Comparison of Early Post-Transplant Outcomes in Ex Vivo αβ+ T Cell-Depleted Haploidentical HSCT with or without Pharmacologic Gvhd Prophylaxis

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3379-3379 ◽  
Author(s):  
Eun Seok Choi ◽  
Sung Han Kang ◽  
Hyery Kim ◽  
Kyung-Nam Koh ◽  
Ho Joon Im ◽  
...  

Abstract Background and purpose: One of the key obstacles to successful haploidenitcal hematopoietic cell transplantation (HHCT) is a development of fatal GVHD. Although much progress in immunosuppressant (IS) has effectively prevented the development of acute GVHD, they have many serious toxicity and drug interactions requiring serial monitoring of drug levels. Recent advances in ex vivo depletion technique enabled to effectively reduce T cells or their subset, αβ+ T cells, leading to residual αβ+ T cells in grafts well below 5×104/kg of recipient weight. We eliminated post-transplant pharmacologic GVHD prophylaxis along with targeting αβ+ T cell dose ≤ 5×104/kg since November 2015. In this study, we compared early post-transplant outcomes between with (IS+) or without (IS-) post-transplant immunosuppressants after ex vivo αβ+ T cell-depleted HHCT. Methods: Between May 2012 and July 2016, 69 pediatric patients received HHCT using TCRαβ-depleted grafts from haploidentical family donors at Asan Medical Center Children's Hospital. Fifty patients received tacrolimus and mycophenolate mofetil to prevent acute GVHD, while 19 did not receive any immunosuppressant after transplant. All donors received G-CSF for 4 consecutive days and peripheral blood stem cells were collected on days -1 and 0. The αβ+ T cells were depleted by negative selection using the CliniMACS® system (Miltenyi-BioTec, Bergisch-Gladbach, Germany) according to manufacturer's instruction. In the earlier trial of IS+, the final doses of αβ+ T cells were adjusted to 1-5×105 cells/kg by add-back from the raw bag. Since November 2015, the cell dose was targeted at ≤ 5×104 αβ+T cells/kg with no post-transplant immunosuppressants (IS-). Results: The median infused CD34+ cells, αβ+ T cells, γδ+ T cells and CD3-CD56+ NK cells per kg of recipient weight were 8.9×106, 33.8×104, 20.0×106, 45.9×106 in IS+ group and 6.1×106, 4.6×104, 17.5×106, 24.6×106 in IS- group, respectively. All 69 patients achieved neutrophil engraftment at a median of 10 days (range, 9-17). Three patients out of 50 in IS+ group experience graft rejection (GR), while no GR occurred in IS- group. The cumulative incidences of acute GVHD grade II-IV were similar (31% vs 33%). Severe acute GVHD ≥ grade III developed in 7 in IS+ group, while none in IS- group developed ≥ grade III. As of July 2016, the median follow-ups were 24 months (range 9.5-50.8) for IS+ group and 5 months (0.5-9.1) for IS- group. Two out of 50 patients in IS+ group died of TRM leading to 2.2% at 6 months and 4.9% at 1 year after HHCT, while no patients in IS- group died of TRM during the follow-up period. The mean time from transplant to discharge were longer in IS+ group compared to IS- group (32 days versus 21 days, P=0.049). While the mean time of hospital stay within 100 days post-HHCT for patients who survived more than 100 days was not different between two groups (47 days versus 34 days, P>0.05). Conclusions: The major findings of our study were less severe acute GVHD and shorter hospital stay from HHCT to discharge in IS- group, even with less T cell dose, compared to IS+ group. Therefore, this HHCT using ex vivo αβ-depleted graft containing αβ+ T cells ≤ 5×104/kg is an effective treatment strategy to prevent acute GVHD without post-transplant IS. In addition, the early clinical outcomes were comparable between with and without post-transplant IS. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4078-4078
Author(s):  
Leslie S. Kean ◽  
Amelia Langston ◽  
Muna Qayad ◽  
H. Jean Khoury ◽  
Divya Tiwari ◽  
...  

Abstract Abstract 4078 Background: Acute GvHD remains the major cause of complications and death following unrelated-donor HSCT. In a non-human primate model, we have previously shown that in vivo costimulatory blockade of donor T-cells could provide effective protection against GVHD. To begin to explore its clinical utility, we are conducting a trial (Clinical Trials.Org # NCT01012492) to determine the feasibility of combining abatacept (CTLA4-Ig) with cyclosporine and methotrexate as acute GVHD prophylaxis for patients undergoing unrelated marrow and peripheral blood stem cell transplants for hematologic malignancies. Methods: Patients older than 12 with advanced hematologic malignancies, conditioned with either TBI/Cytoxan, Busulfan/Cytoxan or Fludarabine/Melphalan are eligible. Abatacept is administered IV on days −1, +5, +14, and +28 at 10 mg/kg in addition to standard GvHD prophylaxis consisting of cyclosporine (day −2 to day 100), and methotrexate (15 mg/m2 on day +1 and 10 mg/m2 on days +3, 6 and 11). Patients are then followed for clinical outcomes and immunologic reconstitution through day +365. Results: 9 patients (planned enrollment = 11 patients) have thus far been enrolled on the study of which 5 are evaluable for engraftment, toxicity and acute GvHD. The other four patients consist of 2 who are currently receiving abatacept, 1 who was discovered to have an ongoing viral infection at the start of the first abatacept infusion so was removed from the treatment regimen, and 1 who is awaiting transplant. The median age for the 5 evaluable patients is 47 years (17–74 years). 3 patients had AML and 2 had ALL. Patients were conditioned with Bu/Cy (n=1), TBI/Cy (n=2) and Flu/Melphalan (n=2). 4 donor-recipient pairs were allele matched at 9 of 10 loci (A, B, C, DRB1 and DQB1), while 1 was fully matched. Four of the 5 patients are currently alive and in remission and 1 relapsed at day +98 (and died on day +121 with refractory AML). The four other patients are surviving without relapse with a follow-up of 155–313 days. All 5 patients received the 4 scheduled abatacept doses. No infusional side effects were noted. All patients achieved neutrophil engraftment (median day +20 (11–47). 4 of 5 patients have achieved platelet engraftment (median day +27 (14–35). Donor engraftment (100% CD33 and 99–100% CD3 at Day +30) occurred in all cases. All patients have demonstrated rapid lymphocyte engraftment, with the mean ALC reconstituting to >500 cells/μL by day +21 post-transplant. At day +100, the mean CD3+ count was 673 +/− 251 cells/μL. Both CD8+ and CD4+ T cells reconstituted by day 100, with the mean CD8+ count = 384 +/− 148 cells/μL and the mean CD4+ count = 229 +/− 119 cells/μL. T cell reconstitution was accompanied by a shift away from naïve (Tn, CCR7+/CD45RA+) toward a CCR7-/CD45RA- effector memory (Tem)-predominant phenotype. Thus, the average proportion of CD4+ Tem cells in the recipient increased from 22 +/− 6% pre-transplant to 46 +/− 7% at day +100 with a concomitant loss of CD4+ Tn cells. Likewise, the proportion of CD8+ Tem also significantly increased, from an average of 15 +/− 4% pre-transplant to 32 +/− 7% at day +100, also with a reciprocal decrease in CD8+ Tn cells. One patient developed steroid responsive grade 3 acute GVHD involving the skin and the liver, followed by steroid responsive liver chronic GvHD. This patient is currently weaning corticosteroids. Another patient developed steroid responsive late-onset (day +217) acute GVHD (liver and GI) during cyclosporine weaning, which was also steroid responsive, and is also currently weaning corticosteroids. No other systemic acute or chronic GvHD has occurred. No unexpected complications or life-threatening infections were observed. 3 patients have experienced 5 episodes of CMV reactivation, all responsive to antiviral therapy. One patient developed polyclonal EBV-related PTLD (plasmacytic hyperplasia) in the absence of EBV viremia, which regressed without intervention. No other EBV-related disease has occurred. Conclusions: These preliminary data suggest that abatacept can be safely added to cyclosporine and methotrexate for GVHD prophylaxis in recipients of hematopoietic grafts from unrelated donors, with encouraging rates of acute GVHD. As such, they support the conduct of a larger, randomized phase 2 study. Disclosures: Off Label Use: Abatacept: It is an immunosuppressive agent that targets the CD28/B7 T cell costimulation pathway. It is approved for use in Rheumatoid arthritis.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 439-439
Author(s):  
Aaron P. Rapoport ◽  
Edward A. Stadtmauer ◽  
Bruce L. Levine ◽  
Ashraf Badros ◽  
Gorgun Akpek ◽  
...  

Abstract Ex-vivo co-stimulation of autologous T-cells with anti-CD3/anti-CD28-conjugated magnetic beads followed by adoptive transfer may augment T-cell responses toward tumor antigens or infectious agents. 54 patients (pts) were treated with ex-vivo co-stimulated autologous T-cells after autotransplantation for myeloma. The median age was 56 (range 38–71), 67% were male, 20% were African-American, 22% had IgA paraproteins, 11% had del 13 or complex karyotypes and the median β2m level at diagnosis was 3.31 mg/L (range 1.09–73.7). After lymphocyte collections, pts received cyclophosphamide (4.5 g/m2) + G-CSF for stem cell mobilization, and high dose melphalan (200 mg/m2 or 140 mg/m2 for pts ≥ 70) for conditioning. T-cells were cultured for ~12 days with anti-CD3/anti-CD28-immobilized immunomagnetic beads + IL-2 supplementation (100 units/ml). During a run-in phase, 12 pts received co-stimulated T-cells post-transplant ( day +12) alone. In a second phase, 42 pts who participated in a 2 x 2 randomization, received T-cells either early (day +12) or late (day + 100) after transplant. These pts also received either 2 immunizations with the pneumococcal conjugate vaccine (PCV, Prevnar) at days +30, +90 or 3 immunizations (prior to T-cell collection, at days + 30, + 90) to test immune responses to a well-defined antigen. 42 pts received PCV immunizations with no grade 3/4 adverse events. Anti-pneumococcal antibody responses developed in 51% of 31 pts tested thus far. Details of these studies will be presented separately. 52 pts received a mean dose of 8.04 x 109 costimulated T-cells (range 1.6–11). Infusion-related adverse effects included grade I–II rigors/chills (40%), grade I–II facial/upper body rashes (12%) at a median of 13 days after T-cells, grade I cardiovascular events (10%), grade I–III hypoxia (5%), grade II fever (5%), and 1 episode of DVT. At T-cell harvesting, the mean % of CD3 + cells in culture was 94.8%, the mean T-cell doubling level was 4.81 (28-fold expansion). Among the 42 randomized pts, at day + 42 post-transplant ( 30 days after T-cell infusion for the early groups), the median CD4/CD3 count was 462/mcl (range 202–1439) for the early T-cell recipients vs 230/mcl (range 50–915) for the late T-cell recipients (T-cells not yet infused) [P=0.004]. The median CD8/CD3 counts were 1399/mcl (range 465–2810) vs 1084/mcl (range 103–3422) for the early and late T-cell recipients respectively at day +42 [P=0.04]. For clinical responses there were 11 CRs, 22 VGPRs ( 90% reduction in paraprotein levels), 18 PRs (50–90% reductions), 2 pts had no response and 1 pt was unevaluable. 2 pts had delayed reductions in M-protein levels of ~ 50% between day 42 and day 180 or 270. For the entire cohort, the probability of overall survival at 1.5 years was 78% [95%CI 64%–92%]. Infusions of ex-vivo expanded autologous T-cells are well-tolerated and associated with accelerated T-cell recovery early after autotransplantation. This study may provide a platform for combining costimulated T-cells and tumor vaccines in the autograft setting.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3020-3020 ◽  
Author(s):  
Denis-Claude Roy ◽  
Mireille Guerin ◽  
Radia Sidi Boumedine ◽  
Silvy Lachance ◽  
Sandra Cohen ◽  
...  

Abstract Abstract 3020FN2 Background: The intensive T-cell depletion of the graft accompanying haploidentical stem cell transplantation (SCT) delays immune reconstitution and results in frequent and rapidly lethal infectious complications. The ability to accelerate immune reconstitution following HLA-haploidentical-SCT would extend safe transplantation to the large number of patients who do not have an HLA-matched donor. Methods: Twenty-seven adults with very high-risk malignancy entered a Phase I clinical trial of haplo-identical T-cell depleted allogeneic SCT followed by an immunotherapeutic strategy consisting of alloreactive T-lymphocyte depleted cells to accelerate immune reconstitution (ATIR) while preventing graft-versus-host disease (GVHD). Selective elimination of host-reactive T cells was achieved using a dibromorhodamine-based photodepletion approach. All stem cell grafts underwent in vitro immunomagnetic T cell depletion using CD34+ positive cell selection (Miltenyi). The myeloablative regimen consisted of TBI (1200 cGy), thiotepa (5 mg/kg), ATG (12.5 mg/kg) and fludarabine (200 mg/m2). No GVHD prophylaxis was administered. Results: Eight patients were enrolled and subsequently removed from the study because of leukemia relapse (n=4) or late identification of an unrelated donor (n=4). All 8 patients died. Nineteen patients (11 M, 8 F) with very high-risk hematologic malignancies (mostly refractory or relapsed acute myeloid leukemia (10) and myelodysplastic syndromes (4), and refractory biphenotypic leukemia (1), CLL (2), CML (1) and NHL (1)) proceeded with the trial. Median age was 54 years (range: 20–62). Patient and disease characteristics were similar between patient cohorts. Patients received incremental doses of ATIR cells, from 1×104 to 5×106 CD3 cells/kg at a median of 30 days (range: 28–39) after SCT. Greater than 90% of activated (CD25+CD44+) CD4 and CD8 T cells (p<0.004) and anti-host cytotoxic T lymphocyte precursors (CTLp) (p=0.0008) were depleted from the donor lymphocyte infusions (DLI). Interferon- γ responses against CMV, EBV and Influenza peptides were maintained post-photodepletion. Naive (TNaive) and central memory (TCM) T cells were also preserved, but mature effector memory populations (TEMRA) decreased after photodepletion. All patients showed complete donor chimerism and durable hematologic engraftment. No patients developed grade III-IV acute GVHD. Acute GVHD grade II developed in 4 patients at a median of 102 days post-transplant (range 45–125), 3 of these belonging to the 2 highest dose groups (2.0–5.0×106 CD3 cells/kg). Five patients developed de novo extensive chronic GVHD at a median of 4.8 months post-transplant. All patients responded rapidly to oral immunosuppression lasting for a median of 6.3 months, with only one patient treated for up to 17 months. NK (CD56) and B (CD19) cells were the first to recover, approximately 4 weeks post-transplant. Patients administered the highest T-cell doses (2.0–5.0×106 CD3 cells/kg) showed earlier reconstitution of CD3, CD4 and CD8 cells (all p≤0.02). The administration of increasing ATIR cell doses yielded gradually increasing proportions of TNaive of CD4 phenotype in the first 18 weeks, and of CD8 phenotype between weeks 18 and 36 post-ATIR. Time to the first infection was delayed in patients receiving high ATIR doses (p=0.015). During the first 6 months post-ATIR, the number of patients without infections was only 1 (14%) of the 7 low T-cell doses patients but increased to 8 (67%) in the high T-cell dose group (p=0.027). The overall survival was 47.4±22.4% (±95% confidence interval) and the event-free-survival 36.8±21.7% at 2 years, with a median follow-up of survivors of 4.0 years (range 3.1–5.1 years). Transplant-related mortality was decreased in the high (19.2%) versus the low ((65.7%) T-cell dose group, with a similar trend for improved survival in the high T-cell dose group (p=0.078). Conclusions: Post-transplant immunotherapy with photodepleted DLI decreased the incidence and severity of infections without inducing severe GVHD. These results document the benefits of administrating selectively depleted T-cells after haploidentical transplantation. Disclosures: Roy: Kiadis Pharma: Research Funding. Mielke:Kiadis Pharma: Research Funding. Egeler:Kiadis Pharma: Employment.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 657-657
Author(s):  
Caroline R. Cantilena ◽  
Sawa Ito ◽  
Xin Tian ◽  
Prachi Jain ◽  
Fariba Chinian ◽  
...  

Abstract INTRODUCTION: Ex-vivo T cell depletion strategies have been widely used to reduce the incidence of graft versus host disease (GVHD) in allogeneic stem cell transplantation (allo-SCT). Although several options of ex-vivo graft manipulation strategy are available, direct comparison between strategies along with relevant biomarkers has been lacking. Here we evaluated cellular and plasma biomarkers in two separate graft manipulation strategies, CD3-CD19 depletion versus CD34+ selection using the Miltenyi CliniMACS and their association with clinical outcomes. METHODS: Forty two subjects with hematological malignancies underwent HLA matched sibling allo-SCT at a single center between 2012 and 2015 and received either an ex-vivo CD3-CD19 depleted, CD34+ negatively selected graft (CD3/19D, n=20) or an ex-vivo CD34+ cell positively selected graft (CD34S, n=22). Both cohorts were treated with the same conditioning regimen of cyclophosphamide, fludarabine, and total body irradiation (600-1200 cGy) and GVHD prophylaxis of low dose cyclosporine. Peripheral blood mononuclear cells and plasma samples were collected at days 14 or 30, 60, 100 post-transplant. Post-transplant cellular immune reconstitution was evaluated by multi-color flow cytometry immunophenotyping, characterizing the subsets of memory T cells, regulatory T cells (Tregs), natural killer (NK) cells, and B cells with various functional markers. The plasma levels of ST2, Reg3α, and sTNFR1 were measured using enzyme-linked immunosorbent assay (ELISA). RESULTS: The median age at transplant was 48 years (range 17-70) in CD3/19D and 45 years (11-73) in CD34S. At a median follow up of 37 months in CD3/19D and 22 months in CD34S, the major clinical outcomes were similar between two groups; the overall survival (70% and 86%), non-relapse mortality (5% and 4.5%), and cumulative incidence of relapse (35% and 39%) at 2 years, respectively. Two subjects in CD3/19D developed late engraftment failure before day 100 but all other subjects achieved primary neutrophil and platelet recovery. Unexpectedly, the cumulative incidence of grade II-IV acute GVHD was higher in CD3/19D (61%) in comparison to the incidence in CD34S (32%, P=0.07, Figure). The cumulative incidence of extensive chronic GVHD was 33% in CD3/19S and 24% in CD34S. The fraction of Helios negative Tregs post-transplant was significantly lower in CD3/19D (median [interquartile range]: 10.4% [7.1-16.4] at day 30; 4.9% [3.0-8.3] at day 60) compared to CD34S (23.8% [10.7-35.8], P=0.03 at day 30; 8.8% [6.8-18.4], P=0.01 at day 60, Figure). Plasma ST2 levels were significantly higher in CD3/19D (45ng/mL [27-67] at day 14; 33ng/mL [27-62] at day 28) in comparison to CD34S (29ng/mL [19-40], P=0.03 at day 14; 25ng/mL [14-33], P=0.03 at day 28, Figure). In addition, significantly higher CD4 naive T cells, lower effector memory and PD-1 bright CD4 T cells were observed in CD3/19D in comparison to CD34S. NK and B cell profiles were not significantly different between the two groups. CONCLUSION: Both methods of ex vivo TCD were associated with extremely low NRM rates (~5%).We observed a higher cumulative incidence of acute GVHD in the recipients of CD3/19 depleted grafts, accompanied with the distinct biomarker profiles of poor Treg reconstitution and high level of ST2. CD3/19 depletion may have disproportionately depleted Tregs in the graft, leading to uncontrolled tissue damage and GVHD evidenced by higher ST2 levels. Further validation is required to confirm the utility of monitoring Treg reconstitution and ST2 level as biomarkers to predict the outcomes of T cell depleted allo-SCT. Figure 1. Figure 1. Disclosures Battiwalla: NIH/NHLBI: Employment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2904-2904 ◽  
Author(s):  
R. Lowsky ◽  
K. Heydari ◽  
B. Sahaf ◽  
J. Shizuru ◽  
G. Laport ◽  
...  

Abstract Murine models of transplantation established that nonmyeloablative conditioning using repeated low doses of irradiation targeted to lymphoid tissues (TLI) and depletive anti-T cell antibodies protects against GVHD by skewing residual host T cell subsets to favor regulatory natural killer (NK) T cells that suppress GVHD by polarizing donor T cells toward secretion of non-inflammatory cytokines such as IL-4. We recently translated the murine protocol to a clinical study using non-myeloablative TLI and ATG host conditioning with HLA matched related and unrelated donors, and showed a marked reduction in the incidence of acute GVHD while retaining graft anti-tumor activity (Lowsky et al., in Press NEJM). Engrafted donor CD4+ T cells showed a marked increase in IL-4 production as compared to CD4+ T cells from controls. We now adapted the TLI and ATG nonmyeloablative host conditioning regimen to a clinical study of allogeneic HCT using haploidentical matched (3/6 HLA matched) related donors to determine if it will result in donor hematopoietic cell engraftment and also protect against acute GVHD. Blood derived hematopoietic progenitor cells were collected by apheresis from donors mobilized with G-CSF and the product was T cell depleted using CD34+ selection. CD3+ T cells were added back to the donor inoculum according to a dose escalation schedule. The initial T cell dose was 1 x105 CD3+ cell/kg with designated increments based on clinical outcomes of up to a maximum of 1 x107 CD3+ cells/kg. The desired CD34+ cell dose was >5 x 106 CD34+ cells/kg for all patients. Seven patients were transplanted; the median age was 53 years (range 27 to 61 years). Five patients had acute myelogenous leukemia, two with disease in remission and three not in remission at the start of TLI and ATG, one with myelodysplastic syndrome, and one with progressive peripheral T cell lymphoma. The median follow-up for all patients is 265 days with three of seven patients alive and free of disease at the last observation period. Sustained donor hematopoietic cell engraftment was achieved in three of three patients only after the T cell dose was increased to 1 x107 CD3+ cells/kg. No patient developed acute GVHD. None of the three patients receiving the highest dose of T cells had any invasive fungal or viral infections. Monitoring of sorted host T cell subsets before TLI and ATG, and immediately after but before the infusion of donor cells, revealed in five of five patients a highly significant skewing of residual host T cells favoring invariant NK T (CD3+ CD161hi Va24 +Vb11 +) cells. The mean absolute number of host CD3+, and CD4+ and CD8+ T cells decreased by 99, 163 and 121 fold, respectively, immediately after conditioning compared to the absolute numbers before the start of TLI and ATG, whereas the mean absolute number of invariant NK T cells decreased by only 11%. In conclusion, we have determined the conditions for successful hematopoietic cell engraftment using a non-myeloablative regimen of TLI and ATG that appears associated with a reduced aGVHD risk yet retained graft anti-tumor activity. As in the pre-clinical model, we show direct evidence that the low incidence of aGVHD is associated with a significant alteration in residual host T cell subsets markedly favoring invariant NK T cells.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3662-3662
Author(s):  
Robert M. Dean ◽  
Daniel H. Fowler ◽  
Nancy M. Hardy ◽  
Jeanne Odom ◽  
Kathleen Castro ◽  
...  

Abstract Allogeneic hematopoietic stem cells (HSC) generally engraft rapidly and completely after myeloablative conditioning. However, with reduced-intensity conditioning (RIC), mixed chimerism and graft failure are more common. Host immune status and HSC number are factors known to affect engraftment after reduced-intensity stem cell transplantation (RIST). In addition, donor T cells within the allograft may also influencethe kinetics of donor engraftment after RIST. To evaluate this, we performed a controlled comparison of engraftment outcomes among 3 groups undergoing RIST, varying by ex vivo T cell depletion (TCD) or in vivo depletion of activated T cells with methotrexate (MTX) to prevent graft-versus-host disease (GVHD). Group I (n = 50) received T cell replete (TCR) peripheral blood stem cells (PBSC) with cyclosporine (CSA) alone for GVHD prophylaxis. Group II (n = 17) received ex vivo TCD PBSC (positive/negative selection with T cell add-back to uniform dose of 1 x 105 CD3+ cells/kg) with CSA alone for GVHD prophylaxis. Group III (n = 31) received TCR PBSC with CSA plus MTX (5 mg/m2 IV x 4 doses) for GVHD prophylaxis. The 3 groups were similarly immunosuppressed from prior therapy before RIST (median absolute lymphocyte counts 330/μL, 260/μL, and 307/μL for Groups I, II, and III, respectively), and received an identical RIC regimen (fludarabine/cyclophosphamide) plus comparable numbers of filgrastim-mobilized PBSC from HLA-matched sibling donors (median 7.9 x 106, 7.6 x 106, and 6.8 x 106 CD34+ cells/kg, respectively; median 3.6 x 108, 1.0 x 105, and 3.2 x 108 CD3+ cells/kg, respectively). Hematopoietic recovery was slowest in Group III, consistent with the myelosuppressive effects of MTX (Table). A greater proportion of patients in Group I achieved complete donor chimerism (≥ 95%) by day +28 than in Groups II or III (P &lt; 0.025), and at day +100, mixed donor chimerism persisted more often in Groups II and III than in Group I patients (P &lt; 0.01). Correspondingly, early (&lt; day +42) occurrence of grade 3–4 acute GVHD, before initiation of planned sequential donor lymphocyte infusions (DLI) in Group II, was more frequent in Group I than in either Groups II or III (p=0.08). Table: Hematopoietic Recovery, Engraftment, and GVHD Group Days to ANC &gt; 500, median (range) Days to plt &gt; 100, median (range) Donor chimerism ≥ 95% Early acute GVHD, grades 3–4 Day +28 Day +100 I 9 (7–13) 15.5 (12-42) 37/44 (84%) 36/38 (95%) 9/50 (18%) II 9 (7–10) 17.5 (11–40) 8/17 (47%) 9/14 (65%) 0/17 (0%) III 14 (7–21) 21.5 (12–85) 23/31 (74%) 21/31 (68%) 2/31 (6%) Thus, the deletion of T cells by either ex vivo TCD or in vivo MTX administration measurably alters the kinetics and degree of donor T cell engraftment after RIST. These observations provide evidence that donor T cells are an independent factor affecting engraftment of allogeneic HSC after RIST by compensating for incomplete host immune ablation. These data also support the hypothesis that a graft-versus-host effect plays a significant role in engraftment after RIST. Manipulation of donor T cells through graft engineering techniques may be a useful strategy to enhance engraftment in the setting of RIST.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 769-769 ◽  
Author(s):  
Aaron P. Rapoport ◽  
Stephan A. Grupp ◽  
Edward A. Stadtmauer ◽  
Robert H. Vonderheide ◽  
Bruce L. Levine ◽  
...  

Abstract Retrospective studies suggest that rapid lymphocyte recovery following autologous stem cell transplants (SCT) may be associated with better outcomes. Previously we showed that adoptive transfer of in-vivo vaccine-primed and ex-vivo (anti-CD3/anti-CD28) costimulated autologous T cells (ex-T) at about day 14 post-transplant increased CD4 and CD8 T cell counts at day 42 post-transplant and induced pneumococcal conjugate vaccine-directed T and B-cell responses [Rapoport et al, Nature Medicine, 2005]. In 2 current studies, we are further investigating the impact of ex-vivo costimulated autologous T cells on vaccine responses after SCT. In the first study, we are investigating whether a similar strategy of pre- and post-transplant immunizations along with an early infusion of vaccine-primed ex-T can induce responses to a putative tumor vaccine composed of 4 HLA-A2-restricted peptides derived from survivin and hTERT in pts undergoing SCT for myeloma. In the second (randomized) trial, the impact of early ex-T on immune recovery and vaccine reponses is being tested in pediatric neuroblastoma pts. Compared to the previous study, two methodologic changes were made: The target number of T cells infused was raised 5-fold to 5 x 1010 (109/kg) T cells were infused on day + 2 to take greater advantage of homeostatic expansion mechanisms. Patients were monitored for delayed hematopoietic recovery because of this switch to early ex-T and the fact that survivin and hTERT are also expressed in hematopoietic stem cells. At the time of submission, 16 adult and 30 pediatric patients have been enrolled on these trials of whom 11 and 21, respectively, are evaluable for post-transplant hematopoietic and T-cell recovery. On the myeloma trial, the mean # of T cells infused was 3.95 x 1010 with 96% viability and a CD4/CD8 ratio of 1.8:1. At day 14 post-transplant, the median CD4 count was 1951/mcl (range 651–7668) and the median CD8 count was 4117/mcl (range 1499–39,354). The median # days to achieve an absolute neutrophil count (ANC) > 500 was 12 (range 11–14) and the median # days to achieve a PLT count >20,000/mcl was 13 days (range 0–28). Similarly, in the pediatric cohort, median CD4 and CD8 counts at day 30 were 1500 and 2100/mcl, respectively, compared to 22 and 14 in a group of pts who did not receive d+2 ex-T, with no impact on engraftment. 1 adult and 3 pediatric pts also developed an “engraftment syndrome” characterized by GHVD-like features with or without fever. The adult pt with day 14 CD4 and CD8 counts of 2,724 and 11,571 cells/mcl had clinical and histologic features of (autologous) gut GVHD. 3 pediatric pts developed pruritic rashes clinically and pathologically indistiguishable from GVHD within 14 d of ex-T infusion, with fever seen in 1. In the adult and 1 pediatric pt, steroid treatment led to complete resolution of symptoms. These combined data sets demonstrate that robust CD4 and CD8 T cells counts can be achieved as early as day 14 post-SCT when adults or children receive ex-T at day +2 post-SCT without exogenous IL-2 or other cytokine support. It appears that a subset of patients develop a T cell “engraftment syndrome” similar to autologous GVHD. The mechanisms responsible for this rapid immune cell recovery are currently under investigation.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3054-3054 ◽  
Author(s):  
Rachel B. Salit ◽  
Frances T. Hakim ◽  
Michael R. Bishop ◽  
Thea M. Friedman ◽  
Robert Korngold ◽  
...  

Abstract Abstract 3054 Background: A clearly superior graft-versus-host disease (GVHD) prophylaxis regimen has not been established for patients undergoing reduced intensity allogeneic hematopoetic stem cell transplantation (HSCT) from matched unrelated donors (URD). Encouraging results have been reported with both the combination of alemtuzumab and cyclosporine (AC) and the regimen of tacrolimus, methotrexate, and sirolimus (TMS) in the URD setting. These two regimens work by biologically distinct mechanisms and may have markedly different effects on immune reconstitution. T-cell receptor (TCR) spectratyping analysis, which provides information on antigen receptor diversity, is a valuable method for monitoring post-transplant immune reconstitution. As part of a randomized pilot study, we prospectively assessed the effects of AC vs. TMS on TCR Vb repertoire diversity in patients undergoing reduced intensity HLA-matched unrelated donor transplantation. Methods: Twenty patients (median age 53 yrs; range 24–70 yrs) with hematologic malignancies received reduced intensity conditioning (fludarabine 30 mg/m2/day and cyclophosphamide 1200 mg/m2/day IV Day -6 to -3) followed by a 10/10 HLA-matched unrelated donor T-cell replete mobilized peripheral blood allograft. Patients were randomized to receive either: AC (n=10): alemtuzumab 20 mg/day IV over 8 hours Days -8 to -4 and cyclosporine starting at Day -1 with a 10% per week taper starting at Day +100 or TMS (n=10): tacrolimus and sirolimus starting at Day -3 with a 33% taper at Day +63 and Day +119 and methotrexate 5 mg/m2 IV, Days +1, +3, +6, and +11. Blood samples were collected from the donor and patient at baseline and the patient at 1, 3, 6 and 12 months post-transplant for TCR spectratyping analysis. All comparisons are based on an exact Wilcoxon rank sum test; p values < 0.01 were significant because of multiple comparisons. Results: Patients on the AC arm had significantly fewer T-cells on Day +14 compared with the TMS arm (median CD3+ = 1 cells/μl vs 356 cells/μl; CD4+ = 0 cells/μl vs 243 cells/μl; CD8+ = 0 cells/μl vs. 59 cells/μl; each p<0.0001); there was less disparity at Day +28 (median CD3+ = 45 cells/μl vs. 398 cells/μl; CD4+ = 36 cells/μl vs. 218 cells/μl; CD8+= 5 cells/μl vs 152 cells/μl; each p 0.002). By Day +100, lymphocyte recovery was not appreciably different between the two arms (median CD3+ = 242 cells/μl vs. 445 cells/μl (p = 0.095): CD4+ = 106 cells/μl vs. 212 cells/μl (p=0.28); CD8+ = 72 cells/μl vs. 135 cells/μl (p = 0.03). NK-cell recovery was slightly less in the AC vs. TMS arm at Day +14 (median NK = 27 cells/μl vs. 70 cells/μl; p = 0.01) and at Day +28 (median NK = 29 cells/μl vs. 150 cells/μl; p=0.02). There was no difference by Day +100 (median NK = 124 cells/μl vs. 88 cells/μl; p=0.31). B-cell reconstitution was negligible in both arms through Day +100. Assessment of CD4+ TCR Vb repertoire diversity by spectratyping demonstrated significantly lower diversity in patients receiving AC at 1 (p = 0.0003), 3 (p = 0.0003) and 6 (p=0.003) months post transplant compared with patients receiving TMS. CD8+ TCR spectratyping similarly revealed significantly reduced diversity in the AC arm at 3 (p = 0.001) and at 6 months (p = 0.003), and a trend toward significance at 12 months (p = 0.07). On each of the 2 arms, 2 of 10 patients developed acute Grade II-IV GVHD. Of the 5 patients on the AC arm who were seropositive for CMV, all 5 reactivated CMV by PCR within the first 60 days and reactivated 2–5 times in the first year. In contrast, only 3 of 5 seropositive patients reactivated CMV on the TMS arm and only one reactivated in the first 60 days. Conclusions: Two factors may have contributed to the loss of repertoire diversity in the AC arm. First, the alemtuzumab regimen may have severely depleted the infused donor T-cells. Second, stimulation by reactivating virus may have induced expansion of CMV-specific memory and effector T-cells, resulting in a skewed and oligoclonal T-cell repertoire. Especially in CD8+ T-cells, CMV has been shown to produce significant oligoclonal expansion (including CD4+: CD8+ ratio inversion). The loss of T-cell numbers and repertoire may in turn have contributed to the prevalence of early CMV reactivation. Thus, despite the similarities in frequency of acute GVHD in this small sample, it appears that these two commonly used GVHD prophylaxis regimens have very different effects on post-transplant immune reconstitution in the first 6 months after allogeneic HSCT. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1101-1101
Author(s):  
Mohammad Sohrab Hossain ◽  
Ghada M Kunter ◽  
Vicky Fayez Najjar ◽  
David L. Jaye ◽  
Edmund K. Waller

Abstract Donor T-lymphocytes are effective adoptive immunotherapy in the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT), but life threatening complications related to GVHD limits its clinical application. Recent advancement in the field of immunotherapy has directed our interest to enhancing the anti-tumor response of donor T cells by modulating expression of checkpoint blockade molecules including programmed death-1 (PD-1), cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) and foxp3, the transcription factor associated with regulatory T cells. The two ligands of PD-1, PD-L1 or PD-L2 are highly expressed in the presence of inflammatory signal induced by infection or cancer and PD-1/PD-L1 interaction negatively regulates T-cell antigen receptor (TCR) signaling and dampen T cell cytotoxic activity. Herein, we studied the role of PD-1, CTLA-4 and transcription factor foxp3 expressing donor CD4+ and CD8+ T cells in the development of GVHD. Methods: We have used two established allo-HSCT murine GvHD models. Lethally irradiated wild type (WT) B6, PD-L1 knock out (KO) B6 and PD-L2 KO B6 mice were transplanted with 2 x 106 splenic T cells and 2 x 106 T cell depleted bone marrow (TCD BM) cells from H-2Kdonors. Lethally irradiated CB6F1 recipients were similarly transplanted with splenocytes and TCD BM cells from B6 donors. Acute GvHD scores were determined by combining scores obtained from histological tissue sections and weight-loss, posture, activity, fur texture and skin integrity following standard published procedures. The activation status of donor T-cells and BM and host-derived non-T cells in GvHD target organs was analyzed by flow cytometry. Data from allo-HSCT recipients were compared with the respective data obtained from B6 à B6 syngenic HSCT (syn-HSCT) recipients. Serum cytokines were determined by Luminex assay. Results: PD-L1 KO B6 allo-HSCT recipients had significantly increased acute GvHD scores compared with WT B6 allo-HSCT recipients (p<0.0005) and B6 PD-L2 KO allo-HSCT recipients (p<0.0005) measured on day 8 after transplant. All PD-L1 KO allo-HSCT recipients died within 10 days post transplant while WT B6 and PD-L2 KO allo-HSCT recipients had 20% mortality until 36 days post transplant. Increased acute GvHD was associated with increased amount of serum inflammatory cytokines and increased numbers of activated PD-1+CD69+CD4+ donor T cells. Interestingly, PD-1 expression on donor CD4+ T cells significantly increased in the spleen of transplant recipients but not in BM, while PD-1 expression was significantly increased on donor CD8+ T cells in both spleen and BM compartments of allo-HSCT recipients compared with the syn-HSCT recipients. CTLA-4 expression on CD4+ and CD8+ donor T cells were significantly increased in spleen in the first two weeks post transplant but decreased at later time points compared with syn-HSCT. Again, CTLA-4 expression on CD4+ donor T cells in the BM remained significantly higher measured on 100+ days post transplant in allo-HSCT recipients compared with the syn-HSCT but similar levels of CTLA-4 expression on CD8+ T cells were measured in BM between these two HSCT recipients. Foxp3 expression on donor T cells and the numbers of CD4+CD25+foxp3+ regulatory T (Tregs) were markedly suppressed in donor T cells on day 4 post HSCT of allo-HSCT recipients compared with the syn-HSCT recipients. Although total numbers of donor T cells in the spleen of allo-HSCT recipients remained low over time, the percentage of PD-L1-expressing donor T cells in spleen were significantly higher (p<0.005) at early time points (day 4) in allo-HSCT recipients compared with the syn-HSCT. While total numbers of host-derived cells in spleen decreased over time in mice that developed GvHD, host-derived PD-L1 expressing CD3+ T cells persisted at higher levels through day 36 post transplant. Additionally, PD-L1 expression was also increased in donor BM-derived T cells and non-T cells populations over time. Collectively, these data indicate that severe GvHD occurs in allo-HSCT recipients in spite of increased numbers of PD-1, CTLA-4 and PD-L1 expressing donor and host cells. The occurrence of severe GvHD in these allo-HSCT models systems was associated with markedly reduced levels of CTLA-4 and foxp3 transcription factor expressing Tregs indicating that these pathways may be more relevant to controlling GvHD than PD-1:PD-L1 expression. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3920-3920
Author(s):  
A. Samer Al-Homsi ◽  
Tara S Roy ◽  
Kelli Cole ◽  
Marlee Bogema ◽  
Stephanie F Williams ◽  
...  

Abstract Graft versus host disease (GvHD) remains a major barrier to the progress of blood and marrow transplantation and limits its wide applicability. Standard prophylactic regimens essentially targeting T lymphocytes are partially effective and burdensome. Cyclophosphamide (Cy) administered post-transplant selectively deletes alloreactive proliferating T cells, promotes expansion of regulatory T cells, and induces long-lasting depletion of intrathymic host-reactive T cells. It is an attractive option for prevention of GvHD and has already been used alone in matched related and unrelated donor transplants. However, despite a low incidence of chronic GvHD, acute GvHD still occurs in 50% of cases and is grade III-IV in 15% of cases. Dendritic cells (DCs) play a pivotal role in the early phase of GvHD. Proteasome inhibitors such as bortezomib (Bor) have a number of immunomodulatory effects including inhibition of DCs maturation and function. We therefore initiated a phase I feasibility study combining post-transplant Cy & Bor. Twelve patients with hematological malignancies undergoing peripheral blood allogeneic transplantation from matched related (n=6) or unrelated (n=6) donors have so far been enrolled. Disease risk index (DRI) was low in 4, intermediate in 3 and high or very high in 5. The conditioning regimen combined fludarabine and busulfan (total 6.4 mg/kg). Patients receiving graft from unrelated donors also received rabbit anti-thymocyte globulin at 5-8 mg/kg. The dose of Bor was escalated in standard fashion. Three patients in each of cohorts 1 and 2 received 0.7 and 1 mg/m2 respectively. The subsequent 6 patients received 1.3 mg/m2. All patients received 2 IV doses, 6 hours after graft infusion and 72 hours thereafter. Cy was given at 50 mg/kg IV on days +3 and +4. Steroids were not allowed after day 0. Engraftment was prompt in all patients. Median time to neutrophil engraftment was 15.5 days (range 14-25). One patient failed to meet criteria for platelet engraftment. The patient had acyclovir-resistant herpes genitalis and CMV reactivation requiring protracted therapy with foscarnet. The remaining patients had a median time to platelet recovery of 28 days (range 15-109). All patients achieved full chimerism by day 20 except one who had residual CLL and did not reach full chimerism until day +119. No patient developed secondary graft failure. Two treatment-related deaths occurred on day +150 due to RSV pneumonitis and on day +200 due to acute sepsis. One patient with recurrent multiple myeloma after autologous transplantation died due to progressive disease. No other Common Toxicity Criteria grade 3 or 4 occurred in any patient. With a median follow-up of 21 months (range 1-27), the overall 2-year predicted disease free survival and overall survival were both 60%. Incidence of acute GvHD in 11 patients with follow-up > 100 days, was 64%: grade I 55%, grade II 9%, and grade III-IV 0%. GI and liver acute GvHD were not encountered. Only 4 patients received systemic steroids for acute GvHD; only one required > 20 mg/day of prednisone. One patient developed chronic GvHD of the liver (biopsy-proven). Another patient developed poor appetite and weight loss on day +138. Endoscopy showed gastric ulceration. No biopsy was obtained. Neither calcineurin nor m-TOR inhibitors were ever used. Two patients developed extensive HSV-genito-rectal ulcers; one had prior history of recurrent flares. When institutional guidelines were changed to start acyclovir at the beginning of conditioning as opposed to day +5, no other cases was noted. Seven patients developed CMV reactivation and required preemptive therapy only. One patient developed BK virus-induced hematuria and 1 patient developed CNS toxoplasmosis. In summary, the calcineurin and m-TOR inhibitor-free post-transplant Cy & Bor combination for GvHD prophylaxis is feasible and safe. Although the small number of patients prevents any definite conclusion, the absence of incidence of grade III-IV acute GvHD and the sparing of the GI tract and liver are promising. Furthermore, the completion of GvHD prophylaxis by day +4 without the need for close renal and drug level monitoring are both practical and appealing. Updated results with longer follow-up will be reported at the meeting. A confirmatory phase II study is underway. Disclosures Al-Homsi: Millennium Pharmaceuticals: Research Funding. Off Label Use: Bortezomib use for aGvHD prevention.


Sign in / Sign up

Export Citation Format

Share Document