scholarly journals Very Early Dynamics of Regulatory T-Cell Chimerism Significantly Varies According to the Donor Sources: Implication for Basic Immune Pathogenesis of Engraftment Phase

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4575-4575
Author(s):  
Miki Iwamoto ◽  
Ken-ichi Matsuoka ◽  
Yusuke Meguri ◽  
Takeru Asano ◽  
Takanori Yoshioka ◽  
...  

Abstract Post-transplant expansion of donor-derived T cells has crucial impact on the early clinical events including graft engraftment and acute graft-versus-host disease. Flowcytometry-based method enables us to analyze the lymphocyte chemerism in the very early phase after HSCT and recent reports have shown that T-cell achieved donor-chimerism in the first two weeks in the majority cases. However, the very early dynamics of each T-cell subset, including CD4+Foxp3+ regulatory T cells (Tregs), has not been well characterized. Since the early expansion of Tregs and other CD4+ and CD8+ conventional T cells (Tcons) are immunologically competitive and might important for the stabilization of immunity in the early phase, we hereby investigated the early dynamics of donor-Treg chimerism comparing with Tcons within each individual patient. Laboratory studies were undertaken in 11 adult patients who received HLA-mismatched allogeneic graft; unrelated cord blood (n=5), unrelated peripheral blood (n=1) and related peripheral blood (n=5). Blood samples were obtained before and at 1, 2, 4, and 6 weeks after HSCT. Peripheral blood mononuclear cells (PBMCs) were isolated from blood samples by density gradient centrifugation and cryopreserved before being analyzed. After thawing, to analyze the subset-specific chimerism, PBMCs were stained with anti-HLA monoclonal antibodies and other subset-specific antibodies as follows: Pacific Blue conjugated anti-CD4, eFluor450 conjugated anti-CD3, PE-Cy7 conjugated anti-CD25, anti-CD14, APC conjugated anti-CD127, anti-CD56, and APC-eFluor780 conjugated anti-CD8a, anti-CD19. Gated lymphotes (CD4+Tcons, CD4+Tregs, CD8+T cells, B cells, NK cells, Monocytes) were analyzed their chimerism by flowcytometry. All 11 patients achieved donor-dominant chimerism of T cells, NK cells and Monocytes (>90%) by 4 weeks after HSCT. As for T-cell subsets, donor-chimerisms of Tregs at the first week were higher than that of CD4+ and CD8+ Tcons in all 5 patients after PBSCT (Average %donor chimerisms: Tregs 81.3%, CD4+Tcon 66.0%, CD8+Tcon 75.2%). Of interest, patients after cord blood transplantation (CBT) showed marked contrast to PBSCT where donor-chimerism of Tregs at the first week was much lower than that of CD4+ and CD8+ Tcons (Average %donor chimerism: Tregs 27.2%, CD4+Tcon 53.2%, CD8+Tcon 47.0%), and it is significantly lower than that of PBSCT (P=0.009). At 4 weeks when Treg achieved complete donor-chimerism in all patients, Treg percentage of total CD4 T cells after CBT was lower than that after PBSCT (average %Treg at w4: 7.8% vs 12.6%, respectively). Clinically, 3 patients with delayed donor-Treg achievement in the first week after CBT developed pre-engraftment immune reaction (PIR) which was followed by the onset of acute GVHD, although patients with donor-Treg dominant recovery in the first week after PBSCT did not develop clinical PIR. These data suggest that cord blood-derived Tregs expanded less aggressively in the very early phase and achieve donor-chimerism behind Tcons within each individual patient. Slower rising-up of cord blood-derived Treg in the first week appears to be associated with the low percentage of Treg at 4 weeks after CBT. In good contrast, PBSC-derived Tregs achieved donor-chimerism prior to Tcons. Taken together, our results suggest that early dynamics of donor-Treg chimerism after HLA-mismatched HSCT might significantly vary according to the donor sources and be critically linked to the clinical immune events in the early phase after HSCT. The careful monitoring of early Treg reconstitution from the point of view might provide a novel strategy to promote immune tolerance in the early phase after transplantation. Disclosures Maeda: Mundipharma KK: Research Funding.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2227-2227
Author(s):  
Satoshi Takahashi ◽  
Nobukazu Watanabe ◽  
Jun Ooi ◽  
Akira Tomonari ◽  
Kashiya Takasugi ◽  
...  

Abstract The immaturity of T cells in cord blood is well known in functional assays and phenotypic analyses. During the first several months after cord blood transplantation (CBT), the T cell compartment is recovered by peripheral expansion from those mature and naïve T cells in cord blood grafts and plays an important role in acute graft-versus-host disease (GVHD) and graft-versus-leukemia reaction. Recently, we have reported that adult patients with hematological malignancies receiving CBT from HLA-partially-mismatched unrelated donors (n=68) had a lower risk of severe acute GVHD (> grade II, 7% versus 26%) and transplant-related mortality (9% versus 29% at 1 year) and a higher probability of disease-free survival (74% versus 44% at 2 years) than HLA-matched unrelated bone marrow transplant (BMT) recipients (n=45) in our multivariate analysis (Takahashi et al., Blood, in press). We speculated that the immune reconstitution process over a period of several months after CBT might have contributed to these promising clinical results. Using four-color analysis with CD4, CD8, CD45RA, and CD62L, more than 90% of cord blood CD4+ and CD8+ T cells in the grafts belonged to the naïve fraction. Cytokine expression in cord blood T cells was also suppressed to 0.1% in CD4+ and to 0.9% in CD8+ with positive interferon-γ by intracellular staining, which were significantly lower than those in adult T cells (16.2% in CD4+ and 37.8% in CD8+). Circulating T cell counts normalized after 3 months for CD8+ and 4 months for CD4+ in our CBT recipients, both of which were significantly faster than in previously published studies, which were 9 months for CD8+ and 12 months for CD4+. After T cell recovery, peripheral blood T cells moved from the naïve to the central memory fraction immediately, and then moved to the effector memory fraction. A naïve subset of CD4+ T cells remained (median: 38 cells/μl on day 90, n=12) during the first 3 months, which was significantly higher than in the BMT control (median: 9 cells/μl on day 90, n=5, p=0.015), but showed a low level of CD8+ T cells (median: 14 cells/μl on day 90, n=12), almost the same as in BMT recipients (median: 13 cells/μl on day 90, n=5). Intracellular interferon-γ-producing T cells were detected at 3.4% (0.1–34.2%) in CD4+ and 32.3% (1.1–86.9%) in CD8+ at 1 month post-CBT (n=16), both of which were comparable to post-BMT. To investigate whether these T cells with memory phenotype are functional, we analyzed antigen-specific T-cell recovery using cytomegalovirus (CMV) as a specific antigen. CMV-responsive CD4+ T cells were detected within the first 4 months in all recipients with positive CMV antigenemia (n=13), but CD8+ T cells were detected only in 5 out of 13 cases, probably because of pre-emptive Gancyclovir administration in most antigenemia-positive patients. To conclude, naïve cord blood T cells rapidly increased in number and adopted a memory phenotype showing cytokine-production and antigen-recognition capacity in the early phase after CBT. These data suggest that mature T lymphocytes in cord blood have unique properties and contribute to the favorable clinical outcome of CBT.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 224-224 ◽  
Author(s):  
Patrick J Hanley ◽  
Caridad Martinez ◽  
Kathryn Leung ◽  
Barbara Savoldo ◽  
Gianpietro Dotti ◽  
...  

Abstract Abstract 224 Adenovirus(Ad), Cytomegalovirus(CMV) and Epstein-Barr virus(EBV) frequently cause severe morbidity and mortality in patients(pts) after stem cell transplantation (SCT) and cord blood transplantation(CBT). We have shown that adoptive immunotherapy with peripheral blood(PB) donor derived multivirus-specific Cytotoxic T Lymphocytes directed against Ad, CMV and EBV can effectively prevent and treat the clinical manifestations of these viruses after SCT. CBT, while less likely to cause GvHD than conventional SCT, is unlikely to provide passive transfer of virus-specific CTL, since CBTs come from virus-naïve donors. Here we report for the first time the transfer of CB-derived multivirus-specific CTL(cbmCTL) to CBT recipients to restore cellular immunity to Ad, CMV and EBV. The development of cbmCTLs for pts undergoing CBT requires the priming and extensive expansion of naïve T cells rather than the more limited and simple direct expansion of pre-existing memory T cell populations from virus-exposed donors. We hypothesize that cbmCTL, derived from naïve T cells, will be efficacious and persist in vivo. Our protocol uses an initial round of stimulation with autologous CB-derived dendritic cells transduced with a recombinant Ad5f35 vector containing a transgene for the immunodominant CMV antigen, pp65 (Ad5f35pp65) in the presence of IL-7, IL-12 (CTEP-NCI) and IL-15. This is followed by 2 rounds of weekly stimulation with autologous Ad5f35pp65-transduced EBV-LCL in the presence of IL-15 or IL-2. Seven cbmCTL cultures generated for clinical use contained a mean of 48% CD8+, and 36% CD4+ cells with a mean of 33% CD45RA-/CD62L+ central memory T cells. In 51Cr release and/or IFNg ELISPOT assays, cbmCTL lines showed specific activity against all viruses. We have treated 7 pts who received the 80% fraction of a fractionated CB unit followed by cbmCTLs generated from the remaining 20% fraction; two pts were treated on each dose level;5×106/m2; 1×107/m2; and 1.5×107/m2 while one pt has been treated with 2.5×107/m2 – dose level 4. Pts received cbmCTLs on days 63–146 after CBT (median: day 83). No early infusion-related toxicities or subsequent GvHD was observed. All pts engrafted neutrophils by day 30 (median: day 20) despite receiving only 80% of the CB unit. Five of 7 pts had no initial infection or reactivation episodes, remaining free of CMV, EBV, and Ad from 2 months to 2 years post-CBT. Of the two remaining pts, pt 1 was transiently viremic for CMV pre-infusion and became highly viremic 4-weeks post-cbmCTL. The pt received a 2nd dose of cbmCTLs and CMV DNA/antigen became undetectable in the PB within 16 days of the 2nd dose and remains asymptomatic and virus free >2 yr post-CBT. Analysis of this pt's PB showed a rise in CMV-T cells even prior to cbmCTL #2, with a 31-fold expansion of CMV-T cells by 4 weeks after the initial CTLs. This pt also had AdV in his stool, which resolved without additional therapy. Shortly after CTL infusion, pt 4 had detectable EBV DNA in the PB that was controlled without additional antiviral therapy. The transferred cells had long-term persistence, since T cell receptor(TCR) deep-sequencing (ImmunoSEQ) allowed us to track infused T cell clones (i.e. clones present in the infused cbmCTL but absent in peripheral blood before cbmCTL infusion) up to 1 year post-CBT in 5/5 pts tested. In summary, none of the recipients of cbmCTL developed viral disease; in two pts with viral infections, the infections resolved without progression to disease, coinciding with the appearance of virus-specific T cells in peripheral blood. Hence, administration of cbmCTL to pts after CBT has so far been safe and can facilitate reconstitution of virus-specific T cells and control viral reactivation/infection in vivo. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 1 (24) ◽  
pp. 2206-2216 ◽  
Author(s):  
Prashant Hiwarkar ◽  
Mike Hubank ◽  
Waseem Qasim ◽  
Robert Chiesa ◽  
Kimberly C. Gilmour ◽  
...  

Key Points Cord blood T cells are ontogenetically distinct from the peripheral blood T cells. Recapitulation of fetal ontogeny after cord blood transplantation results in rapid CD4+ T-cell reconstitution.


Blood ◽  
2009 ◽  
Vol 114 (9) ◽  
pp. 1958-1967 ◽  
Author(s):  
Patrick J. Hanley ◽  
Conrad Russell Young Cruz ◽  
Barbara Savoldo ◽  
Ann M. Leen ◽  
Maja Stanojevic ◽  
...  

The naive phenotype of cord blood (CB) T cells may reduce graft-versus-host disease after umbilical cord blood transplantation, but this naivety and their low absolute numbers also delays immune reconstitution, producing higher infection-related mortality that is predominantly related to CMV, adenovirus (Adv), and EBV. Adoptive immunotherapy with peripheral blood-derived virus-specific cytotoxic T lymphocytes (CTLs) can effectively prevent viral disease after conventional stem cell transplantation, and we now describe the generation of single cultures of CTLs from CB that are specific for multiple viruses. Using EBV-infected B cells transduced with a clinical-grade Ad5f35CMVpp65 adenoviral vector as sources of EBV, Adv, and CMV antigens, we expanded virus-specific T cells even from CB T cells with a naive phenotype. After expansion, each CTL culture contained both CD8+ and CD4+ T-cell subsets, predominantly of effector memory phenotype. Each CTL culture also had HLA-restricted virus-specific cytotoxic effector function against EBV, CMV, and Adv targets. The CB CTLs recognized multiple viral epitopes, including CD4-restricted Adv-hexon epitopes and immunosubdominant CD4- and CD8-restricted CMVpp65 epitopes. Notwithstanding their naive phenotype, it is therefore possible to generate trivirus-specific CTLs in a single culture of CB, which may be of value to prevent or treat viral disease in CB transplant recipients. This study is registered at www.clinicaltrials.gov as NCT00078533.


2018 ◽  
Vol 215 (2) ◽  
pp. 459-479 ◽  
Author(s):  
Ghada Ben Youssef ◽  
Marie Tourret ◽  
Marion Salou ◽  
Liana Ghazarian ◽  
Véronique Houdouin ◽  
...  

Mucosal-associated invariant T (MAIT) cells are semi-invariant Vα7.2+ CD161highCD4− T cells that recognize microbial riboflavin precursor derivatives such as 5-OP-RU presented by MR1. Human MAIT cells are abundant in adult blood, but there are very few in cord blood. We longitudinally studied Vα7.2+ CD161high T cell and related subset levels in infancy and after cord blood transplantation. We show that Vα7.2+ and Vα7.2− CD161high T cells are generated early during gestation and likely share a common prenatal developmental program. Among cord blood Vα7.2+ CD161high T cells, the minority recognizing MR1:5-OP-RU display a TRAV/TRBV repertoire very similar to adult MAIT cells. Within a few weeks of life, only the MR1:5-OP-RU reactive Vα7.2+ CD161high T cells acquire a memory phenotype. Only these cells expand to form the adult MAIT pool, diluting out other Vα7.2+ CD161high and Vα7.2− CD161high populations, in a process requiring at least 6 years to reach adult levels. Thus, the high clonal size of adult MAIT cells is antigen-driven and likely due to the fine specificity of the TCRαβ chains recognizing MR1-restricted microbial antigens.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3240-3240
Author(s):  
Sylvia Borchers ◽  
Michael Stadler ◽  
Susanne Luther ◽  
Tina Ganzenmueller ◽  
Brigitte Pabst ◽  
...  

Abstract Abstract 3240 Analysis of donor chimerism is a well established technique to monitor engraftment and detect pending relapse in patients after allogeneic hematopoietic stem cell transplantation (HSCT). Over the last decade, use of unrelated and/or mismatched donors as well as alternative grafts like cord blood (CB) has increased, and, in addition reduced intensity conditioning regimens are widely applied. Thus, recipients are increasingly being exposed to both persistent mixed chimerism and infectious complications because of delayed immune reconstitution. Donor chimerism is analyzed routinely from peripheral blood cells in all recipients. In addition, in a prospective study to evaluate usefulness of subset chimerism, T cell chimerism is analyzed in selected patients since 2007. Reconstitution of CMV-specific CD8 T cells (CMV-CTL) is monitored by multimers (multimeric dye-labeled recombinant-MHC-I-peptide-complexes) since 2006 to evaluate CMV-specific immune reconstitution post HSCT. Using this method, HLA-restriction of the multimers enables detection of residual recipient CMV-CTLs in mismatched transplantations. Interestingly, we found that CMV reactivation is accompanied by a decline in donor chimerism in some patients and that recipient CMV-CTLs persisting post HSCT expand upon CMV reactivation. Table 1 summarizes first data of this analysis in patients transplanted between 2007 and 2011 in our centre. Table 1: Data from patients transplanted between 2007 and 2011 at MHH # underlying disease R/D gender Donor Conditioning regimen GvHD prophylaxis Graft 1 AML f/f MMUD Flamsa(TBI)/ATG CSA/MMF PBPC 2 AML f/f MMRD Flu/Melph/Thiotepa/ATG TCD PBPC 3 AML m/m MMUD Flamsa(TBI)/Thymo CSA/MMF PBPC 4 NHL m/f MUD Flu/Cy/Thymo CSA/MMF PBPC 5 ALL m/m MMUD TBI/Cy/ATG CSA/MMF PBPC 6 AA f/m hla-ident sibl. Flu/Cy/TBI/Thymo CSA/MTX BM 7 MDS m/m MMUD Flu/Cy/TBI/ATG CSA/MMF cord blood 8 NHL m/m MMUD Flu/Cy/ATG CSA/MTX PBPC # R/D CMV-serostatus aGvHD CMV reactivation (CMV-R) leukocyte chimerism decline post CMV-R T cell chimerism decline post CMV-R persisting recipient CMV-CTLs 1 R+D- yes 40, 61 yes yes 2 R+D- no 27, 90, 188 no yes 3 R+D+ yes no no yes 4 R+D+ no 18 yes yes yes 5 R+D- suspected 34, 90 no no yes 6 R+D- no 55 yes yes 7 R+D- yes 85, 113 yes yes yes* 8 R+D- no (HvG) 33 yes yes yes* * confirmed by chimerism analysis in enriched CMV-CTL Patient 7 received a double mismatched cord-blood graft. As expected the recipient-CMV-CTLs declined after HSCT and by day +50 post-HSCT no CMV-CTLs (A*0201-NLVP multimer) could be detected anymore. The patient had a CMV reactivation on day +85 as shown by pp65 antigenemia assay. On day +90, 72 CMV-CTLs/μl were detected, further increasing to over 200/μl by day +152. To further analyze the origin and functionality, CMV-CTLs detected on day +90 were enriched by MACS to a purity of 97% in the CD3+CD8+ T cells. Donor chimerism was only 4%. After reconstitution of autologous CMV-CTLs, the patient experienced an additional subclinical CMV reactivation on day +113, not requiring any treatment at this time. In patient 8 a subclinical CMV reactivation on day +33 led to proliferation of CMV-CTLs and HLA-A*24 restricted and -B*35 restricted CMV-CTLs rose from 0 cells/μl (A*24 0.05%, B*35 0.04% of CD3CD8 T-cells) to 1 cell/μl and 21 cells/μl (A*24 0.28%, B*35 4.05% of CD3CD8 T-cells), respectively. Donor chimerism decreased from 51% on day +33 to 0% by day +62. Chimerism analysis of T-cell subsets on day +62 and of CMV-CTLs on day +69 revealed a 0% donor chimerism in these subsets. We speculate that in this patient CMV reactivation led to an inflammatory environment, which might have promoted loss of the graft. Our data indicate that T cell-subset chimerism analyses may contribute to a better understanding of chimerism kinetics. Furthermore, recipient-derived CMV-CTLs may be able to control CMV reactivation, especially after reduced intensity conditioning but also after standard conditioning regimens (i.e. in patient 5), but can severely influence donor chimerism and thus might have negative effects as well. We are currently investigating donor chimerism in T cell subsets and CMV-CTL reconstitution to gain insight into the complex immune responses and reconstitution processes occurring after allogeneic HSCT or CB-SCT. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Demi Brownlie ◽  
Inga Rødahl ◽  
Renata Varnaite ◽  
Hilmir Asgeirsson ◽  
Hedvig Glans ◽  
...  

AbstractRespiratory viral infections with SARS-CoV-2 or influenza viruses commonly induce a strong infiltration of immune cells into the lung, with potential detrimental effects on the integrity of the lung tissue. Despite comprising the largest fractions of circulating lymphocytes in the lung, little is known about how blood natural killer (NK) cells and T cell subsets are equipped for lung-homing in COVID-19 and influenza. Using 28-colour flow cytometry and re-analysis of published RNA-seq datasets, we provide a detailed comparative analysis of NK cells and T cells in peripheral blood from moderately sick COVID-19 and influenza patients, focusing on the expression of chemokine receptors known to be involved in leukocyte recruitment to the lung. The results reveal a predominant role for CXCR3, CXCR6, and CCR5 in COVID-19 and influenza patients, mirrored by scRNA-seq signatures in peripheral blood and bronchoalveolar lavage from publicly available datasets. NK cells and T cells expressing lung-homing receptors displayed stronger phenotypic signs of activation as compared to cells lacking lung-homing receptors, and activation was overall stronger in influenza as compared to COVID-19. Together, our results indicate migration of functionally competent CXCR3+, CXCR6+, and/or CCR5+ NK cells and T cells to the lungs in moderate COVID-19 and influenza patients, identifying potential common targets for future therapeutic interventions in respiratory viral infections.Author summaryThe composition of in particular CXCR3+ and/or CXCR6+ NK cells and T cells is altered in peripheral blood upon infection with SARS-CoV-2 or influenza virus in patients with moderate disease. Lung-homing receptor-expression is biased towards phenotypically activated NK cells and T cells, suggesting a functional role for these cells co-expressing in particular CXCR3 and/or CXCR6 upon homing towards the lung.


1983 ◽  
Vol 158 (2) ◽  
pp. 571-585 ◽  
Author(s):  
A Moretta ◽  
G Pantaleo ◽  
L Moretta ◽  
M C Mingari ◽  
J C Cerottini

In order to directly assess the distribution of cytolytic T lymphocytes (CTL) and their precursors (CTL-P) in the two major subsets of human T cells, we have used limiting dilution microculture systems to determine their frequencies. The two subsets were defined according to their reactivity (or lack thereof) with B9.4 monoclonal antibody (the specificity of which is similar, if not identical, to that of Leu 2b monoclonal antibody). Both B9+ and B9- cells obtained by sorting peripheral blood resting T cells using the fluorescence-activated cell sorter (FACS) were assayed for total CTL-P frequencies in a microculture system that allows clonal growth of every T cell. As assessed by a lectin-dependent assay, approximately 30% of peripheral blood T cells were CTP-P. In the B9+ subset (which represents 20-30% of all T cells), the CTL-P frequency was close to 100%, whereas the B9- subset had a 25-fold lower CTL-P frequency. It is thus evident that 90% and 10% of the total CTL-P in peripheral blood are confined to the B9+ or B9- T cell subsets, respectively. Analysis of the subset distribution of CTL-P directed against a given set of alloantigens confirmed these findings. CTL-P frequencies were also determined in B9+ and B9- subsets derived from T cells that had been activated in allogenic mixed leucocyte cultures (MLC). Approximately 10% of MLC T cells were CTL-P. This frequency was increased 3.5-fold in the B9+ subset, whereas the B9- subset contained only a small, although detectable number of CTL-P. Moreover, the great majority of the (operationally defined) CTL-P in MLC T cell population were found to be directed against the stimulating alloantigens, thus indicating a dramatic increase in specific CTL-P frequencies following in vitro stimulation in bulk cultures.


2009 ◽  
Vol 296 (5) ◽  
pp. G1054-G1059 ◽  
Author(s):  
Satoshi Kuboki ◽  
Nozomu Sakai ◽  
Johannes Tschöp ◽  
Michael J. Edwards ◽  
Alex B. Lentsch ◽  
...  

Helper T cells are known to mediate hepatic ischemia/reperfusion (I/R) injury. However, the precise mechanisms and subsets of CD4+ T cells that contribute to this injury are still controversial. Therefore, we sought to determine the contributions of different CD4+ T cell subsets during hepatic I/R injury. Wild-type, OT-II, or T cell receptor (TCR)-δ-deficient mice were subjected to 90 min of partial hepatic ischemia followed by 8 h of reperfusion. Additionally, wild-type mice were pretreated with anti-CD1d, -NK1.1, or -IL-2R-α antibodies before I/R injury. OT-II mice had diminished liver injury compared with wild-type mice, implicating that antigen-dependent activation of CD4+ T cells through TCRs is involved in hepatic I/R injury. TCR-δ knockout mice had decreased hepatic neutrophil accumulation, suggesting that γδ T cells regulate neutrophil recruitment. We found that natural killer T (NKT) cells, but not NK cells, contribute to hepatic I/R injury via CD1d-dependent activation of their TCRs, as depletion of NKT cells by anti-CD1d antibody or depletion of both NKT cells and NK cells by anti-NK1.1 attenuated liver injury. Although regulatory T cells (Treg) are known to suppress T cell-dependent inflammation, depletion of Treg cells had little effect on hepatic I/R injury. The data suggest that antigen-dependent activation of CD4+ T cells contributes to hepatic I/R injury. Among the subsets of CD4+ T cells, it appears that γδ T cells contribute to neutrophil recruitment and that NKT cells directly injure the liver. In contrast, NK cells and Treg have little effects on hepatic I/R injury.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 42-43
Author(s):  
Prajish Iyer ◽  
Lu Yang ◽  
Zhi-Zhang Yang ◽  
Charla R. Secreto ◽  
Sutapa Sinha ◽  
...  

Despite recent developments in the therapy of chronic lymphocytic leukemia (CLL), Richter's transformation (RT), an aggressive lymphoma, remains a clinical challenge. Immune checkpoint inhibitor (ICI) therapy has shown promise in selective lymphoma types, however, only 30-40% RT patients respond to anti-PD1 pembrolizumab; while the underlying CLL failed to respond and 10% CLL patients progress rapidly within 2 months of treatment. Studies indicate pre-existing T cells in tumor biopsies are associated with a greater anti-PD1 response, hence we hypothesized that pre-existing T cell subset characteristics and regulation in anti-PD1 responders differed from those who progressed in CLL. We used mass cytometry (CyTOF) to analyze T cell subsets isolated from peripheral blood mononuclear cells (PBMCs) from 19 patients with who received pembrolizumab as a single agent. PBMCs were obtained baseline(pre-therapy) and within 3 months of therapy initiation. Among this cohort, 3 patients had complete or partial response (responders), 2 patients had rapid disease progression (progressors) (Fig. A), and 14 had stable disease (non-responders) within the first 3 months of therapy. CyTOF analysis revealed that Treg subsets in responders as compared with progressors or non-responders (MFI -55 vs.30, p=0.001) at both baseline and post-therapy were increased (Fig. B). This quantitative analysis indicated an existing difference in Tregs and distinct molecular dynamic changes in response to pembrolizumab between responders and progressors. To delineate the T cell characteristics in progressors and responders, we performed single-cell RNA-seq (SC-RNA-seq; 10X Genomics platform) using T (CD3+) cells enriched from PBMCs derived from three patients (1 responder: RS2; 2 progressors: CLL14, CLL17) before and after treatment. A total of ~10000 cells were captured and an average of 1215 genes was detected per cell. Using a clustering approach (Seurat V3.1.5), we identified 7 T cell clusters based on transcriptional signature (Fig.C). Responders had a larger fraction of Tregs (Cluster 5) as compared with progressors (p=0.03, Fig. D), and these Tregs showed an IFN-related gene signature (Fig. E). To determine any changes in the cellular circuitry in Tregs between responders and progressors, we used FOXP3, CD25, and CD127 as markers for Tregs in our SC-RNA-seq data. We saw a greater expression of FOXP3, CD25, CD127, in RS2 in comparison to CLL17 and CLL14. Gene set enrichment analysis (GSEA) revealed the upregulation of genes involved in lymphocyte activation and FOXP3-regulated Treg development-related pathways in the responder's Tregs (Fig.F). Together, the greater expression of genes involved in Treg activation may reduce the suppressive functions of Tregs, which led to the response to anti-PD1 treatment seen in RS2 consistent with Tregs in melanoma. To delineate any state changes in T cells between progressors and responder, we performed trajectory analysis using Monocle (R package tool) and identified enrichment of MYC/TNF/IFNG gene signature in state 1 and an effector T signature in state 3 For RS2 after treatment (p=0.003), indicating pembrolizumab induced proliferative and functional T cell signatures in the responder only. Further, our single-cell results were supported by the T cell receptor (TCR beta) repertoire analysis (Adaptive Biotechnology). As an inverse measure of TCR diversity, productive TCR clonality in CLL14 and CLL17 samples was 0.638 and 0.408 at baseline, respectively. Fifty percent of all peripheral blood T cells were represented by one large TCR clone in CLL14(progressor) suggesting tumor related T-cell clone expansion. In contrast, RS2(responder) contained a profile of diverse T cell clones with a clonality of 0.027 (Fig. H). Pembrolizumab therapy did not change the clonality of the three patients during the treatment course (data not shown). In summary, we identified enriched Treg signatures delineating responders from progressors on pembrolizumab treatment, paradoxical to the current understanding of T cell subsets in solid tumors. However, these data are consistent with the recent observation that the presence of Tregs suggests a better prognosis in Hodgkin lymphoma, Follicular lymphoma, and other hematological malignancies. Figure 1 Disclosures Kay: Pharmacyclics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Oncotracker: Membership on an entity's Board of Directors or advisory committees; Rigel: Membership on an entity's Board of Directors or advisory committees; Juno Theraputics: Membership on an entity's Board of Directors or advisory committees; Agios Pharma: Membership on an entity's Board of Directors or advisory committees; Cytomx: Membership on an entity's Board of Directors or advisory committees; Astra Zeneca: Membership on an entity's Board of Directors or advisory committees; Morpho-sys: Membership on an entity's Board of Directors or advisory committees; Tolero Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol Meyer Squib: Membership on an entity's Board of Directors or advisory committees, Research Funding; Acerta Pharma: Research Funding; Sunesis: Research Funding; Dava Oncology: Membership on an entity's Board of Directors or advisory committees; Abbvie: Research Funding; MEI Pharma: Research Funding. Ansell:AI Therapeutics: Research Funding; Takeda: Research Funding; Trillium: Research Funding; Affimed: Research Funding; Bristol Myers Squibb: Research Funding; Regeneron: Research Funding; Seattle Genetics: Research Funding; ADC Therapeutics: Research Funding. Ding:Astra Zeneca: Research Funding; Abbvie: Research Funding; Octapharma: Membership on an entity's Board of Directors or advisory committees; MEI Pharma: Membership on an entity's Board of Directors or advisory committees; alexion: Membership on an entity's Board of Directors or advisory committees; Beigene: Membership on an entity's Board of Directors or advisory committees; DTRM: Research Funding; Merck: Membership on an entity's Board of Directors or advisory committees, Research Funding. OffLabel Disclosure: pembrolizumab


Sign in / Sign up

Export Citation Format

Share Document