scholarly journals Functionally active virus-specific T cells that target CMV, adenovirus, and EBV can be expanded from naive T-cell populations in cord blood and will target a range of viral epitopes

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 ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2238-2238
Author(s):  
Paul Szabolcs ◽  
Young-Ah Lee ◽  
Luciana Marti ◽  
Melissa Reese ◽  
Joanne Kurtzberg

Abstract Introduction: Unrelated umbilical cord blood transplantation (UUCBT) is a viable option for those who lack HLA-matched sibling donors. However, opportunistic infections (OI) occurring in the first 100 days, remain the major cause of morbidity and mortality. Viral infections are the primary cause of OI death. As previously shown, cord blood T cells have significantly less preformed effector molecules available intracellularly to kill virally infected cells via the perforin-granzyme pathway than adult PB T cells. Since several virally infected patients control their infections in the absence of specific antiviral therapy ( e.g adenoviral enteris, polyoma cystitis) we postulated that the T cell compartment of those UUCBT recipients who experience early viral infections maty upregulate expression of the perforin exocytosis pathway. In parallel the impact of viral infection on T cell turnover would also be appreciable. Here we report on 19 prospectively studied pediatric patients, all full donor chimera, following myeloablative therapy. Methods: On day +50 we determined by 4-color FACS the expression of intracellular Granzyme, A, B, along with perforin. To monitor T cell turnover proliferating cells were identified by monitoring for the KI-67 nuclear antigen. The expression of the antiapoptosis gene BCL-2 was similarly monitored in both CD4+ and CD8+ T cells. We analyzed their association with the development of de novo OI up to day +100 employing Student’s t-test. Results: Mean age of patients was 6.2 years. 10 of 19 patients developed OI (adenovirus x 4, CMV x 7, EBVx1, parainfluenza x 1) with 5/10 patients experiencing more than one viral infections simultaneously) at a median of 29 days after UUCBT. Of those with OI 6/10 died due to their infections while 8/9 without OI are alive at a median of 15.8 months after UUCBT with one death due to leukemic relapse. Table I presents the correlation between the tested parameters with the development of OI. Patients experiencing viral infections had significantly higher % of their T cells in particular CD8+ T cells equipped with effectors of cytotoxicity and were proliferating in higher percentage compared to those with no active infections. However, the anti-apoptotic protein BCL-2 expression was significantly lower in patients experiencing OI that may lead to their shorter life span and overall T cell lymphopenia observed in OI patietns that we have previously detected in a larger cohort of 102 patients (ASBMT 2004 abstract#48). Conclusion: Correlating with active viral infections significant maturation of cord blood T cells is evident as early as 50 days after UUCBT towards acquiring effector molecules of the perforin pathway. Enhanced T cell proliferation is counteracted by reduced expression of BCL-2 that may lead to the lymphopenia in patients with OI. Future strategies aiming to enhance the longevity of antiviral T cells may protect from death due to viral infections. Univariate analysis VARIABLE MEDIAN VALUE FOR PATIENTS WITH OI MEDIAN VALUE FOR PATIENTS WITHOUT OI t-Test p value % Granzyme A+ T cells 52% 9% 0.006 % Granzyme A+ CD8+ T cells 91% 47% <0.001 % Granzyme B+ T cells 36% 6% 0.036 % Granzyme B+ T cells 87% 39% <0.001 % Perforin+ T cells 38% 4% 0.009 % Perforin+ CD8+ T cells 61% 21% <0.001 % Ki-67+ T cells 27% 16% 0.0041 % Ki-67+ CD8+ T cells 35% 16% 0.0037 BCL-2 expression level (MFI) 87 117 0.028


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.


2014 ◽  
Vol 89 (2) ◽  
pp. 1058-1069 ◽  
Author(s):  
T. W. H. Flinsenberg ◽  
L. Spel ◽  
M. Jansen ◽  
D. Koning ◽  
C. de Haar ◽  
...  

ABSTRACTReactivation of human cytomegalovirus (CMV) is hazardous to patients undergoing allogeneic cord blood transplantation (CBT), lowering survival rates by approximately 25%. While antiviral treatment ameliorates viremia, complete viral control requires CD8+T-cell-driven immunity. Mouse studies suggest that cognate antigen-specific CD4+T-cell licensing of dendritic cells (DCs) is required to generate effective CD8+T-cell responses. For humans, this was not fully understood. We here show that CD4+T cells are essential for licensing of human DCs to generate effector and memory CD8+T-cell immunity against CMV in CBT patients. First, we show in CBT recipients that clonal expansion of CMV-pp65-specific CD4+T cells precedes the rise in CMV-pp65-specific CD8+T cells. Second, the elicitation of CMV-pp65-specific CD8+T cells from rare naive precursors in cord blood requires DC licensing by cognate CMV-pp65-specific CD4+T cells. Finally, also CD8+T-cell memory responses require CD4+T-cell-mediated licensing of DCs in our system, by secretion of gamma interferon (IFN-γ) by pp65-specific CD4+T cells. Together, these data show that human DCs require licensing by cognate antigen-specific CD4+T cells to elicit effective CD8+T-cell-mediated immunity and fight off viral reactivation in CBT patients.IMPORTANCESurvival rates after stem cell transplantation are lowered by 25% when patients undergo reactivation of cytomegalovirus (CMV) that they harbor. Immune protection against CMV is mostly executed by white blood cells called killer T cells. We here show that for generation of optimally protective killer T-cell responses that respond to CMV, the early elicitation of help from a second branch of CMV-directed T cells, called helper T cells, is required.


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 ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2352-2352
Author(s):  
Nicole J. Gormley ◽  
Aleah Smith ◽  
Maria Berg ◽  
Lisa Cook ◽  
Catalina Ramos ◽  
...  

Abstract Abstract 2352 Introduction/Methods: The administration of highly purified haploidentical peripheral blood CD34+ cells combined with an unrelated cord blood transplant results in earlier neutrophil engraftment than is typically seen with a cord blood transplant alone. Chimerism data from pilot trials evaluating this strategy have reported 3 phases of engraftment: 1) early myeloid engraftment from transplanted haplo-CD34+ cells followed by 2) cord blood engraftment resulting in dual chimerism and 3) the subsequent disappearance of haploidentical donor cells with resultant full donor cord chimerism. The mechanism accounting for the disappearance of haploidentical cells has not been defined. Here the clinical results and an in vitro assessment of alloreactivity in three patients that underwent combined haploidentical CD34+ cell and cord blood transplantation for severe aplastic anemia (SAA) are described. The conditioning regimen consisted of cyclophosphamide (60mg/kg/day on days -7 and -6), fludarabine (25mg/m2/day on days -5 to -1), horse ATG (40mg/kg/day on days -5 to -2), and total body irradiation (200cGy on day -1). GVHD prophylaxis consisted of tacrolimus and mycophenolate mofetil. PCR of STRs was used to assess chimerism in T-cell and myeloid lineages and mixed lymphocyte reaction assays(MLR) were performed on peripheral blood samples collected at different time-points post-transplant to assess for alloreactivity against the recipient, the haploidentical donor, or the cord unit. Stimulator cord blood cells for the MLR were obtained from residual cord blood cells remaining in the infusion bag after patient administration and expanded in vitro using anti-CD28/CD3 Dynabeads. Results: Prior to transplantation, all three pts had transfusion dependent SAA associated with severe neutropenia that was refractory to conventional immunosuppressive therapy. Pt 1 had an early transient myeloid recovery (ANC 400 on day+11) from the haploidentical donor followed by engraftment of the cord unit (Cord ANC > 500) on day 21. The patient is currently 2 years post transplant and has 100% cord blood chimerism and is transfusion independent. An MLR assay performed when donor T-cell chimerism was 100% cord, showed evidence for rejection of the haploid cells by cord blood T-cells, with the MLR response to haploidentical donor cells being seven fold higher than the response to fully HLA-mismatched 3rd party cells. In pt 2, neutrophil recovery from the transplanted haploidentical donor occurred on day +10, with chimerism studies showing no evidence for cord engraftment in either myeloid or T-cell lineages at any point post-transplant. The patient is currently 15 months post transplant and is transfusion independent with normal blood counts and sustained “split” chimerism (T-cells recipient in origin with myeloid cells being 100% haploidentical donor). MLR assays showed that the recipient was tolerant to the haploid donor, with no statistically significant difference in the alloreactive response to the haploid donor compared to self. In pt 3, neutrophil recovery from the transplanted haploidentical donor occurred on day +10, with chimerism studies showing split chimerism (T-cell chimerism >90% cord and myeloid chimerism 88–100% haploid donor in origin). MLR assays again showed evidence of rejection of the haploid cells by cord blood T-cells, with a trend towards greater alloreactivity against the haploid donor compared to an HLA mismatched 3rd party on post-transplant day +63. Conclusions: Combined haploidentical CD34+ cell and unrelated cord blood transplantation following highly immunosuppressive conditioning represents a viable treatment option for patients with SAA who lack an HLA-matched donor. Using this approach, 2 of 3 pts had cord blood engraftment associated with early neutrophil recovery from the haploidentical donor. In one pt, the cord unit failed to engraft. Remarkably, sustained engraftment from the haploidentical donor in this pt resulted in transfusion independence. MLR appears to be a useful approach to assess the in vitro alloreactivity of this unique stem cell graft source. In the two pts who had cord engraftment, in vitro MLR assessments established that the disappearance of haploid cells occurred as a consequence of rejection of the haploidentical cells by engrafting cord blood T-cells, rather than from non-immunological haploidentical cell graft failure. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (13) ◽  
pp. 4543-4551 ◽  
Author(s):  
Krishna V. Komanduri ◽  
Lisa S. St. John ◽  
Marcos de Lima ◽  
John McMannis ◽  
Steven Rosinski ◽  
...  

Advances in immune assessment, including the development of T-cell receptor excision circle (TREC) assays of thymopoiesis, cytokine-flow cytometry assays of T-cell function, and higher-order phenotyping of T-cell maturation subsets have improved our understanding of T-cell homeostasis. Limited data exist using these methods to characterize immune recovery in adult cord blood (CB) transplant recipients, in whom infection is a leading cause of mortality. We now report the results of a single-center prospective study of T-cell immune recovery after cord blood transplantation (CBT) in a predominantly adult population. Our primary findings include the following: (1) Prolonged T lymphopenia and compensatory expansion of B and natural killer (NK) cells was evident; (2) CB transplant recipients had impaired functional recovery, although we did observe posttransplantation de novo T-cell responses to cytomegalovirus (CMV) in a subset of patients; (3) Thymopoietic failure characterized post-CBT immune reconstitution, in marked contrast to results in other transplant recipients; and (4) Thymopoietic failure was associated with late memory T-cell skewing. Our data suggest that efforts to improve outcomes in adult CB transplant recipients should be aimed at optimizing T-cell immune recovery. Strategies that improve the engraftment of lymphoid precursors, protect the thymus during pretransplant conditioning, and/or augment the recovery of thymopoiesis may improve outcomes after CBT.


Blood ◽  
2011 ◽  
Vol 118 (16) ◽  
pp. 4480-4488 ◽  
Author(s):  
Natacha Merindol ◽  
Martin A. Champagne ◽  
Michel Duval ◽  
Hugo Soudeyns

Abstract Recipients of umbilical cord blood (UCB) transplantation (UCBT) face a high risk of morbidity and mortality related to opportunistic infections (OI) and leukemic relapse. To understand the molecular basis of these UCBT-related complications, the characteristics of UCB-derived antigen-specific CD8+ T cells were examined in a group of pediatric UCBT recipients. Compared with the UCB graft inoculum and the late post-UCBT period (12-36 months), declining clonal diversity of UCB-derived CD8+ T cells specific for the Melan-A26-35 A27L peptide and high frequencies of PD-1-expressing CD8+ T cells were observed in the first 3 months after UCBT, a period during which OIs are most frequent. The CD8+ T-cell compartment predominantly comprised CD45RA+ CCR7− terminally differentiated effector-memory T cells until 6 months after UCBT, at which time the polyfunctionality of antigen-specific CD8+ T cells was reestablished. Finally, the frequency of PD-1+ CD8+ T cells was significantly higher in subjects who subsequently experienced leukemic relapse. This study informs the biologic properties of UCB-derived CD8+ T cells and provides a rationale for the characteristics of UCBT in terms of immune reconstitution and OI. These results also suggest that the elevated frequency of PD-1+ CD8+ T cells could be associated with leukemic relapse in pediatric UCBT recipients.


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.


Sign in / Sign up

Export Citation Format

Share Document