Adoptive Immunotherapy with Tregs and Tcons Ensures Low TRM and a Low Incidence of Post Transplant Leukaemia Relapse After HLA Haploidentical Transplants for Acute Leukemia

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 154-154
Author(s):  
Mauro Di Ianni ◽  
Franca Falzetti ◽  
Alessandra Carotti ◽  
Adelmo Terenzi ◽  
Loredana Ruggeri ◽  
...  

Abstract Abstract 154 In full haplotype mismatched (HLA-haploidentical) stem cell transplantation we showed adoptive transfer of freshly isolated donor CD4+CD25+ FoxP3+ T regulatory cells (Tregs) followed by donor T cells (Tcons) prevented acute and chronic GvHD without any post-transplant immunosuppression, promoted lymphoid reconstitution and improved immunity against opportunistic pathogens (Di Ianni et al., Blood 2011). The major drawback was the extra-haematological toxicity of the conditioning regimen which included TBI, thiotepa, fludarabine and cyclophosphamide. To reduce regimen related toxicity we replaced cyclophosphamide with alemtuzumab, given 22 days before the Treg infusion to prevent it from interfering with adoptive T cell immunotherapy (Fig 1). The graft consisted of immunoselected Tregs (median 2×106/kg; range 1.6–4.8; FoxP3+ cells 92% ± 8 SD;), CD34+ cells (median 9.1×106/kg; range 8.1–10.9) and Tcons (median 1×106/kg; range 0.5–3). No post-transplant prophylaxis against GvHD was given. Since May 2010 18 patients (median age 43 years, range 23–61) with high risk acute leukaemia (16 AML, 2 ALL) have been transplanted. All sustained full donor-type engraftment. Neutrophils reached 0.5×109/L at a median of 12 days (range 9–28 days). Platelets reached 20×109/L and 50×109/L at median of 12 and 15 days, respectively (range 10–36 days and 11–55 days). CD4+ and CD8+ peripheral blood counts reached, respectively, 50/μL medianly on days 36 (range 27 – 120 days) and 34 (range 15– 85); 100/μL medianly on days 55 (range 27 – 147 days) and 48 (range 27 – 114); 200/μL on days 62 (range 37 – 177 days) and 49 (range 28 – 147). We observed a rapid development of a wide T-cell repertoire with specific CD4+ and CD8+ T cells for opportunistic pathogen antigen such as Aspergillus, Candida, CMV, ADV, HSV, VZV, Toxoplasma. Treg immunotherapy did not compromise post-transplant generation of donor-vs-recipient alloreactive natural killer (NK) cell repertoires in patients who received transplants from NK alloreactive donors (Ruggeri et al., Science 2002). Three of 16 valuable patients developed acute GvHD. Two responded to a short course of immunosuppressive therapy and at present (288 and 360 days after transplant) are alive and well with very good immunological reconstitution. The 3rd patient died of infectious complications. Two other patients died of non-leukemic causes (1 fulminant hepatitis 17 days post-transplant, 1 pneumonia 14 days post-transplant). The incidence of TRM is 17% (3/18). As hoped, extra-haematological toxicity was mild. One AML patient, who received a transplant from a non-NK alloreactive donor, relapsed 77 days post-transplant. Fourteen of the 18 patients are alive and well at a minimum follow-up of 3 months. This study shows adoptive immunotherapy with freshly isolated, naturally occurring Tregs is a feasible option in HLA-haploidentical stem cell transplantation since alloantigen-specific Tregs were efficiently activated in vivo and controlled alloreactivity of at least 1×106/kg Tcons without clinically significant inhibition of general immunity. Moreover Treg infusion did not weaken the GvL effect. The incidence of post-transplant leukaemia relapse was surprisingly low as only 1 patient has relapsed to date and even in our previous series no patient who was transplanted in CR has relapsed at a median follow-up of 25 months. Infusion of high numbers of Tcons in the absence of post-transplant immunosuppression can be hypothesized to exert a GvL effect. In addition, in patients who were transplanted from NK alloreactive donors, preservation of alloreactive NK cell repertoires played a key role in reducing the incidence of relapse. Disclosures: No relevant conflicts of interest to declare.

Hematology ◽  
2003 ◽  
Vol 2003 (1) ◽  
pp. 350-371 ◽  
Author(s):  
A. John Barrett ◽  
Katayoun Rezvani ◽  
Scott Solomon ◽  
Anne M. Dickinson ◽  
Xiao N. Wang ◽  
...  

Abstract After allogeneic stem cell transplantation, the establishment of the donor’s immune system in an antigenically distinct recipient confers a therapeutic graft-versus-malignancy effect, but also causes graft-versus-host disease (GVHD) and protracted immune dysfunction. In the last decade, a molecular-level description of alloimmune interactions and the process of immune recovery leading to tolerance has emerged. Here, new developments in understanding alloresponses, genetic factors that modify them, and strategies to control immune reconstitution are described. In Section I, Dr. John Barrett and colleagues describe the cellular and molecular basis of the alloresponse and the mechanisms underlying the three major outcomes of engraftment, GVHD and the graft-versus-leukemia (GVL) effect. Increasing knowledge of leukemia-restricted antigens suggests ways to separate GVHD and GVL. Recent findings highlight a central role of hematopoietic-derived antigen-presenting cells in the initiation of GVHD and distinct properties of natural killer (NK) cell alloreactivity in engraftment and GVL that are of therapeutic importance. Finally, a detailed map of cellular immune recovery post-transplant is emerging which highlights the importance of post-thymic lymphocytes in determining outcome in the critical first few months following stem cell transplantation. Factors that modify immune reconstitution include immunosuppression, GVHD, the cytokine milieu and poorly-defined homeostatic mechanisms which encourage irregular T cell expansions driven by immunodominant T cell–antigen interactions. In Section II, Prof. Anne Dickinson and colleagues describe genetic polymorphisms outside the human leukocyte antigen (HLA) system that determine the nature of immune reconstitution after allogeneic stem cell transplantation (SCT) and thereby affect transplant outcomethrough GVHD, GVL, and transplant-related mortality. Polymorphisms in cytokine gene promotors and other less characterized genes affect the cytokine milieu of the recipient and the immune reactivity of the donor. Some cytokine gene polymorphisms are significantly associated with transplant outcome. Other non-HLA genes strongly affecting alloresponses code for minor histocompatibility antigens (mHA). Differences between donor and recipient mHA cause GVHD or GVL reactions or graft rejection. Both cytokine gene polymorphisms (CGP) and mHA differences resulting on donor-recipient incompatibilities can be jointly assessed in the skin explant assay as a functional way to select the most suitable donor or the best transplant approach for the recipient. In Section III, Dr. Nelson Chao describes non-pharmaceutical techniques to control immune reconstitution post-transplant. T cells stimulated by host alloantigens can be distinguished from resting T cells by the expression of a variety of activation markers (IL-2 receptor, FAS, CD69, CD71) and by an increased photosensitivity to rhodamine dyes. These differences form the basis for eliminating GVHD-reactive T cells in vitro while conserving GVL and anti-viral immunity. Other attempts to control immune reactions post-transplant include the insertion of suicide genes into the transplanted T cells for effective termination of GVHD reactions, the removal of CD62 ligand expressing cells, and the modulation of T cell reactivity by favoring Th2, Tc2 lymphocyte subset expansion. These technologies could eliminate GVHD while preserving T cell responses to leukemia and reactivating viruses.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4475-4475
Author(s):  
Jessica C. Harskamp ◽  
Esther H.M. van Egmond ◽  
Hans L. Vos ◽  
Stijn J.M. Halkes ◽  
Roel Willemze ◽  
...  

Abstract Abstract 4475 Allogeneic stem cell transplantation (alloSCT) is frequently complicated by life-threatening graft versus host disease (GVHD). Previous studies demonstrated that T cell depletion (TCD) of the graft significantly decreases the incidence and severity of GVHD, and is associated with a higher percentage of patients with mixed chimerism (MC). In most studies chimerism analysis is performed on the total bone marrow (BM) leukocyte fraction, and changes in chimerism are related to engraftment. In this study we investigated whether MC in the total BM leukocyte fraction truly reflects engraftment or if it is influenced by survival and expansion of donor and recipient residual mature T cells, and whether hematopoietic lineage specific chimerism analysis is therefore a better method to determine engraftment. It is likely that chimerism analysis of the stem cell compartment is best reflected in peripheral blood (PB) in those cells that are continuously produced and short lived, such as monocytes and granulocytes, and therefore PB myeloid chimerism primarily reflects engraftment. In contrast, previous studies have shown by T cell receptor excision circle analysis that T cell neogenesis is virtually absent in the first 6 months after alloSCT, and that predominantly memory T cells are present in PB and BM. Therefore, we hypothesize that MC of these long lived T cells merely reflects survival and expansion of recipient and donor residual T cells. Since the life span of B and NK cells is longer than myeloid cells, but shorter than T cells, we anticipate that in the first 6 months after alloSCT, B and NK cell chimerism reflects a combination of survival and neogenesis. To analyze these hypotheses we performed hematopoietic lineage specific chimerism analysis on PB cells of 22 patients (median age 52 years, range 23-73, 11 males) receiving a TCD alloSCT between June and November 2008 after a myeloablative (n=11) or non myeloablative conditioning regimen (n=11) for AML, ALL, high risk MDS, multiple myeloma, CML, CLL or NHL. At intervals of 6 weeks PB was collected, and monocytes, granulocytes, B and NK cells, CD4+ and CD8+ T cells were sorted. The total leukocyte fraction was obtained by erythrocyte lysis of BM. DNA was isolated to perform chimerism analysis using short tandem repeats - PCR. Our results show that in the BM leukocyte fraction 47% of the patients were MC at 3 months after alloSCT, with a median frequency of patient cells of 4%. However, of the patients with MC in the total leukocyte fraction, 67% was complete chimeric in the myeloid subsets and MC in the T cell compartment. In the PB myeloid subsets (monocytes and granulocytes) less than 28% of the patients were MC during the first 6 months after alloSCT with a median frequency of patient cells less than 5%. In the B and NK cell subsets, at most time points more patients were MC (7-43%) with higher frequencies of patient cells (2-14%) compared to the myeloid subsets. The CD4 and CD8 T cell subsets showed the highest frequencies of MC in numbers of patients (31-61%) as well as the highest MC frequencies of patient cells (13-80%). Phenotypic analysis of the T cell compartment showed that 98% of the CD4 and CD8 T cells were memory cells during the first 6 months after alloSCT. Preliminary data indicate that the median percentage of donor derived T cells increased during the first 6 months after alloSCT, correlating with development of mild GVHD, suggesting that T cell chimerism is influenced by immunogenic triggers. In conclusion, these results illustrate that for engraftment and neogenesis of donor hematopoiesis, myeloid chimerism analysis provides more accurate information than total BM leukocyte chimerism analysis, since the results are greatly influenced by T cell chimerism. Since almost all T cells were memory cells within the first 6 months after alloSCT, T cell chimerism analysis reflects survival and expansion of mature donor as well as recipient T cells, and can therefore not be used to measure engraftment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3471-3471
Author(s):  
Farid Boulad ◽  
Stella M. Davies ◽  
David A. Williams ◽  
David A Margolis ◽  
Elizabeth G Klein ◽  
...  

Abstract Abstract 3471 Fanconi anemia (FA) is associated with a high risk of secondary malignancies, more specifically solid tumors. In allogeneic stem cell transplantation of the “non-Fanconi anemia host”, two factors have been consistently associated with increased risk of secondary malignancies post transplant and include graft-versus-host disease (GvHD) and radiation. The goal of reducing the risk of GvHD post transplant, has been achieved successfully with T-cell depleted transplants, as previously reported. Our second aim was to reduce the risk of radiation toxicity post transplant. We therefore initiated a multi-center trial using the fludarabine (Flu)/cyclophosphamide (Cy)/ATG cytoreduction backbone, and substituting busulfan (Bu) instead of radiation. This was followed by a CD34+ T-cell depleted peripheral blood stem cell graft. From June 2009 to July 2012, 27 patients, including 13 males and 14 females aged 4.3 – 31.4 (median 8.1), enrolled on this phase II multicenter protocol in 4 institutions. Indications for transplant included severe single lineage cytopenia (N=3), aplastic anemia (N=18) and myelodysplastic syndrome (N=6) - including refractory anemia RA (N=3), RA with excess blasts 1 RAEB1 (N=2), RAEB-2 (N=1). Prior treatment included transfusions (N=27) and androgens (N=12). Ten patients had a history of prior infections. Donors were Related mismatched (N=6) or Unrelated matched (N=16) or mismatched (N=5). Preparative regimen was: Bu 0.8–1.0 mg/Kg/dose Q 12H × 4 doses (days -7, -6), Cy 10 mg/Kg/day × 4, Flu 35 mg/m2/day × 4 days and Rabbit ATG 2.5 mg/Kg/day × 4 days (days -4 to -2). Patients also received filgrastim and GvHD prophylaxis with cyclosporine. Busulfan doses were adjusted to keep the steady state concentration below 350 in most cases. All grafts were T-cell depleted using the CliniMacs CD34 columns (Miltenyi). Cell doses of the grafts were: 3.1–42.7 × 106̂ CD34 cells/Kg and 2.2–49.9 × 103̂ CD3 cells/Kg. All 27 evaluable patients engrafted; one patient suffered a secondary graft failure. Two patients developed grade 1 and only one patient developed grade 2 GvHD. Grade 2–4 toxicity included mucositis (N=10), pulmonary toxicity (N=6), hepatotoxicity (N=6) with one patient developing hepatic veno-occlusive disease, renal toxicity (N=5), and hypertension (N=7). Infections included: CMV viremia (N=4), EBV viremias (N=1), clostridium difficile (N=1), bacteremia (N=5) and clostridium botulinum (N=1). Cause of death for four patients was acute respiratory failure (N=1), multi-organ failure (N=1), severe pulmonary hypertension (N=1) and infection (N=1). With a median follow-up of 7.9 months (range 0.5–37.8 months) 19 of 23 evaluable patients are alive and well (4 pts too early for outcome analysis). Although the study is ongoing an includes a relatively small number of patients with modest follow-up, the results appear promising. Engraftment, GVHD, early toxicity, infection and outcome data appear similar to historical TBI-based protocols. Further enrollment and longer follow up will define the comparative toxicity profile and relative risk of second tumor. Disclosures: Williams: bluebird bio: Consultancy; Wyeth: ; Takara bio:.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2031-2031
Author(s):  
Simone A Minnie ◽  
David Smith ◽  
Kate H Gartlan ◽  
Thomas S Watkins ◽  
Kate A Markey ◽  
...  

Abstract Autologous stem cell transplantation (ASCT) remains an important consolidation treatment for multiple myeloma (MM) patients, even in the era of novel agents. The prolongation of plateau-phase induced by ASCT is generally attributed to intensive cytoreduction. However, ASCT generates inflammation and profound lymphodepletion, which may result in hitherto unexpected immunological effects. To investigate potential immunological contributions to myeloma control after ASCT, we developed preclinical models of transplantation for MM using Vk*MYC myeloma that generates bony lytic lesions, a serum M band and marrow plasmacytosis that are hallmarks of clinical disease. Myeloma-bearing B6 recipients underwent myeloablative conditioning and were transplanted with naïve B6 bone marrow (BM) grafts with or without T cells from donors that were myeloma-naïve (SCT) or had low M bands at the time of harvest to mimic ASCT. Surprisingly, we demonstrate the broad induction of T cell-dependent myeloma control with enhanced median survival in recipients of grafts containing T cells compared to T cell depleted (TCD) BM alone (SCT= 91 days and ASCT > 100 days post-transplant vs TCD BM alone= 44 days; p<0.0001). Myeloma was most efficiently controlled when recipients were transplanted with memory T cells (CD44+) from autologous grafts (median survival: ASCT-CD44+ T cells >90 days post-transplant vs. CD44─ T cells = 50 days; p = 0.0006). Importantly, T cells adoptively transferred from recipients surviving > 120 days (MM-primed) protected secondary recipients compared to T cells from naïve donors (median survival: MM-primed > 120 days post-transplant vs 65 days naïve T cells; p = 0.0003). Furthermore, MM-primed CD8 T cells were restricted in TCR repertoire and provided protection in a myeloma clone-specific fashion, indicative of a tumor-specific T cell response. Despite this immune-mediated control of myeloma after SCT, progression still occurred in the majority of recipients. We phenotyped CD8+ T cells from the BM of MM-relapsed, MM-controlled and MM-free (that had never seen myeloma) mice 8 weeks after SCT. Expression of the inhibitory receptors, programmed cell death protein 1 (PD-1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) on BM CD8+ T-cells strongly correlated with myeloma cell number (r = 0.729, p<0.0001 and r = 0.796, p<0.0001 respectively). Additionally, the co-stimulatory/adhesion receptor CD226 (DNAM-1) was markedly downregulated as myeloma progressed (r = - 0.865, p<0.0001), as was interferon-γ secretion (r = - 0.76, p = 0.0022). t-SNE analysis confirmed an irreversible exhaustion signature at myeloma progression, characterized by the absence of DNAM-1 and co-expression of PD-1, TIM-3, TIGIT together with CD101 and CD38. Immune-checkpoint inhibition (CPI) early post-SCT, using antibodies against PD-1 or TIGIT facilitated long-term myeloma control (median survival in both treatment arms > 120 days post-SCT vs. 60 and 68 days respectively; p <0.05). Furthermore, TIGIT blockade limited CD8+ T cell exhaustion, increased CD107a and IFNγ secretion and expanded a memory CD8+ T cell population in the BM. Genetic deletion of either IFNγ or the IFNγ receptor from the donor graft resulted in dramatic myeloma progression after SCT. Consequently, treatment with a CD137 (4-IBB) agonist early after SCT profoundly augmented CD8+IFNγ+GranzymeB+ T-cell expansion in the BM, such that majority of treated animals eliminated myeloma and survived long-term. These data provide insights into an unappreciated mechanism of action of ASCT whereby myeloma immune-equilibrium is established and suggest that combination with immunotherapeutic strategies is a rational approach to generate long term disease control. Disclosures Smyth: Bristol Myers Squibb: Other: Research agreement; Tizona Therapeutics: Research Funding.


2021 ◽  
Vol 11 ◽  
Author(s):  
Toshiki Terao ◽  
Ken-ichi Matsuoka ◽  
Kentaro Narita ◽  
Takafumi Tsushima ◽  
Satoshi Yuyama ◽  
...  

The prevention of chronic graft-versus-host disease (cGVHD) is important for recipients of hematopoietic stem-cell transplantation (HSCT). As one of the etiologies, the relationship between early T-cell recovery and subsequent cGVHD development has been the focus of attention. Recently, letermovir (LTV) was approved for preventing cytomegalovirus (CMV) reactivation in the early transplantation phase. Although CMV affects the immune reconstitution after HSCT, the impacts of LTV to prevent CMV reactivation on early T-cell recovery and cGVHD have not been fully investigated. We aimed to identify early T-cell recovery under LTV at day 30 in 15 and 33 recipients from matched related donors (MRDs) and haploidentical donors with post-transplant cyclophosphamide (PTCy-haplo), respectively. Early increases in the levels of total lymphocytes and HLA-DR+ activated T-cells at day 30 were observed under CMV prophylaxis by LTV only in PTCy-haplo recipients and not in MRD recipients. Moreover, PTCy-haplo recipients with LTV showed a significantly higher incidence of cGVHD, but not acute GVHD. Our observations suggest that an early increase in the levels of HLA-DR+ activated T-cells may be implicated in the development of cGVHD in patients treated with PTCy who received LTV. Further studies are warranted to validate our results and elucidate the detailed mechanisms of our new insights.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 191-191
Author(s):  
Karl S. Peggs ◽  
Stephanie Verfuerth ◽  
Christine Chow ◽  
Kirsty Thomson ◽  
Anthony H. Goldstone ◽  
...  

Abstract Adoptive transfer of virus-specific T-cells offers the potential for accelerating reconstitution of antigen-specific immunity and limiting the morbidity and mortality of viral infections following allogeneic stem cell transplantation. However, the logistics of producing virus-specific T-cells and the risk of inducing GvHD secondary to the infusion of alloreactive clones has limited the application of cellular therapies (CT). We have treated 37 allogeneic transplant recipients on 2 consecutive CT studies with CMV-specific T-cell lines generated by short-term ex-vivo culture of donor lymphocytes with donor monocyte-derived dendritic cells pulsed with virus-lysate. Culture resulted in generation of both CD4+ and CD8+ CMV-specific T cells as demonstrated by antigen-specific γ-interferon production (1.2–4.8% of CD4+ cells, 0.2–6.5% of CD8+ cells) and HLA-tetramer binding. These cells were capable of HLA-restricted CMV-specific target lysis. Thirty-one patients were treated on a pre-emptive protocol based on PCR surveillance, expanded to include those with multiple reactivations/disease. Total cultured cell dose infused was 1 x 105/kg, returned at a median of 39 (range 4–410) days post-transplant. Six have been entered on a subsequent prophylactic study, receiving the same dose at day 28 post-transplant. Five of these had CMV DNA detectable by PCR at the time of CT. The preparative regimen included T-cell depletion (alemtuzumab) in thirty-one. Ten had unrelated donors (5 mismatched at one and 1 at two HLA loci). Thirty-four recipients were CMV seropositive. Thirty-two were receiving cyclosporine at the time of infusion. One was also receiving mycophenolate, and one both mycophenolate and steroids for hemolysis. Eleven had prior GvHD (5 Grade I, 5 Grade II, 1 extensive chronic). Twenty-eight were treated during their first episode of post-transplant CMV infection, five during the second, two the third and one the seventh. Two had active CMV disease (biopsy proven CMV colitis) and six were receiving anti-viral drug therapy at the time of infusion. Fifteen patients required no antiviral therapy. In 16/30 cases given ACT at a time when CMV DNA was detectable antiviral therapy was given for subsequently increasing viral titre. Both cases of CMV disease resolved and there were no additional cases following CT. Following viral clearance there were only 4 episodes of subsequent CMV infection requiring therapy in 32 evaluable cases (including 1 treated prophylactically) compared to 45/72 historical controls (p<0.0001) Two occurred following increased immune suppression (including steroids) for ongoing or newly developed immune-mediated hemolysis. GvHD occurred in 12/32 evaluable patients following ACT (4 Grade I, 1 Grade III acute GvHD, 3 limited and 4 extensive chronic GvHD). Six cases were in unrelated donor or T-replete transplants, and six were in patients with a prior history of GvHD. Massive expansions of mainly CD8+ T-cells developed in concert with resolving viremia. Infused CMV-specific clones were demonstrated to expand (contributing up to 35% of the CD8+ population) and persist for at least 6 months following infusion. In conclusion, our updated experience demonstrates that cellular therapy for CMV is both feasible and effective in a clinical environment, and that it can be delivered with minimal toxicity allowing consideration of expanded prophylactic application.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2909-2909 ◽  
Author(s):  
Paul J. Travers ◽  
Andrea Knight ◽  
Sarah Grace ◽  
Panos Kottaridis ◽  
Stephen Mackinnon ◽  
...  

Abstract In previous studies we and others have shown that the production of new T cells from the thymus declines after the third decade and that while patients below the age of 30 reconstitute primarily new T cells, those over thirty reconstitute primarily by expansion of pre-existing mature T cells. However little attention has been paid to the γδ subset of T cells, which form an important component of mucosal immune protection and which represent approximately 5% of peripheral T cells. Two major subsets of γδ T cells are defined by the expression of Vδ1 versus Vδ2, with Vδ1+ cells predominating in the fetal circulation and in mucosal sites, while Vδ2+ cells predominate in adult life and in the peripheral circulation. In light of the differerential preponderance of the two subsets in the fetal versus adult circulation, we have examined the reconstitution of these two subsets of γδ T cells following hemopoietic stem cell transplantation in a cohort of 28 patients sampled at 3 monthly intervals to ask whether both subsets recover adequately from adult stem cells. In 44 normal individuals, the median levels of Vδ1 and Vδ2 cells are 12.46 (0.22 to 167.8) and 32.78 (4.48 to 190.1) cells/mm3 respectively. In patients under 30, the reconstitution of the Vδ1 and Vδ2 subsets follow similar kinetics, reaching a plateau at 9 months post transplant with comparable numbers of Vδ1 and Vδ2 cells (note that the normal ratio of Vδ1 to Vδ2 is 0.38, so in the patients there is a significant increase in the proportion of Vδ1 cells in the peripheral circulation). In patients over 30 years of age, there is an even more significant disparity in the reconstitution of the two subsets. The Vδ1 subset recovers with similar kinetics as is seen in the patients under 30, although to slightly lower final levels. The Vδ2 subset, however, shows very little recover y, reaching a plateau at 6 months at the bottom of the normal range for up to 2 years post transplant. In these patients the ratio between Vδ1 and Vδ2 is inverted with an increasingly greater proportion of Vδ1 cells at longer times after transplant, with ratios in excess of 10 by 24 months post transplant, a 30-fold increase in the normal proportion of Vδ1 + T cells. Given that the Vδ1 subset shows a very restricted repertoire compared to the Vδ2 subset, the overall capacity of the circulating γδ T cell population to recognise and respond to antigen will be significantly compromised in older transplant recipients.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2993-2993
Author(s):  
Bipin N. Savani ◽  
Stephan Mielke ◽  
Katayoun Rezvani ◽  
Agnes Yong ◽  
Nancy Hensel ◽  
...  

Abstract One hundred and fifty-seven patients with leukemia (80 CML, 48 AML/MDS, 29 ALL) received a T cell depleted myeloablative allogeneic stem cell transplantation (SCT) from an HLA-matched sibling between 09/1993–09/2005. Conditioning consisted of TBI (12–13.6 Gy) + cyclophosphamide (96) or cyclophosphamide and fludarabine (61). The stem cell source was G-CSF mobilized peripheral blood stem cell (PBSC) in 129 and bone marrow in 28 patients. T cell dose with graft ranged from 0.2 – 2 × 105/kg CD3+ cells. GVHD prophylaxis was with low dose cyclosporine (level 100–200 ng/ml) in 103 and standard dose in 54. Patients without ≥ grade II acute GVHD received 1–2 donor lymphocyte infusion of 107 CD3+ cells/kg between days 45 and 100. Absolute lymphocytes on day 30 (LC30) was available in 154 patients (3 patients died before day 30) and 54 day +30 post-transplant samples were available for lymphocyte subset analysis. Median lymphocyte count on day +30 (LC30) was 400/μl, (range 10–3295) and 150/μl, (range 6–1005) for CD56+, CD16+ CD3− NK cells (NK30). Statistical analysis was performed on SPSS14 software. Median age of the group was 34 years (range 10–56). 78 patients had standard risk (SR) disease in first remission or first chronic phase of CML. The remaining 79 had high risk (HR) disease. At the time of analysis 85 patients are (51.5±4%) are alive with median follow-up of surviving patients 1392 days (range 147–4208). Only 9 patients (3 above median LC30) developed aGVHD before day+30. Patients with ≥ median LC30 had significantly better transplant outcome: survival 71±5 vs. 36 ±5.5%, p<0.0001; DFS 71±5 vs. 31±5 %, p<0.0001; NRM 10±3.5 vs. 38 ±6%, p<0.0001; relapse 22±5 vs. 51±7.5%, p=0.004; ≥ II aGVHD 34 ±5 vs. 51±6%, p=0.05. In multivariate analysis only disease risk and LC30 emerged as independent prognostic factors: LC30 above 400/μl was associated with improved survival (RR 4.3), DFS (RR 4.5), less relapse (RR 10.3), NRM (RR 3.3) and aGVHD (RR 2.3). LC30 impacted on outcome of both HR and SR disease groups (Figure). LC30 and NK30 were highly correlated (r2– 0.45, p<0.0001) and NK30 above 150/μl was also associated with improved transplant outcome: In multivariate analysis of this subset of 54 patients, SR disease and NK30 emerged as the only independent factors with better outcome for NK30 >150/μl: higher survival (RR 3), and DFS (RR 3), less relapse (RR 4.8), less NRM (RR 3) and less aGVHD (RR 5.3). This study does not define whether LC30 is a surrogate for NK cell count or whether both are a surrogate for some other undetected prognostic factor. However the inverse relationship between NK count and aGVHD suggests an NK-mediated effect through elimination of host antigen presenting cells as has been described in mismatched SCT but also in HLA identical SCT by Cook et al (Blood2004, 103, 1521) Prospective studies to correlate transplant outcome with NK cell recovery and function after HLA identical SCT are indicated. Figure Figure


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 578-578
Author(s):  
David Avigan ◽  
Jacalyn Rosenblatt ◽  
Baldev Vasir ◽  
Zekui Wu ◽  
Adam Bissonnette ◽  
...  

Abstract Autologous transplantation results in the transient reversal of tumor mediated tolerance due to the reduction in disease bulk, the depletion of regulatory T cells, and in the increased presence of tumor reactive lymphocytes during the period of lymphopoietic reconstitution. As a result, cancer vaccines are being explored as a means of targeting residual myeloma cells following stem cell transplant. We have developed a cancer vaccine in which patient derived tumor cells are fused with autologous dendritic cells (DCs). In this way multiple tumor antigens are presented in the context of DC mediated costimulation. We are conducting a study in which patients with multiple myeloma (MM) undergo stem cell transplantation followed by vaccination with 3 doses of DC/MM fusions. DCs were generated from adherent mononuclear cells cultured with GM-CSF and IL-4 for 5–7 days and matured with TNFa. DCs strongly expressed costimulatory and maturation markers. Myeloma cells were isolated from bone marrow aspirates and were identified by their expression of CD38, CD138, and/or MUC1. DC and MM cells were fused with polyethylene glycol as previously described and fusion cells were quantified by determining the percentage of cells that coexpress unique DC and myeloma antigens. To date, 19 patients have been enrolled and 18 have completed vaccine generation. Mean yield of the DC and myeloma preparations was 1.84 × 108 and 8.3 × 107 cells, respectively. Mean fusion efficiency was 40% and the mean cell dose was 4.3 × 106 fusion cells. As a measure of their potency as antigen presenting cells, fusion cells prominently stimulated allogeneic T cell proliferation in vitro. Mean stimulation indexes were 12, 57, and 31 for T cells stimulated by myeloma cells, DCs, and fusion cells, respectively. Adverse events judged to be potentially vaccine related included injection site reactions, pruritis, myalgias, fever, chills, and tachycardia. Six patients have completed the follow up period and 3 patients are currently undergoing vaccination. All patients achieved a partial response to transplant. Three patients demonstrated resolution of post-transplant paraprotein levels following vaccination. One patient with highly aggressive disease who experienced disease progression in the early post-transplant period, demonstrated initial response and then stabilization of disease with vaccination. We are examining the effect of transplant and vaccination on measures of cellular immunity, anti-tumor immunity and levels or activated as compared to regulatory T cells. T cell response to PHA mitogen was transiently depressed post-transplant. In contrast, a transient increase was noted post-transplant in mean T cell expression of IFNγ in response to autologous myeloma cell lysate. In preliminary studies, a relative increase in the ratio of activated (CD4/CD25low) to regulatory (CD4/CD25high) T cells was observed. To date, all evaluable patients demonstrated evidence of vaccine stimulated anti-tumor immunity as manifested by a rise in IFNγ expression by CD4 and/or CD8+ T cells following ex vivo exposure to autologous tumor lysate. In this ongoing study, fusion cell vaccination in conjunction with stem cell transplantation has been well tolerated, induced anti-tumor immunity and clinical responses in patients with multiple myeloma.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4597-4597
Author(s):  
Lorea Beloki ◽  
Miriam Ciaurriz ◽  
Natalia Ramirez ◽  
Amaya Zabalza ◽  
Cristina Mansilla ◽  
...  

We present the results of a pilot study using pentamer (PM) and streptamer (ST) multimer complexes for monitoring CMV-specific CD8+ T-cells (CTLs). We analysed 15 patients that underwent allogeneic Stem Cell Transplantation (HSCT). Patient characteristics are summarized in Table 1. All patients and donors were positive for the HLA-A*02:01 allele. PM and ST were directed against the epitope NLVPMVATV (495-503) of the CMV phosphoprotein 65 (pp65). Samples were obtained at 15-day intervals until day +90 and monthly thereafter.PatientCMV status (D/R)GenderAgeDiagnosisDonorConditioningEngraftment (day)Follow up (months)GVHD (day)1+/+F43NHLSIBRIC2415-2-/+F33MDSURDRIC1912-3+/+M55AMLSIBMAC1714474-/+F41AMLURDMAC2121-5+/+F32AMLSIBMAC2013256+/+F64MDSSIBRIC2714897-/+M58CLLURDRIC2313928+/+M57ALLURDMAC155-9+/+F39AMLURDMAC2012-10-/+M42ALLURDMAC2125-11-/+M44AMLURDMAC12815312+/+F65AMLSIBRIC258-13-/+M27AMLSIBMAC3031814+/+M65AMLSIBRIC1966215+/-F30SAASIBRIC133- Three patterns were observed. In 3 patients (20%) no CMV-specific-CTLs could be detected despite several CMV reactivations, requiring prolonged cumulative antiviral therapy (median 68 days; range 67-136). In 7 patients (47%) CMV reactivation occurred at a mean of 41 days (10-94) and triggered a rapid increase of CMV-specific-CTLs with a median of 22.7 x 105/L (range 1.3-279.7). The CMV-PCR became immediately negative and antiviral therapy was stopped promptly after a median of 15.5 days (6-23). Finally, 5 patients (33%) showed an early immune reconstitution with CMV-specific-CTLs detected with a median of 0.7 x 105/L (range 0.2-2.8) in the absence of CMV-PCR reactivation at a median of 21 days (10-34) post-SCT. No CMV-PCR reactivation was observed in this group with a median follow-up of 12 months (5-14). Discussion Monitoring CMV-specific-T-cells might be able to distinguish patients at higher risk of recurrent virus reactivation and in need of prolonged antiviral therapy. Patients with increasing CMV-specific-CTLs detectable at the time of CMV-PCR reactivation may only need a short course of antiviral therapy, while those with early CMV-specific-CTLs may be protected from CMV reactivation. Conclusion Using Multimer-based (Pentamer and Streptamer) monitoring of CMV-specific T-cell immune reconstitution after allogeneic HSCT may contribute to the clinical decision regarding when and for how long to commence anti-CMV therapy. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document