scholarly journals Novel Immunosuppressive Approach Attenuates Severe Aplastic Anemia in Mouse Lymphocyte Infusion AA Models

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2942-2942
Author(s):  
Jessica Lorente ◽  
Divya Kannegenti ◽  
Brandon Theall ◽  
Lisandra Hernandez ◽  
Esha Vallabhaneni ◽  
...  

Abstract Aplastic Anemia (AA) is an immune-mediated and life-threatening form of acquired bone marrow failure. AA ranges from moderate to severe AA, and is characterized by development and expansion of self-reactive effector T cells, which cause apoptosis of mature blood cells, progenitors and hematopoietic stem cells (HSCs). Current treatments for AA, which are not always effective or feasible, include immunosuppressive therapy (IST) and allogeneic HLA-identical sibling or well-matched unrelated donor BM transplant. Because the self-antigens triggering AA remain to be identified, mouse lymphocyte infusion models of AA with striking similarities to human AA have been developed utilizing alloantigen recognition. The AA in these models is induced by infusing lymph node cells (LNCs) from C57BL/6J mice into MHC partially mismatched F1 hybrid B6D2F1 or CByB6F1 recipients, or into minor-H antigen mismatched C.B10 recipients. The host mice develop SAA without any clinical signs of generalized GVHD, and characterized by BM infiltration and oligoclonal expansion of donor effector T cells, apoptosis of all host BM cells, severe BM aplasia and death within 3-5 weeks after LNC infusion. These preclinical mouse models represent a very useful in vivo system for testing new therapeutic approaches to treat and manage SAA. Activation of self-reactive T cells in human AA and alloreactive donor T cells in mouse AA models involves interaction of T cells with dendritic cells (DCs) as professional APCs. DCs express β2 integrin CD11b/CD18 (Mac-1), which plays an important role in inflammation, cell-mediated killing and cell activation. Notably, Mac-1 expressed on DCs is inactive and is not activated on contact with T cells. More importantly, activation of Mac-1 on DCs by Mg2+ treatment significantly reduces their T cell-activating capacity, and active CD11b represses DC cross-priming of cytotoxic T cells. Thus, activation of Mac-1 on DCs represents a potential new immunosuppressive strategy for reducing pathological T cell responses in AA. Dr. Gupta has identified novel Mac-1 agonists, termed Leukadherins (LAs1-3) that bind to and activate Mac-1. Multiple lines of experimental evidence generated by Dr. Gupta’s and Dr. Jurecic’s groups have shown that LAs have potent anti-inflammatory and immunosuppressive properties. For example, treatment with LA1 is safely and effectively reducing the onset and severity of Experimental Autoimmune Encephalomyelitis (EAE) in mice, induced by Myelin Oligodendrocyte Glycoprotein (MOG). Moreover, in EAE mice LA1 efficiently decreased the activation of myelin-reactive T cells and their IFN-γ production. We hypothesized that by activating Mac-1 on DCs LAs could effectively (a) reduce T cell-activating capacity of DCs and attenuate allo-reactive T cell responses, and (b) reduce severity of AA in mouse models. Indeed, in mixed lymphocyte reaction, which depends on stimulation of allogeneic T cells by DCs, LA1 significantly suppressed proliferation of lymph node T cells from C57BL6/J mice in the presence of irradiated splenocytes from allogeneic DBA/2J mice. SAA was induced in B6D2F1 mice by tail vein injection of 5 x 10e7 LNCs from C57BL/6J mice. The untreated AA mice died within 21 days of LNC infusion and exhibited (a) severe BM aplasia, (b) ~5-fold expansion of CD4+ T cells and >20-fold expansion of CD8+ T cells in comparison to Control B6D2F1 mice, and (c) severe depletion of HSCs (LSK CD150+ CD48- BM cells); Multipotent progenitors (MPPs, LSK CD150- CD48- BM cells); and Hematopoietic progenitors (HPC-1, LSK CD150- CD48+ cells; HPC-2, LSK CD150+ CD48+ cells). In contrast, AA mice treated IP with 1 mg/kg/day of LA1 for 7 or 21 days after LNC infusion exhibited (a) mild BM aplasia and improved BM cellularity, (b) significantly reduced expansion of CD4 (~2-fold) and CD8 (~12-fold) T cells in the BM, and (c) significantly improved frequency and total numbers of HSCs and progenitors in comparison to untreated AA mice. More importantly, treatment of AA mice with LA1 for 21 days resulted in 40-50% of AA mice surviving for more than 7 weeks after LNC infusion. These results demonstrate that treatment with LA1 can safely convert SAA into a moderate disease in preclinical mouse AA models and provide a platform for testing of LAs as new alternative or adjuvant therapy to manage ongoing AA in patients who (1) are not responding to IST and are not candidates for BMT, and/or (2) are undergoing IST and awaiting BMT. Disclosures No relevant conflicts of interest to declare.

2019 ◽  
Vol 20 (4) ◽  
pp. 514-514
Author(s):  
Giorgio Napolitani ◽  
Prathiba Kurupati ◽  
Karen Wei Weng Teng ◽  
Malick M. Gibani ◽  
Margarida Rei ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2886-2886
Author(s):  
Jochen Greiner ◽  
Li Li ◽  
Krzysztof Giannopoulos ◽  
Christian Brunner ◽  
Konstanze Döhner ◽  
...  

Abstract Specific immunotherapies for CML patients targeting T cell antigens might eliminate residual CML cells after therapy with imatinib or chemotherapy and might enhance a specific graft versus leukemia effect after allogeneic stem cell transplantation without aggravating the graft versus host disease. However, for effective specific immunotherapies in CML, extended studies on the expression, the function and the immunology of leukemia-associated antigens (LAAs) and on the LAA presentation by leukemia cells are required. Here, we investigated on expression and functional aspects of tumor/leukemia-associated antigens (TAAs/LAAs) in CML. Several LAAs are expressed and are therefore candidate structures for specific immunotherapies: bcr-abl (100%), G250 (24%), hTERT (53%), MPP11 (91%), NEWREN60 (94%), PRAME (62%), Proteinase3 (71%), RHAMM/CD168 (83%) and WT1 (53%), but not BAGE, MAGE-A1, SSX2 or NY-ESO-1. The expression of LAAs varied according to leukocyte subsets and the expression of RHAMM/CD168, Proteinase3 and PRAME was upregulated in accelerated phase and blast crisis. Quantitative expression of several TAAs/LAAs correlated with the clinical course. In flow cytometry, CD34+ CML progenitor cells typed positive for HLA-molecules, but were deficient for CD40, CD80, CD83 and CD86. This lack of costimulatory molecules might constitute a tumor escape mechanism. However, RHAMM/CD168-R3-specific T cell responses were demonstrated by ELISPOT analysis and specific lysis of R3-peptide pulsed T2 cells in chromium-51 release assays. These CD8+ cells could be phenotyped as CCR7-CD27-CD45RA+ early effector T cells by tetramer staining. In conclusion, several LAAs are expressed in CML and are therefore candidate structures for specific immunotherapies. CML progenitor cells express HLA-molecules, but lack costimulatory molecules. However, as CML patients show RHAMM/CD168-R3-specific early effector T cells, peptide vaccination might be therefore a promising approach to enhance specific immune responses in CML.


2018 ◽  
Vol 19 (7) ◽  
pp. 742-754 ◽  
Author(s):  
Giorgio Napolitani ◽  
Prathiba Kurupati ◽  
Karen Wei Weng Teng ◽  
Malick M. Gibani ◽  
Margarida Rei ◽  
...  

2013 ◽  
Vol 81 (4) ◽  
pp. 1064-1077 ◽  
Author(s):  
Adriana Pina ◽  
Eliseu Frank de Araujo ◽  
Maíra Felonato ◽  
Flávio V. Loures ◽  
Claudia Feriotti ◽  
...  

ABSTRACTThe protective adaptive immune response in paracoccidioidomycosis, a mycosis endemic among humans, is mediated by T cell immunity, whereas impaired T cell responses are associated with severe, progressive disease. The early host response toParacoccidioides brasiliensisinfection is not known since the disease is diagnosed at later phases of infection. Our laboratory established a murine model of infection where susceptible mice reproduce the severe disease, while resistant mice develop a mild infection. This work aimed to characterize the influence of dendritic cells in the innate and adaptive immunity of susceptible and resistant mice. We verified thatP. brasiliensisinfection induced in bone marrow-derived dendritic cells (DCs) of susceptible mice a prevalent proinflammatory myeloid phenotype that secreted high levels of interleukin-12 (IL-12), tumor necrosis factor alpha, and IL-β, whereas in resistant mice, a mixed population of myeloid and plasmacytoid DCs secreting proinflammatory cytokines and expressing elevated levels of secreted and membrane-bound transforming growth factor β was observed. In proliferation assays, the proinflammatory DCs from B10.A mice induced anergy of naïve T cells, whereas the mixed DC subsets from resistant mice induced the concomitant proliferation of effector and regulatory T cells (Tregs). Equivalent results were observed during pulmonary infection. The susceptible mice displayed preferential expansion of proinflammatory myeloid DCs, resulting in impaired proliferation of effector T cells. Conversely, the resistant mice developed myeloid and plasmacytoid DCs that efficiently expanded gamma interferon-, IL-4-, and IL-17-positive effector T cells associated with increased development of Tregs. Our work highlights the deleterious effect of excessive innate proinflammatory reactions and provides new evidence for the importance of immunomodulation during pulmonary paracoccidioidomycosis.


2017 ◽  
Vol 41 (4) ◽  
pp. 1271-1284 ◽  
Author(s):  
Astrid M. Westendorf ◽  
Kathrin Skibbe ◽  
Alexandra Adamczyk ◽  
Jan Buer ◽  
Robert Geffers ◽  
...  

Background/Aims: Hypoxia occurs in many pathological conditions, including inflammation and cancer. Within this context, hypoxia was shown to inhibit but also to promote T cell responses. Due to this controversial function, we aimed to explore whether an insufficient anti-tumour response during colitis-associated colon cancer could be ascribed to a hypoxic microenvironment. Methods: Colitis-associated colon cancer was induced in wildtype mice, and hypoxia as well as T cell immunity were analysed in the colonic tumour tissues. In addition, CD4+ effector T cells and regulatory T cells were cultured under normoxic and hypoxic conditions and examined regarding their phenotype and function. Results: We observed severe hypoxia in the colon of mice suffering from colitis-associated colon cancer that was accompanied by a reduced differentiation of CD4+ effector T cells and an enhanced number and suppressive activity of regulatory T cells. Complementary ex vivo and in vitro studies revealed that T cell stimulation under hypoxic conditions inhibited the differentiation, proliferation and IFN-γ production of TH1 cells and enhanced the suppressive capacity of regulatory T cells. Moreover, we identified an active role for HIF-1α in the modulation of CD4+ T cell functions under hypoxic conditions. Conclusion: Our data indicate that oxygen availability can function as a local modulator of CD4+ T cell responses and thus influences tumour immune surveillance in inflammation-associated colon cancer.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A159-A159
Author(s):  
Alma-Martina Cepika ◽  
Pauline P Chen ◽  
Molly Uyeda ◽  
Brandon Cieniewicz ◽  
Mansi Narula ◽  
...  

BackgroundGraft-vs-host-disease (GvHD) is a life-threatening complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT), limiting the use of this potentially curative treatment for hematological malignancies. To address this, we have developed T-allo10 cell therapy, which is enriched with type 1 regulatory T (Tr1) cells. Tr1 cells are peripherally inducible, CD49b+LAG3+IL-10+FOXP3- regulatory T cells that can confer alloantigen-specific tolerance, making them an attractive alternative to existing GvHD therapies, which non-discriminately impair both GvHD and protective immunity. T-allo10 cells are currently being evaluated in a phase I clinical trial in patients with hematological malignancies undergoing allo-HSCT (NCT03198234). Herein, we aimed to confirm that Tr1 cells are the active ingredient responsible for the T-allo10 suppressive function, and reveal the underlying molecular signatures to elucidate the mechanisms of Tr1 cell-mediated suppression.MethodsT-allo10 cells were generated in a co-culture of healthy host or patient tolerogenic dendritic cells (DC-10) with allogeneic healthy donor CD4+ T cells, then tested for Tr1 phenotype, anergy, suppression and cytokine production. Sorted T-allo10-derived Tr1 cells and non-Tr1 cells, as well as control effector T cells (Teff) and parental CD4+ T cells, were analyzed by TCR- and RNA-seq. Protein expression for key differentially expressed genes were validated, and the functional roles for IL-10, CTLA-4 and PD-1 in T-allo10-mediated suppression were confirmed in a suppression assay.ResultsWe show that the T-allo10 cell product is: i) enriched for Tr1 cells, ii) anergic in response to alloantigen re-challenge, but not to non-specific stimuli or 3rd party antigens, and iii) suppresses host-reactive T cells, but not T cell responses to other antigens. Furthermore, T-allo10-derived, isolated Tr1 cells had a restricted TCR repertoire, suggesting they clonally expand in response to alloantigens. T-allo10-derived Tr1 cells have a distinct signature compared to non-Tr1 cells, and, in addition to IL-10, express high levels of CTLA-4 and PD-1 (but not FOXP3). Notably, blockade of CTLA-4 and the PD-1 pathway completely abolishes T-allo10-mediated suppression of T cell responses.ConclusionsOur data shows that Tr1 cells are the active, suppressive, and antigen-specific ingredient of T-allo10 cells. Furthermore, while the role of IL-10 in Tr1 cell-mediated suppression is well known, we uncover that Tr1 suppress in addition through CTLA-4 and PD-1. Collectively, these intriguing findings underscore the importance of CTLA-4 and PD-1 pathways in conferring cell-mediated immunological tolerance. Further, they define the key characteristics and modes of action of antigen-specific Tr1 cells, providing crucial information for the ongoing T-all10 trial and future design of novel Tr1 cell-based therapies.Ethics ApprovalThe patient study was approved by Administrative Panels on Human Subjects in Medical Research, Stanford University, Tallo10 eProtocol # 38734. Healthy donor samples were purchased as deidentified human blood products from the Stanford Blood Center, and are thus exempt.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2246-2246
Author(s):  
Craig A. Mullen ◽  
Ulker Kocak ◽  
Joanne L. Shaw ◽  
Shahram Mori

Abstract After transplant the immune system is reconstituted by cells derived from both hematopoietic stem cells and peripheral expansion of differentiated donor T cells. Immune function is poor despite transplantation of mature lymphocytes from immune competent donors. We tested the hypothesis that early antigen encounter at the time of cell transplant would enhance desired donor T cell responses in the post-transplant repertoire. 2 independent models of peptide-specific T cell responses were studied. Model 1 : The model for CD4 cells employed T cells from transgenic DO11.11 mice that constitutively express the T cell receptor for the class II restricted ovalbumin (OVA) peptide 323–339. Fig 1: Early exposure to OVA antigen enhances clonal expansion of OVA specific transgenic T-cells following syngeneic BMT. Lethally irradiated BALB/c mice were injected with 300 μg of OVA peptide in CFA or CFA alone subcutaneously one day before transplantation (D-1). The transplanted mice received 2x106 transgenic OVA specific T-cells and 6x106 non-transgenic naive BALB/c bone marrow cells. At 2 days (A) and 7weeks (B) following BMT, draining lymph nodes were isolated and examined for the presence of OVA-specific T-cells using FITC-labeled KJ-126 antibody and PE-labeled anti mouse CD4 antibody. Naïve BALB/c animals were used as negative controls (C). The absolute number of antigen-specific T-cells was determined by multiplying the total cells recovered with the percentage of OVA-specific CD4+ T-cells identified by flow. Figure Figure Model 2: The model for CD8 cells employed nontransgenic H2-Db-restricted T cell responses to the influenza nucleoprotein peptide 366–374. Fig 2: Antigen specific CD8+ cells in antigen-exposed animals are functionally active. Donor SW mice were immunized three times by ip injection of virus-infected spleen cells. Recipient C57BL/6 animals underwent BMT using influenza-immune donors spleen cells and bone marrow (10x106 and 4x106 respectively). Some transplant recipients were exposed to influenza virus on D-1. Ten days following BMT, the animals were sacrificed and spleens were isolated and stimulated in vitro with 2 μg of NP peptide. After two rounds of stimulation, the splenocytes were assayed by intracellular cytokine assay for the secretion of IFNg by staining with PE-anti IFNγ and FITC-anti-CD8 antibodies. The results are representative of three experiments (total number n=4/experimental group). Figure Figure Encounter with specific antigen at the time of T cell transplantation led to clonal expansion of donor T cells and preservation of donor T cell function in the post-transplant immune environment. Antigen-specific donor T cell function was poor if antigen encounter was delayed or omitted. Severe parent>F1 graft versus host reactions blocked the effect of early antigen exposure.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 2537-2537
Author(s):  
T. B. Hunter ◽  
R. P. Gladue ◽  
S. J. Antonia

2537 Background: CD40-mediated interactions play an important role in the response to a variety of diseases, including cancer. Engagement of CD40 on antigen-presenting cells (APC) by CD40L leads to maturation and upregulation of co-stimulatory molecules, B7.1 and B7.2 (CD80 and CD86), which are requisite in the activation of T cells. Clinical trials involving immunologic interventions have shown clinical responses confirming that the immune system can be harnessed for the treatment of cancer. However, the clinical response rate has been low, signifying the need for new immunotherapeutic strategies. To this end, an agonist antibody specific for CD40 has been developed and is being evaluated as a potential anti-cancer agent. Methods: The activation capacity of anti-CD40 antibody CP-870,893 was analyzed by performing flow cytometric analysis of APC maturation markers following incubation of monocyte derived dendritic cells (DC) with the antibody. IL-12 and macrophage inflammatory protein-1α (Mip1 α) secretion were also analyzed. The effect of the antibody on anti-tumor T cell responses was tested in an autologous human model consisting of tumor cells as stimulator cells and tumor-draining lymph node cells as responders from a series of cancer patients. Results: Cultured DC treated with CP-870,893 consistently display a mature phenotype: robust upregulation of CD80, CD83, CD86 and HLA-DR expression, increased Mip1 α secretion, and the loss of antigen presenting capability. IL-12 secretion was not detected. CP-870,893 also promotes the responsiveness of lymph node derived T cells to autologous tumor, indicated by IFNγ and IL-2 ELISpot. Conclusions: These data demonstrate that CP-870,893 binds to and activates DC. A fully autologous mixed lymph node cell/tumor cell model was used to demonstrate that this activation promotes tumor-specific T cell responses. T cells from the tumor draining lymph node are not responsive to autologous tumor cells, however in the presence of CP-870,893 this unresponsiveness is reversed. These data indicate that CP-870,893 warrants further study as an immunotherapeutic agent in the treatment of cancer. No significant financial relationships to disclose.


2021 ◽  
Vol 12 ◽  
Author(s):  
Chenchen Zhao ◽  
Yi Zhang ◽  
Hong Zheng

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for hematological malignancies. This beneficial effect is derived mainly from graft-versus-leukemia (GVL) effects mediated by alloreactive T cells. However, these alloreactive T cells can also induce graft-versus-host disease (GVHD), a life-threatening complication after allo-HSCT. Significant progress has been made in the dissociation of GVL effects from GVHD by modulating alloreactive T cell immunity. However, many factors may influence alloreactive T cell responses in the host undergoing allo-HSCT, including the interaction of alloreactive T cells with both donor and recipient hematopoietic cells and host non-hematopoietic tissues, cytokines, chemokines and inflammatory mediators. Interferons (IFNs), including type I IFNs and IFN-γ, primarily produced by monocytes, dendritic cells and T cells, play essential roles in regulating alloreactive T cell differentiation and function. Many studies have shown pleiotropic effects of IFNs on allogeneic T cell responses during GVH reaction. Epigenetic mechanisms, such as DNA methylation and histone modifications, are important to regulate IFNs’ production and function during GVHD. In this review, we discuss recent findings from preclinical models and clinical studies that characterize T cell responses regulated by IFNs and epigenetic mechanisms, and further discuss pharmacological approaches that modulate epigenetic effects in the setting of allo-HSCT.


Sign in / Sign up

Export Citation Format

Share Document