The Role of CMV Serostatus of the Donor in the Graft Lymphoid Composition and Prediction of Gvhd and CMV Reactivation

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2422-2422
Author(s):  
Anna Kreutzman ◽  
Itxaso Portero Sainz ◽  
Valle Gomez Garcia De Soria ◽  
Carlos Fernandez ◽  
Royg Mercedes ◽  
...  

Abstract Background. Cytomegalovirus (CMV) is a widespread persistent β–herpesvirus, which can cause severe complications during primary infection or reactivation in immunocompromised patients, such as after allogeneic stem cell transplantation (alloSCT). Another major complication associated with alloSCT is graft-versus-host disease (GVHD). The pathogenesis of GVHD involves migration of the transplanted donor naïve T-cells into the secondary lymphoid organs in the recipient, which is mainly steered by CD62L and CCR7. As these homing molecules have been associated with both acute GVHD (aGVHD) and chronic GVHD (cGVHD), we studied whether the CMV serostatus of the donor affects the lymphoid composition of the graft product and whether this phenotype can predict CMV reactivation and GVHD. Methods. This single-center study included 77 donor-recipient pairs who underwent alloSCT. 64 pairs were HLA identical, 12 had 1 mismatch and 3 had 2 mismatch. 36 donors were related to their recipients. All recipients were followed at least for 100 (aGVHD) or 360 days (cGVHD) after transplantation. 43 donors were CMV-seropositive (CMVpos) and 34 were CMV-seronegative (CMVneg). 62 recipients were CMVpos, and 32 of them developed CMV reactivation, 25 aGVHD and 30 cGVHD. Samples from the graft product (donor) were phenotyped by flow cytometry (CD45, CD3, CD8, CD4, CD62L, CCR7) and both frequency (freq) and absolute number (abs) of each T-cell subpopulation were analyzed. Results. When the donors were divided based on their CMV serostatus, we observed that the grafts from CMVpos donors had a lower freq of naïve (CCR7+CD62L+) CD4+ T-cells (of lymphocytes p=0.06, of CD3 p=0.06, of CD4 p=0.07) and naïve CD8+ T-cells (of leukocytes p=0.03, of lymphocytes p=0.041, of CD3 p=0.011, of CD8 p=0.012) compared to CMVneg donors. Further, the abs of transplanted naïve CD8+ T-cells was significantly lower in the grafts from CMVpos donors (p=0.048). No differences were observed in T-cells (CD3+, CD4+, CD8+). We next studied if the CMV-serostatus and T-cell phenotype of the graft associates with GVHD. CMVpos donors whose recipients developed aGVHD had higher abs (p=0.05) and freq of naïve CD8+ T-cells (of lymphocytes p=0.08, of CD3 p=0.08, of CD8 p=0.11) compared to those without aGVHD. The same trend was observed with abs (p=0.11) and freq of CCR7+CD4+ T-cells (of leukocytes p=0.15). Similarly, those CMVpos donors whose recipients developed cGVHD had higher abs (p=0.05) and freq of CCR7+CD8+ T-cells (of leukocytes p=0.03, of lymphocytes p=0.06). Further, cGVHD patients who received the transplant from CMVpos donors were infused with a higher freq of CD3+ (of leukocytes p=0.03) and CD4+ T cells (of leukocytes p=0.04) than patients who received a graft from CMVpos donors but did not develop cGVHD. In contrast, CMVneg donors who developed aGVHD had a higher freq of CD3+ (p=0.018) and CD4+ T-cells (p=0.09), whereas no differences were seen in the T-cell subpopulations. Conversely, the abs (p=0.08) and freq of CCR7+CD4+ T-cells (of leukocytes p=0.11) were higher in those who later developed cGVHD. To study whether the graft lymphoid composition can be used to predict CMV reactivation, we analyzed the lymphoid composition in the graft product of those donors (both CMVpos and CMVneg) whose recipients were CMV seropositive but did not develop any form of GVHD (to avoid the influence of GVHD in the reactivation of CMV). Despite the low number of patients (CMV reactivation n=9, and no CMV reactivation n=13), we observed trends of higher portion of CD4+ T-cells (p=0.09 of lymphocytes, of CD3 p=0.20) and CCR7+CD4+ T-cells (of lymphocytes p=0.18, of CD4 p=0.16) in those grafts that were transplanted into CMV seropositive recipients who did not reactivate CMV. Conclusions. CMVpos donors whose recipient developed either aGVHD or cGVHD had a higher abs and freq of naïve CD8+ T-cells, which was not seen with CMVneg donors. This suggests that seropositivity sets the abs and freq of CD8 subpopulations near to a decisive cutoff for the development of GVHD. Conversely, other factors influences the development of GVHD in those patients whose donors were seronegative. In other words, seropositivity of the donor affects the graft composition and thus the risk of GVHD. Finally, our data indicate that a higher proportion of naïve or central memory CCR7+ CD4+ T cells in the donor graft could prevent CMV reactivation suggesting that graft composition affects also CMV reactivation. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 581-581
Author(s):  
Yi Zhang ◽  
Elizabeth Hexner ◽  
Dale Frank ◽  
Joe Gerard ◽  
Frank Kung ◽  
...  

Abstract Although mature CD8+ T cells are known to be major effectors of acute GVHD, patients receiving T cell-depleted allografts remain at high risk for chronic GVHD. To what extent CD8+, CD4+ or both T cell subsets contribute to this chronic immunopathology is not known. We have recently demonstrated that alloreactive memory T cells develop in mice with acute GVHD and account for the persistence of host tissue injury (Journal of Immunology, 2005;174:3051). Based on these findings, we now ask whether de novo generated donor T cells from engrafted T-BM themselves contribute to persistent host tissue injury in GVHD. Confirming previous observations, we found that transplantation of lethally irradiated C57BL/6SJL (B6, CD45.1) mice with highly purified C3H.SW (CD45.2) CD4+ naïve T cells did not cause GVHD, but mice receiving highly purified CD8+ naïve T cells together with C3H.SW T-BM, suffered severe acute GVHD. Surprisingly, in these mice receiving only CD8+ T cells, a substantial number of donor CD4+ T cells as well as CD8+ T cells were detected in GVHD target tissues, indicating that these infiltrating CD4+ T cells had arisen de novo from the transplanted T-BM. Donor CD4+ T cells recovered from GVHD mice expressed surface markers of activated effector/effector memory T cells, including CD25, CD69, CXCR3, and CD44hiCD62Llo. In response to host DCs, purified GVHD CD4+ T cells proliferated and expanded 4-5X more, and produced 10X higher levels of IFN-γ than did CD4+ T cells derived from B6 mice receiving C3H.SW T-BM alone. Furthermore, adoptive transfer of these in vivo generated GVHD CD4+ T cells, without CD8+ T cells, into secondary irradiated B6 recipients induced clinical GVHD characterized by delayed onset, weight loss, diarrhea, and lymphopenia, but without cutaneous inflammation. Histologic examination demonstrated chronic inflammation in the liver and intestinal tract, including epithelial apoptosis. Thymic pathology was dramatic in secondary B6 recipients of GVHD CD4+ T cells, including thymic atrophy, loss of thymic cortex, and infiltration of large amount of tingible macrophages. Taken together, these results demonstrate that donor bone marrow derived, de novo generated CD4+ T cells also contribute to GVHD together with transferred mature CD8+ T cells. Moreover, they suggest that these CD4+ T cells, in concert with alloreactive memory CD8+ T cells that develop during the evolution of GVHD, cause the persistence of acute GVHD and its subsequent progression into chronic GVHD. Thus, donor BM-derived, de novo generated CD4+ T cells are the “Hidden Dragon” of CD8+ T cell-mediated GVHD. Understanding how these CD4+ T cells are generated and regulated will prove to be critical to the prevention and treatment of both acute and chronic GVHD.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1158-1158
Author(s):  
Miguel Perales ◽  
Ying Taur ◽  
Ingrid Leiner ◽  
Marissa N Lubin ◽  
Boze Susac ◽  
...  

Abstract Introduction: Double-unit cord blood transplantation (DCBT) is a viable therapy for adults with high-risk hematologic malignancies who lack an adult donor. However, lack of transfer of memory T cells in the graft is associated with an increased risk of viral infections. To study immune reconstitution, we recently described a novel method that combines 5' rapid amplification of complementary DNA ends (RACE) PCR and deep sequencing to quantify T cell receptor (TCR) diversity after allogeneic hematopoietic stem cell transplant (van Heijst, Nat Med 2013). In that study, we showed that recipients of DCBT recover TCR diversity comparable to healthy donors by 12 months. We now report results of a prospective analysis of CD4+ and CD8+ T cell repertoire recovery in DCBT recipients and correlation with clinical outcomes. Methods: We prospectively collected samples from 33 DCBT recipients. The median age was 45 years (range 26-71), 18 (55%) were CMV seropositive, and the majority (n = 17, 52%) had non-European ancestry. Diagnoses included 20 (61%) acute leukemias and 13 (39%) lymphomas. Conditioning was myeloablative (n=1, 3%), reduced intensity (n=28, 85%), or non-myeloablative (n=4, 12%), and all patients received GVHD prophylaxis with cyclosporine-A and mycophenolate mofetil and no ATG. Patients received double-unit CB grafts (4-6/6 HLA-A,-B antigen, -DRB1 allele donor-recipient matched); this was supplemented with haploidentical CD34-selected PBSC in 18 patients. The 66 units had a median donor-recipient HLA-allele match of 6/8 (range 3-8). Infused total nucleated cell doses were 2.3 (1.7-3.3) and 1.9 (1.3-2.5) for the larger and smaller units, respectively. Samples were collected from the DCB grafts, recipient day+21 bone marrow, and peripheral blood at days +30, 60, 90, 120, 180 and 365 post-transplant. TCR-β sequences from each sample were amplified and sequenced using the Illumina/MiSeq sequencing platform after isolation of CD4+ and CD8+ T cells. TCR abundances were assessed at the level of clonotype and TCR diversity was calculated using inverse Simpson index. Results: Of the 33 patients, long-term samples were obtained in 25 patients, short-term samples (≤ day 100) in 6 patients who died early after DCBT, and no samples other than the graft for 2 patients. The remainder of the results focuses on 25 patients with complete samples. As previously shown, there is a 1-2-log increased diversity in CD4+ vs. CD8+ T cells (Figure). Furthermore, median CD4+ steady-state diversity is achieved early by day 60. In contrast, there is a higher rate of clonal dominance in CD8+ compared to CD4+ T cells (24/25 vs. 11/25, p=0.0001 by Fisher test). Several patterns of clonal dominance were observed, including two main patterns in CD8+ T cells. In 7/24 patients, clonal dominance is established by day 60 and persists throughout, whereas in 12/24 patients, clonal dominance fluctuates throughout follow-up. In CD4 +T cells, where less dominance is observed, a similar distribution is seen, though prolonged clonal dominance is rare. Interestingly, some of the dominant clones can be detected in the graft and are present in the day 21 marrow sample. Persistent clonal dominance in CD8+ T cells was seen 6/9 patients with CMV reactivation, whereas ongoing fluctuation was seen in 9/12 patients without CMV reactivation. In 2 patients with fluctuating clones who reactivated CMV, 1 had low level and the other a late viremia. In contrast, no link to a specific pattern was observed in patients with HHV6 viremia or acute GVHD. Finally, when we assessed similarity in clonal distribution between time points, there was more similarity in CD8+ than CD4+ T cells. Conclusions: This novel deep TCR repertoire sequencing provides a quantitative picture of T cell recovery after DCBT and supports the following: 1) separate analysis of CD4+ and CD8+ T cell populations is critical given different patterns of recovery in T cell subsets; 2) there is significant turnover in CD4+ clones but with overall limited dominance, whereas there is less turnover in CD8+ clones; 3) although the grafts contain predominantly naïve T cells, the clonal evolution of CD8+ T cells strongly suggests generation of virus-specific T cells that control viral infection; and 4) CMV appears to be an important driver of CD8+ T cell clonal expansion after CBT. Ongoing analyses are correlating immune recovery with cord blood unit dominance as well as the biology of GVHD and relapse. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1882-1882
Author(s):  
Xiong Ni ◽  
Qingxiao Song ◽  
Ruishu Deng ◽  
Hua Jin ◽  
Kaniel M. Cassady ◽  
...  

Abstract Recipient tissue expression of programmed death-ligand 1 (PD-L1, B7H1) down-regulates graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (HCT), but this information has not been translated clinically. Here we demonstrate a critical role for recipient PD-L1 expression in preventing both acute and chronic GVHD while preserving graft-versus-leukemia (GVL) effects when donor CD4+ T cells are temporarily depleted in vivo early after HCT. Depletion of donor CD4+ T cells increases serum IFN-γ concentrations and enhances recipient tissue expression of PD-L1 and donor CD8+ T cell expression of the PD-L1 receptors CD80 and PD-1. In GVHD target tissues, depletion of CD4+ T cells increases anergy and apoptosis of infiltrating CD8+ T cells in a manner that depends on recipient PD-L1 expression, thereby preventing damage to intestinal Paneth cells and stem cells, hepatocytes, and thymic medullary epithelial cells, and preventing both acute and chronic GVHD. In lymphoid tissues, depletion of CD4+ T cells augments CD8+ T cell proliferation without increasing CD8+ T cell anergy or apoptosis, resulting in expansion of donor CD8+ T cells and strong GVL effects. Therefore, temporary depletion of donor CD4+ T cells early after HCT represents a novel approach for preventing GVHD while preserving GVL effects. This work was supported by NIH R01 AI066008 and Nesvig Lymphoma Fundation (to D.Z.) and by NSFC 81090413, 81270638 (to J.W.). Disclosures Forman: Amgen: Consultancy; Mustang: Research Funding. Martin:Neovii: Research Funding; RegImmune: Research Funding; Enlivex: Consultancy; Janssen: Consultancy; Pfizer: Consultancy; Pharmacyclics: Consultancy.


2010 ◽  
Vol 2010 ◽  
pp. 1-8 ◽  
Author(s):  
German Bernal-Fernandez ◽  
Patricia Espinosa-Cueto ◽  
Rosario Leyva-Meza ◽  
Nathalie Mancilla ◽  
Raul Mancilla

Patients with tuberculosis frequently develop anergy, a state of T-cell hyporesponsiveness in which defective T-cell costimulation could be a factor. To know if the expression of T-cell costimulatory molecules was altered in tuberculosis, we analyzed the peripheral blood T-cell phenotype of 23 Mexican patients with pulmonary tuberculosis. There was severe CD4 (P<.001) and CD8 (P<.01) lymphopenia and upregulation of costimulatory molecule CD30 on CD4 and CD8 T cells (P<.05); this increase was higher in relapsing tuberculosis. The main finding was severe downregulation of the major costimulatory molecule CD28 on both CD8 and CD4 T cells (P<.001). Depletion of the CD4/CD28 subset, a hitherto undescribed finding, is relevant because CD4 T cells constitute the main arm of the cell-mediated antimycobacterial immune response.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 645-645
Author(s):  
Toshiki Ochi ◽  
Hiroshi Fujiwara ◽  
Sachiko Okamoto ◽  
Hiroaki Asai ◽  
Yukihiro Miyazaki ◽  
...  

Abstract Abstract 645 [Purpose] Redirected T-cell-based anti-cancer adoptive therapy using cancer antigen-specific T-cell receptor (TCR) gene transfer has clinically shown promise, however there still remain considerable issues of achieving better clinical efficacy. For this purpose, helper function provided by concurrent CD4+ T cells should be evidently considerable. In this study, to achieve the enhanced anti-leukemia functionality mediated by redirected CD8+ T cells using WT1-specific TCR gene transfer, we in detail examined functionalities mediated by similarly redirected CD4+ T cells using the identical TCR gene transfer.[Methods] HLA-A*24:02-restricted and WT1235-243-specific codon-optimized TCR α/β genes were inserted into the novel retroviral vector encoding shRNAs for endogenous TCRs (WT1-siTCR vector). (1) Cognate antigen-responsive cytokine production mediated by WT1-siTCR transduced CD4+ T cells was assessed using cytokine beads array and ELISA assay. (2) Expression of CD40L and OX40 on redirected CD4+ T cells stimulated by WT1 peptide ligation was assessed using flow cytometer. (3) Impact caused by redirected CD4+ T cells on each magnitude of WT1-specific cytotoxicity, target-responsive proliferation and transition to central memory T-cell phenotype of WT1-siTCR transduced CD8+ T cells was measured using 51Cr-release assay, CD107a assay, intracellular IFN-γ assay and CFSE assay. (4) Chemokines produced by redirected CD4+ T cells stimulated using WT1 peptide was comprehensively assessed using real-time PCR. Consequent chemotaxis of redirected CD8+ T cells toward those stimulated redirected CD4+ cells was validated using transwell experiments. (6) Finally, anti-leukemia reactivity against autologous leukemia cells mediated by patients' redirected CD8+ T cells was similarly examined in the presence or absence of such autologous CD4+ T cells. [Results] First, in this study, those redirected CD4+ T cells hardly became positive for intracellular FoxP3, a crucial marker for regulatory T cell phenotype. In response to the WT1 peptide, WT1-siTCR transduced CD4+ T cells produced Th1 cytokines; IL-2, IFN-γ and TNF-α, in the context of HLA-A*24:02, which also needed HLA class II molecules on target cells. Magnitudes of WT1-responsive CD107a expression, IFN-γ production and cytotoxicity mediated by WT1-siTCR transduced CD8+ T cells were efficiently enhanced in the presence of redirected, but not non-redirected CD4+ T cells. Similarly, in the presence of those redirected CD4+ T cells, redirected CD8+ T cells expressing WT1-specific TCR increased in number and the transition to central memory T-cell phenotype (CD45RA−CD62L+) of those CD8+ T cells was stimulated in response to the stimulation with WT1 peptide. WT1 peptide ligation stimulated those redirected CD4+ T cells to express membrane-bound OX40, which is involved in the formation of central memory CD8+ T cells. WT1 peptide ligation also stimulated the redirected CD4+ T cells to produce chemokines; CCL3 and CCL4. Redirected CD8+ T cells expressed the receptor for these chemokines, CCR5; efficient migration of redirected CD8+ T cells toward redirected CD4+ T cells stimulated with WT1 peptide was obviously observed in transwell experiments. Finally, redirected CD4+ T cells isolated from patients with leukemia successfully provided Th1 helper function to autologous redirected CD8+ T cells to enhance the anti-leukemia reactivity; cytotoxicity, proliferation and formation of central memory T cells, in response to autologous leukemia cells, in vitro. [Conclusion] Although further investigations are warranted, concurrently adopted WT1-siTCR introduced CD4+ T cells seems feasible to enhance the efficacy of WT1-targeting redirected T-cell-based adoptive therapy for treatment of human leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2668-2668 ◽  
Author(s):  
Alice Bertaina ◽  
Perla Filippini ◽  
Valentina Bertaina ◽  
Barbarella Lucarelli ◽  
Aurelie Bauquet ◽  
...  

Abstract Background Blinatumomab is a bi-specific monoclonal antibody designed to engage and tether cytotoxic T-cells (CTL) to CD19-expressing target B cells. An ongoing phase I multicenter study in pediatric patients with relapsed/refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL) has shown that blinatumomab induces morphological and molecular remissions, defined as minimal residual disease (MRD) levels <10-4, in 47% of patients [Gore L, et al. J Clin Oncol 31, 2013 (suppl; abstr 10007)]. It is presently unknown whether and to what extent blinatumomab affects T-cell phenotype and function in pediatric patients with BCP-ALL. Patients and Methods Eight children diagnosed with relapsed/refractory BCP-ALL at the Bambino Gesù Children’s Hospital in Rome (median age at diagnosis 5.8 years, range 0.5-14.6) received blinatumomab as continuous intravenous infusion for 28 consecutive days, followed by a 2-week drug-free period. Four out of 8 patients were given repeated treatment courses. Peripheral blood samples were collected before treatment (day 0) and weekly thereafter, for 4 consecutive weeks. Bone marrow (BM) aspirates were available on days 0 and +29 of each drug course. Peripheral blood mononuclear cells (PBMC) were labeled with appropriate combinations of fluorochrome-conjugated monoclonal antibodies to quantitate naïve/memory T cells, αβ/γδ-expressing T cells and other immune effectors with potential anti-leukemia activity, such as CD3+CD56+ natural killer (NK) T cells and CD3-CD56+ NK cells. T-cell production of interferon (IFN)-γ, interleukin (IL)-4 and IL-17 was measured at the single-cell level, after short-term (4-hour) stimulation with phorbol myristate acetate (PMA) and ionomycin. The TCR-Vβ Repertoire Kit® (Beckman Coulter, Milan, Italy) allowed the flow cytometry analysis of 24 different Vβ specificities on T cells, thus covering approximately 70% of the normal human TCR-Vβ repertoire. Results Peripheral blood lymphocytes reached their nadir on day +1 (median 300/µL of blood [inter-quartile range 40-380] compared with 1,080/µL of blood at baseline [inter-quartile range 360-2,310]; p=0.0037 by Mann-Whitney U test for paired data), expanded within 7 days up to 3.5-fold above baseline, and included both CD4+ and CD8+ T cells. By contrast, the frequency of both CD3+CD56+ NK T cells and CD3-CD56+ NK cells remained unchanged compared to baseline. IFN-γ production by patient-derived CD4+ T cells exceeded that observed in CD4+ T cells from healthy controls by 2-fold, indicating robust T helper type 1 (Th1) polarization. The frequency of Th2/Th17 cells, defined as CD4+IL-4+ and CD4+IL-17+ cells, respectively, was not different after treatment compared to baseline. CD31 expression on recovering CD45RA+ naïve T cells, a surrogate phenotypic feature for recent thymic emigrants (RTEs), suggested that thymic output may contribute to T-cell expansion after blinatumomab administration. Non-significant changes in the relative proportion of TCR-αβ and TCR-γδ-expressing CD3+ T cells were detected after treatment (median 79.5% TCR-αβ+ T cells and 19.3% TCR-γδ+ T cells among total CD3+ cells) compared with baseline (median 87.4% TCR-αβ+ T cells and 12.2% TCR-γδ+ T cells among total CD3+ cells). Importantly, both CD3+CD8bright T cells and NK cells expressed lytic granule proteins, such as perforin and granzyme-B, at levels that increased during treatment. The analysis of Vβ TCR repertoire revealed a restricted usage of single Vβ domains by BM-resident CD8+ T cells, but not by CD4+ T cells. Specifically, the sum of Vβ within CD8+ T cells in the BM averaged 56.7±6.2% after blinatumomab, compared with 78±5.1% in healthy controls (p=0.04; Mann-Whitney U test for unpaired data). Conclusions Blinatumomab expands both CD31+CD45RA+ thymic-naïve and memory T cells with heightened IFN-γ production and is highly effective at clearing MRD in children with BCP-ALL. Skewing of the Vβ repertoire within BM-resident CD8+ T cells may be consistent with clonal expansions. Disclosures: Zugmaier: Amgen: Employment.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4530-4530
Author(s):  
Mohammad Raeiszadeh ◽  
Annette Pachnio ◽  
Charles Craddock ◽  
Paul Moss ◽  
Frederick Chen

Haematopoietic stem cell transplant (HSCT) patients commonly suffer from Cytomegalovirus(CMV) reactivation due to the eradication and delayed reconstitution of CMV-specific T cell immunity resulting from T cell depletion and conditioning chemotherapy. Our knowledge of T cell immunity and in particular of antigen specific CD8+ T cell responses has advanced rapidly following the introduction of HLA- class I tetramers which enables the direct enumeration and characterisation of CMV-specific CD8+ T cells. In order to study the broader CMV specific T cells reconstitution post HSCT, we used a novel HLA-class II tetramer to monitor the reconstitution of CD4+ T cells specific to a CMV-derived peptide restricted to HLA-DRB1*0701, in parallel with different HLA class I tetramers identifying CMV specific CD8+ T cells. We analysed longitudinally the immune reconstitution of a cohort of thirteen HLA-DRB1*0701 –matched HSCT patients treated for haematological malignancies and who were at high risk of CMV reactivation where both donors and recipients were CMV seropositive. Twelve received reduced intensity conditioning and T-cell depletion with in vivo Alemtuzumab, and one had non-T cell depleted myeloablative conditioning. Twelve out of 13 longitudinally studied HSCT patients experienced CMV reactivation which included multiple episodes of viremia in 8 out of 12 .The viremia resolved in less than a month of onset in 8 patients where rapid expansion of CMV specific CD4+ and CD8+ T cells was observed in response to onset of viral reactivation. In four patients, late reconstitution of CMV-specific CD4 and CD8 T cells was observed between three and six months post HSCT; these patients suffered from prolonged and multiple episodes of CMV reactivation. In reconstituting patients, there was a considerable increase in the number of CMV-specific CD4+ and CD8+ T cells, from undetectable levels before viral reactivation up to 50x10^3 cells/ml and 370x10^3 cells/ml, respectively. CMV-specific CD4+ and CD8+ T cells expanded in parallel and statistically significant correlation between these cells were observed((p=0.06)). The patient treated with myeloablative conditioning chemotherapy retained considerable numbers of (CMV-specific CD4 cells (28.7x10^3 cells/ml) at four weeks post HSCT and did not have CMV reactivation. Phenotypic analysis showed that CMV specific CD4+ T cells were predominantly effector memory cells (CCR7-CD45RA-) whilst CD8+ T cells were predominantly effector memory/CD45RA+ cells. The majority of CMV specific T cells expressed CD57 molecule and we documented a strong correlation between expansion of specific CD4+ T cells and generation of CD4+CD57+ cells post HSCT. Both CD4 and CD8 T cells specific to CMV appear to be required for the control of viremia. The use of HLA class I and class II tetramers in combination with antibodies against surface markers such as CD57 provides a broader picture of the global T cell immune response to CMV and may inform on clinical outcome and treatment guidance. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 18 (1) ◽  
Author(s):  
Stephanie M. Dillon ◽  
Tezha A. Thompson ◽  
Allison J. Christians ◽  
Martin D. McCarter ◽  
Cara C. Wilson

Abstract Background The etiology of the low-level chronic inflammatory state associated with aging is likely multifactorial, but a number of animal and human studies have implicated a functional decline of the gastrointestinal immune system as a potential driver. Gut tissue-resident memory T cells play critical roles in mediating protective immunity and in maintaining gut homeostasis, yet few studies have investigated the effect of aging on human gut T cell immunity. To determine if aging impacted CD4 T cell immunity in the human large intestine, we utilized multi-color flow cytometry to measure colonic lamina propria (LP) CD4 T cell frequencies and immune-modulatory marker expression in younger (mean ± SEM: 38 ± 1.5 yrs) and older (77 ± 1.6 yrs) adults. To determine cellular specificity, we evaluated colon LP CD8 T cell frequency and phenotype in the same donors. To probe tissue specificity, we evaluated the same panel of markers in peripheral blood (PB) CD4 T cells in a separate cohort of similarly aged persons. Results Frequencies of colonic CD4 T cells as a fraction of total LP mononuclear cells were higher in older persons whereas absolute numbers of colonic LP CD4 T cells per gram of tissue were similar in both age groups. LP CD4 T cells from older versus younger persons exhibited reduced CTLA-4, PD-1 and Ki67 expression. Levels of Bcl-2, CD57, CD25 and percentages of activated CD38+HLA-DR+ CD4 T cells were similar in both age groups. In memory PB CD4 T cells, older age was only associated with increased CD57 expression. Significant age effects for LP CD8 T cells were only observed for CTLA-4 expression, with lower levels of expression observed on cells from older adults. Conclusions Greater age was associated with reduced expression of the co-inhibitory receptors CTLA-4 and PD-1 on LP CD4 T cells. Colonic LP CD8 T cells from older persons also displayed reduced CTLA-4 expression. These age-associated profiles were not observed in older PB memory CD4 T cells. The decline in co-inhibitory receptor expression on colonic LP T cells may contribute to local and systemic inflammation via a reduced ability to limit ongoing T cell responses to enteric microbial challenge.


Immuno ◽  
2021 ◽  
Vol 1 (3) ◽  
pp. 119-131
Author(s):  
Jana Palmowski ◽  
Kristina Gebhardt ◽  
Thomas Reichel ◽  
Torsten Frech ◽  
Robert Ringseis ◽  
...  

CD4+ T cells are sensitive to peripheral changes of cytokine levels and metabolic substrates such as glucose and lactate. This study aimed to analyze whether factors released after exercise alter parameters of human T cell metabolism, specifically glycolysis and oxidative phosphorylation. We used primary human CD4+ T cells activated in the presence of autologous serum, which was collected before (CO) and after a 30-min exercise intervention (EX). In the course of activation, cells and supernatants were analyzed for cell viability and diameter, real-time oxygen consumption by using PreSens Technology, mRNA expression of glycolytic enzymes and complexes of the electron transport chain by real-time PCR, glucose, and lactate levels in supernatants, and in vitro differentiation by flow cytometry. EX did not alter T cell phenotype, viability, or on-blast formation. Similarly, no difference between CO and EX were found for CD4+ T cell activation and cellular oxygen consumption. In contrast, higher levels of glucose were found after 48 h activation in EX conditions. T cells activated in autologous exercise serum expressed lower HK1 mRNA and higher IFN-γ receptor 1. We suggest that the exercise protocol used was not sufficient to destabilize the immune metabolism of T cells. Therefore, more intense and prolonged exercise should be used in future studies.


Sign in / Sign up

Export Citation Format

Share Document