction of T(H)9 cells and airway inflammation. Nat Immunol 2012; 13:981-90. [PMID: 22842344 PMCID: PMC3806044 DOI: 10.1038/ni.2390] [Citation(s) in RCA: 191] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 07/06/2012] [Indexed: 12/11/2022]
Abstract
The mechanisms that regulate the T(H)9 subset of helper T cells and diseases mediated by T(H)9 cells remain poorly defined. Here we found that the costimulatory receptor OX40 was a powerful inducer of T(H)9 cells in vitro and T(H)9 cell-dependent airway inflammation in vivo. In polarizing conditions based on transforming growth factor-β (TGF-β), ligation of OX40 inhibited the production of induced regulatory T cells and the T(H)17 subset of helper T cells and diverted CD4(+)Foxp3(-) T cells to a T(H)9 phenotype. Mechanistically, OX40 activated the ubiquitin ligase TRAF6, which triggered induction of the kinase NIK in CD4(+) T cells and the noncanonical transcription factor NF-κB pathway; this subsequently led to the generation of T(H)9 cells. Thus, our study identifies a previously unknown mechanism for the induction of T(H)9 cells and may have important clinical implications in allergic inflammation.
Collapse
Affiliation(s)
- Xiang Xiao
- Transplant Research Center, Brigham and Women’s hospital and Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Savithri Balasubramanian
- The Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Wentao Liu
- Transplant Research Center, Brigham and Women’s hospital and Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Xiufeng Chu
- Transplant Research Center, Brigham and Women’s hospital and Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Haibin Wang
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | | | - Yang-Xin Fu
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew C. Walsh
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Xian Chang Li
- Transplant Research Center, Brigham and Women’s hospital and Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
90
|
Abstract
Understanding immunoregulatory mechanisms is essential for the development of novel interventions to improve long-term allograft survival. Programmed death 1 (PD-1) and its ligands, PD-L1 and PD-L2, have emerged as critical inhibitory signaling pathways that regulate T cell response and maintain peripheral tolerance. PD-1 signaling inhibits alloreactive T cell activation, and can promote induced regulatory T cell development. Furthermore, the upregulation of PD-L1 on nonhematopoietic cells of the allograft may actively participate in the inhibition of immune responses and provide tissue-specific protection. In murine transplant models, this pathway has been shown to be critical for the induction and maintenance of graft tolerance. In this review, we discuss the current knowledge of the immunoregulatory functions of PD-1 and its ligands and their therapeutic potential in transplantation.
Collapse
Affiliation(s)
- Leonardo V. Riella
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital & Children's Hospital Boston, Harvard Medical School, Boston, MA,Corresponding author: Leonardo V. Riella, MD, PhD Transplantation Research Center Brigham and Women's Hospital and Children's Hospital Boston 221 Longwood Ave, Boston MA 02115, USA. Tel: 617-732-5898; Fax: 617-732-5254
| | - Alison M. Paterson
- Department of Microbiology and Immunobiology, Harvard Medical School and Department of Pathology, Brigham & Women's Hospital, Boston, MA
| | - Arlene H. Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School and Department of Pathology, Brigham & Women's Hospital, Boston, MA
| | - Anil Chandraker
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital & Children's Hospital Boston, Harvard Medical School, Boston, MA
| |
Collapse
|
91
|
Magee CN, Boenisch O, Najafian N. The role of costimulatory molecules in directing the functional differentiation of alloreactive T helper cells. Am J Transplant 2012; 12:2588-600. [PMID: 22759274 PMCID: PMC3459149 DOI: 10.1111/j.1600-6143.2012.04180.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Costimulatory molecules are a heterogenous group of cell surface molecules that act to amplify or counteract the initial activating signals provided to T cells from the T cell receptor following its interaction with an antigen/major histocompatibility complex, thereby influencing T cell differentiation and fate. Although costimulation was previously thought to be indispensable for T cell activation at all stages of development, it is now known that the requirements for costimulation, and the costimulatory molecules involved, vary according to the stage of T cell differentiation. The ability to influence T cell fate is of paramount interest in the field of transplantation as we seek therapeutic options that inhibit detrimental alloimmune responses whilst simultaneously promoting allograft tolerance. As with many immune mechanisms, there is a degree of functional overlap between certain costimulatory molecules, whereas some have diametrically opposite effects on different T cell subsets despite sharing common ligands. This is a critical point when considering these molecules as therapeutic targets in transplantation, as blockade of a costimulatory pathway, although desirable in itself, may prevent the ligation of an essential regulatory coinhibitory molecule. This review discusses the T helper cell lineages pertinent to transplantation and the costimulatory molecules involved in their differentiation.
Collapse
Affiliation(s)
- Ciara N. Magee
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Olaf Boenisch
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Nader Najafian
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
92
|
Charbonnier LM, Vokaer B, Lemaître PH, Field KA, Leo O, Le Moine A. CTLA4-Ig restores rejection of MHC class-II mismatched allografts by disabling IL-2-expanded regulatory T cells. Am J Transplant 2012; 12:2313-21. [PMID: 22759373 DOI: 10.1111/j.1600-6143.2012.04184.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Allograft acceptance and tolerance can be achieved by different approaches including inhibition of effector T cell responses through CD28-dependent costimulatory blockade and induction of peripheral regulatory T cells (Tregs). The observation that Tregs rely upon CD28-dependent signals for development and peripheral expansion, raises the intriguing possibility of a counterproductive consequence of CTLA4-Ig administration on tolerance induction. We have investigated the possible negative effect of CTLA4-Ig on Treg-mediated tolerance induction using a mouse model of single MHC class II-mismatched skin grafts in which long-term acceptance was achieved by short-term administration of IL-2/anti-IL-2 complex. CTLA4-Ig treatment was found to abolish Treg-dependent acceptance in this model, restoring skin allograft rejection and Th1 alloreactivity. CTLA4-Ig inhibited IL-2-driven Treg expansion, and prevented in particular the occurrence of ICOS(+) Tregs endowed with potent suppressive capacities. Restoring CD28 signaling was sufficient to counteract the deleterious effect of CTLA4-Ig on Treg expansion and functionality, in keeping with the hypothesis that costimulatory blockade inhibits Treg expansion and function by limiting the delivery of essential CD28-dependent signals. Inhibition of regulatory T cell function should therefore be taken into account when designing tolerance protocols based on costimulatory blockade.
Collapse
Affiliation(s)
- L-M Charbonnier
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium.
| | | | | | | | | | | |
Collapse
|
93
|
Dormond O, Dufour M, Seto T, Bruneau S, Briscoe DM. Targeting the intragraft microenvironment and the development of chronic allograft rejection. Hum Immunol 2012; 73:1261-8. [PMID: 22863981 DOI: 10.1016/j.humimm.2012.07.334] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 06/29/2012] [Accepted: 07/25/2012] [Indexed: 01/10/2023]
Abstract
In this review, we discuss a paradigm whereby changes in the intragraft microenvironment promote or sustain the development of chronic allograft rejection. A key feature of this model involves the microvasculature including (a) endothelial cell (EC) destruction, and (b) EC proliferation, both of which result from alloimmune leukocyte- and/or alloantibody-induced responses. These changes in the microvasculature likely create abnormal blood flow patterns and thus promote local tissue hypoxia. Another feature of the chronic rejection microenvironment involves the overexpression of vascular endothelial growth factor (VEGF). VEGF stimulates EC activation and proliferation and it has potential to sustain inflammation via direct interactions with leukocytes. In this manner, VEGF may promote ongoing tissue injury. Finally, we review how these events can be targeted therapeutically using mTOR inhibitors. EC activation and proliferation as well as VEGF-VEGFR interactions require PI-3K/Akt/mTOR intracellular signaling. Thus, agents that inhibit this signaling pathway within the graft may also target the progression of chronic rejection and thus promote long-term graft survival.
Collapse
Affiliation(s)
- Olivier Dormond
- The Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
94
|
Priyadharshini B, Greiner DL, Brehm MA. T-cell activation and transplantation tolerance. Transplant Rev (Orlando) 2012; 26:212-22. [PMID: 22074786 PMCID: PMC3294261 DOI: 10.1016/j.trre.2011.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 08/16/2011] [Accepted: 09/02/2011] [Indexed: 12/28/2022]
Abstract
Transplantation of allogeneic or "nonself" tissues stimulates a robust immune response leading to graft rejection, and therefore, most recipients of allogeneic organ transplants require the lifelong use of immune suppressive agents. Excellent outcomes notwithstanding, contemporary immunosuppressive medications are toxic, are often not taken by patients, and pose long-term risks of infection and malignancy. The ultimate goal in transplantation is to develop new treatments that will supplant the need for general immunosuppression. Here, we will describe the development and application of costimulation blockade to induce transplantation tolerance and discuss how the diverse array of signals that act on T cells will determine the balance between graft survival and rejection.
Collapse
Affiliation(s)
- Bhavana Priyadharshini
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Biotech 2, Worcester, MA 01605
| | - Dale L. Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Biotech 2, Worcester, MA 01605
| | - Michael A. Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Biotech 2, Worcester, MA 01605
| |
Collapse
|
95
|
CD28 family and chronic rejection: "to belatacept...And beyond!". J Transplant 2012; 2012:203780. [PMID: 22720132 PMCID: PMC3376773 DOI: 10.1155/2012/203780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 03/19/2012] [Accepted: 03/26/2012] [Indexed: 12/15/2022] Open
Abstract
Kidneys are one of the most frequently transplanted human organs. Immunosuppressive agents may prevent or reverse most acute rejection episodes; however, the graft may still succumb to chronic rejection. The immunological response involved in the chronic rejection process depends on both innate and adaptive immune response. T lymphocytes have a pivotal role in chronic rejection in adaptive immune response. Meanwhile, we aim to present a general overview on the state-of-the-art knowledge of the strategies used for manipulating the lymphocyte activation mechanisms involved in allografts, with emphasis on T-lymphocyte costimulatory and coinhibitory molecules of the B7-CD28 superfamily. A deeper understanding of the structure and function of these molecules improves both the knowledge of the immune system itself and their potential action as rejection inducers or tolerance promoters. In this context, the central role played by CD28 family, especially the relationship between CD28 and CTLA-4, becomes an interesting target for the development of immune-based therapies aiming to increase the survival rate of allografts and to decrease autoimmune phenomena. Good results obtained by the recent development of abatacept and belatacept with potential clinical use aroused better expectations concerning the outcome of transplanted patients.
Collapse
|
96
|
Zhang J, Miao Q, Yang Y, Xiao B, Liu B, Cao J, Hao XY, Wang SW, Guo SZ. Effect of combined OX40Ig and CTLA4Ig gene local transfer on allograft rejection and the underlying mechanisms. J Surg Res 2012; 178:949-58. [PMID: 22694937 DOI: 10.1016/j.jss.2012.05.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/26/2012] [Accepted: 05/09/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND OX40Ig and CTLA4Ig fusion proteins have been suggested to induce immune tolerance and prevent rejection in allografts. The present study aims to investigate and compare the effects of ex vivo combined OX40Ig and CTLA4Ig lentivirus-mediated gene transfer on the long-term survival of the graft, as well as potential underlying mechanisms. METHODS We ex vivo transferred Brown Norway rats' superficial groin free flap with lentivirus vectors expressing OX40Ig or CTLA4Ig, or OX40Ig and CTLA4Ig combined, and transplanted the free flaps to Lewis rats. Short-course rapamycin was administered after transfection and transplantation. RT-PCR and Western blot were employed to evaluate expression of OX40Ig and CTLA4Ig. We assessed the survival time of the grafts and the degree of acute graft rejection after indicated treatment. Mixed lymphocyte reaction, flow cytometry, and ELISA were also used to evaluate systemic immune reactions. RESULTS Ex vivo transfer of OX40Ig or CTLA4Ig lentivirus vectors led to local expression of corresponding mRNA and proteins in the donor flap without affecting other organs of the recipient. The graft survival time was significantly expanded and rejection was markedly attenuated after transfection. Mixed lymphocyte reaction, flow cytometry (CD4(+) and CD8(+) T lymphocyte proportions), and serum ELISA analysis (IL-2, IFN-γ, IL-4, and IL-10) also showed decreased immune response following transfection. Combined OX40Ig and CTLA4Ig transfer exerted superior effect on improving graft survival and preventing graft rejection, inhibiting the immune response and decreasing the production of proinflammatory cytokines, compared with singular transfer of either OX40Ig or CTLA4Ig. CONCLUSION Combined ex vivo transfer of OX40Ig and CTLA4Ig lentivirus vectors provided superior benefits on long-term survival and restoration of the graft through inhibiting immune response and decreasing the production of proinflammatory cytokines.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Riella LV, Liu T, Yang J, Chock S, Shimizu T, Mfarrej B, Batal I, Xiao X, Sayegh MH, Chandraker A. Deleterious effect of CTLA4-Ig on a Treg-dependent transplant model. Am J Transplant 2012; 12:846-55. [PMID: 22300534 DOI: 10.1111/j.1600-6143.2011.03929.x] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Blockade of the B7:CD28 costimulatory pathway has emerged as a promising therapy to prevent allograft rejection. However, results from the belatacept phase III clinical trial demonstrated a higher rejection rate when compared to cyclosporine, raising concern about potential deleterious effects of this agent. In this study, we investigated the consequences of B7:CD28 blockade by hCTLA4Ig on regulator T cell (Treg) generation in different major histocompatibility complex (MHC) mismatch transplant models. Administration of hCTLA4Ig significantly decreased the amount of Tregs in B6 WT animals and this effect was predominant in thymus-induced Tregs (Helios(+) ). Although hCTLA4Ig prevented rejection in a fully allogeneic mismatch model, it accelerated rejection in a MHC class-II mismatch model (MST = 26, p < 0.0001), in which long-term allograft survival is dependent on Tregs. This accelerated rejection was associated with a marked reduction in thymus-induced Tregs and led to a higher effector/regulatory T-cell ratio in secondary lymphoid organs and in the allograft. This study confirms the importance of the B7:CD28 pathway in Treg homeostasis in an in vivo transplant model and suggests that hCTLA4Ig therapy may be deleterious in circumstances where engraftment is dependent on Tregs.
Collapse
Affiliation(s)
- L V Riella
- Renal Division, Transplantation Research Center, Brigham and Women's Hospital, Children's Hospital Boston, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
Rejection is the major barrier to successful transplantation. The immune response to an allograft is an ongoing dialogue between the innate and adaptive immune system that if left unchecked will lead to the rejection of transplanted cells, tissues, or organs. Activation of elements of the innate immune system, triggered as a consequence of tissue injury sustained during cell isolation or organ retrieval and ischemia reperfusion, will initiate and amplify the adaptive response. T cells require a minimum of two signals for activation, antigen recognition, and costimulation. The activation requirements of naive T cells are more stringent than those of memory T cells. Memory T cells are present in the majority of transplant recipients as a result of heterologous immunity. The majority of B cells require help from T cells to initiate antibody production. Antibodies reactive to donor human leukocyte antigen molecules, minor histocompatibility antigens, endothelial cells, RBCs, or autoantigens can trigger or contribute to rejection early and late after transplantation. Antibody-mediated rejection triggered by alloantibody binding and complement activation is recognized increasingly as a significant contribution to graft loss. Even though one component of the immune system may dominate and lead to rejection being described in short hand as T cell or antibody mediated, it is usually multifactorial resulting from the integration of multiple mechanisms. Identifying the molecular pathways that trigger tissue injury, signal transduction and rejection facilitates the identification of targets for the development of immunosuppressive drugs.
Collapse
|
99
|
Gilson CR, Patel SR, Zimring JC. CTLA4-Ig prevents alloantibody production and BMT rejection in response to platelet transfusions in mice. Transfusion 2012; 52:2209-19. [PMID: 22321003 DOI: 10.1111/j.1537-2995.2011.03550.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Platelet (PLT) transfusions can induce humoral and cellular alloimmunity. HLA antibodies can render patients refractory to subsequent transfusion, and both alloantibodies and cellular alloimmunity can contribute to subsequent bone marrow transplant (BMT) rejection. Currently, there are no approved therapeutic interventions to prevent alloimmunization to PLT transfusions other than leukoreduction. Targeted blockade of T-cell costimulation has shown great promise in inhibiting alloimmunity in the setting of transplantation, but has not been explored in the context of PLT transfusion. STUDY DESIGN AND METHODS We tested the hypothesis that the costimulatory blockade reagent CTLA4-Ig would prevent alloreactivity against major and minor alloantigens on transfused PLTs. BALB/c (H-2(d)) mice and C57BL/6 (H-2(b)) mice were used as PLT donors and transfusion recipients, respectively. Alloantibodies were measured by indirect immunofluorescence using BALB/c PLTs and splenocytes as targets. BMTs were carried out under reduced-intensity conditioning using BALB.B (H-2(b) ) donors and C57BL/6 (H-2(b)) recipients to model HLA-identical transplants. Experimental groups were given CTLA4-Ig (before or after PLT transfusion) with control groups receiving isotype-matched antibody. RESULTS CTLA4-Ig abrogated both humoral alloimmunization (H-2(d) antibodies) and transfusion-induced BMT rejection. Whereas a single dose of CTLA4-Ig at time of transfusion prevented alloimmunization to subsequent PLT transfusions, administration of CTLA4-Ig after initial PLT transfusion was ineffective. Delaying treatment until after PLT transfusion failed to prevent BMT rejection. CONCLUSIONS These findings demonstrate a novel strategy using an FDA-approved drug that has the potential to prevent the clinical sequelae of alloimmunization to PLT transfusions.
Collapse
Affiliation(s)
- Christopher R Gilson
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Aflac Cancer Center and Blood Disorders Service, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | |
Collapse
|
100
|
Abstract
PURPOSE OF REVIEW The goal of this study is to review key recent findings related to the immunopathogenesis of hepatitis C virus (HCV) infection, especially in regards to T lymphocytes. It aims to complement other reviews in this issue on the roles of host genetics (IL-28B), acute HCV infection (when disease outcome is determined) and other factors that may influence fibrosis progression (microbial translocation). The main focus is on specific immunity and T cells in the context of success and failure to control viral infection. RECENT FINDINGS This review focuses on two areas of intense interest in the recent literature: the relationship between the human leukocyte antigen (HLA), class I-restricted T-cell responses and the evolution of the virus and the role of inhibitory markers on T cells in the immunopathogenesis of HCV. When appropriate, we compare findings from studies of HIV-specific immunity. SUMMARY From examining the virus and the mutational changes associated with T-cell responses and from analyzing the markers on T cells, there have been numerous advances in the understanding of immune evasion mechanisms employed by HCV.
Collapse
|