In Chronic Myeloid Leukemia Cytotoxic T Cell Responses to BMI-1 Protein Correlate with Higher Expression of BMI-1 in Leukemia Progenitors, and May Contribute to Improved Outcome After Allogeneic Stem Cell Transplantation.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 192-192
Author(s):  
S. M. Agnes Yong ◽  
Nicole Stephens ◽  
Bipin N. Savani ◽  
Yixin Li ◽  
Rhoda Eniafe ◽  
...  

Abstract Abstract 192 The polycomb group (PcG) proteins BMI-1 and EZH2 are key regulators of self-renewal processes in normal and leukemic stem cells. Of these two PcG proteins, BMI-1 is more highly expressed in chronic myeloid leukemia (CML) than in normal stem cells, and is associated with a more rapid disease progression in patients who are treated with drug therapy alone, implying that an increased level of “stem-ness” conferred by BMI-1 contributes to leukemogenicity. Conversely, CML patients with high BMI-1 expression prior to allogeneic stem cell transplantation (SCT) have better overall survival post-transplant (Mohty, et al, Blood 2008). To investigate the potential of PcG proteins as leukemia-associated antigens, and targets for graft-versus-leukemia (GVL) effects, we studied a cohort of 86 CML patients (54 chronic phase, 32 advanced phase) who received T-cell depleted SCT with T-cell add-back on day 45-100 post-SCT from HLA-identical sibling donors. Using quantitative real-time PCR, we measured the expression of EZH2, BMI-1 and its target for repression, CDKN2A (encoding p16INK4A) in CD34+ progenitors and their CD34-negative counterparts. Using flowcytometric detection of intracellular cytokines IFN-γ or TNF-α, and degranulation marker CD107a, in CD8+ cytotoxic T lymphocytes (CTL), we assessed immune responses to BMI-1 (TLQDIVYKL and CLPSPSTPV) and EZH2 (YMCSFLFNL and SQADALKYV) peptides in 25 HLA-A*0201+ patient-donor pairs. Seven of 17 (41%) HLA-A*0201+ CML patients had native immune responses to BMI-1 peptide, which was associated with higher BMI-1 expression in CD34+ progenitors (p=0.04, Mann-Whitney U test). Five of 25 (20%) healthy HLA-A*0201+ sibling donors had detectable immune responses to BMI-1 peptide. EZH2 was less immunogenic compared to BMI-1 in both patients and donors. The majority of peptide-specific CTLs analyzed by peptide-specific dextramers had central memory phenotype. BMI-1- or EZH2-specific T cells were readily detected after 7-day cultures using an ELISPOT assay in 75% of donors or patients where peptide-specific CTLs were detected ex-vivo. A higher expression of BMI-1 in CML patients pre-SCT and correspondingly lower expression of its target for repression, CDKN2A, was associated with improved leukemia-free survival (p=0.01), and reduced disease-related death (p=0.0001). In four HLA-A*0201+ patients whose donors had immune responses to PcG peptides, BMI-1 or EZH2-specific T cell responses were detected in the first 120 days post-SCT. CML patients who had donors with immune responses to BMI-1 peptide had improved leukemia-free survival compared to patients whose donors were non-responders (80% vs. 60% respectively). Immune responses to PcG proteins, in particular to BMI-1, may be relevant for disease control by GVL effects. Unlike CTLs specific for primary granular proteins such as proteinase 3 and elastase, which are also highly expressed in CML cells, CTLs against BMI-1 and EZH2 may be less susceptible to selective deletion processes resulting in tolerance, as these proteins are less ubiquitously expressed by mature progenitors which are expanded in CML, and are therefore good GVL and immunotherapy target candidates in CML. Furthermore, these CTLs have the potential to target leukemia stem cells. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1993-1993 ◽  
Author(s):  
Eleanor Tyler ◽  
Achim A Jungbluth ◽  
Richard J. O'Reilly ◽  
Guenther Koehne

Abstract Abstract 1993 Wilm's tumor protein-1 (WT1) is over-expressed in a number of solid and hematologic malignancies including multiple myeloma (MM). The emergence of WT1-specific T cells has been shown to correlate with better relapse-free survival after allogeneic stem cell transplantation in patients (pts) with hematologic malignancies, such as leukemia. In MM, the expression of WT1 in the bone marrow has been shown to correlate with numerous negative prognostic factors, including disease stage and M protein ratio. Taken together, these findings suggest that immunotherapeutic augmentation of WT1-specific immune responses, such as adoptive transfer of WT1-specific T cells, may be capable of eradicating minimal residual disease and preventing relapse in MM. Thus, we examined the significance of WT1-specific cellular immune responses in pts with relapsed MM and high-risk cytogenetics who are undergoing allogeneic T cell-depleted hematopoietic stem cell transplantation (TCD HSCT). In this study, pts were eligible to receive low doses of donor lymphocyte infusions (DLI, 5×105-1×106 CD3+/kg) no earlier than 5 months post TCD HSCT. WT1-specific T-cell frequencies were measured in freshly isolated peripheral blood and bone marrow specimens. Frequencies were detected by staining for intracellular IFN-γ production in response to WT1 peptides, and/or by tetramer analysis, where available. Of 17 pts evaluated, all pts exhibited low frequencies of WT1-specific T-cell responses pre TCD HSCT. Ten of these pts received DLI post TCD HSCT. All 10 pts developed WT1-specific T cell responses post DLI. These increments in WT1-specific T-cell frequencies were associated with reduction in circulating myeloma proteins in all pts. Long-term evaluation demonstrated fluctuations in persisting WT1-specific T-cell frequencies following DLI. In one representative patient, a peak of 3.5% (72/ml) WT1-specific CD8+ T cells were detected in the peripheral blood by staining with the tetramer HLA-A*0201 RMF. This peak T-cell response occurred post TCD HSCT and DLI, and coincided with disease regression. This patient has remained in complete remission for more than 3 years post transplant, with fluctuating levels of WT1-specific CD8+ T cells ranging from 0.3–1.5% still persisting. Findings from concurrent molecular chimerism studies conducted on isolated T cells post TCD HSCT suggest that the WT1-specific T cells are of donor origin. Immunohistochemical analyses of WT1 and CD138 staining in MM bone marrow specimens demonstrated consistent co-expression within malignant plasma cells. WT1 expression in the bone marrow of all 6 pts tested correlated with the extent of malignant plasma cell infiltration. In contrast, no WT1 expression was observed when disease was low or absent. Taken together, our findings suggest a correlation between the emergence of WT1-specific T cells post DLI, and disease regression in pts being treated for relapsed MM. The present data support the development of adoptive immunotherapeutic approaches utilizing WT1-specific T cells for pts with MM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4136-4136
Author(s):  
Willemijn Hobo ◽  
Kelly Broen ◽  
Walter J.F.M. van der Velden ◽  
Annelies Greupink-Draaisma ◽  
Niken Adisty ◽  
...  

Abstract Abstract 4136 Purpose: Allogeneic stem cell transplantation (allo-SCT) can induce remission in patients with hematological malignancies due to graft-versus-tumor (GVT) responses. This, however, is often accompanied by graft-versus-host disease (GVHD). Both the GVT effect and GVHD are mediated by minor histocompatibility antigen (MiHA)-specific T cells recognizing peptide products from polymorphic genes that differ between recipient and donor. Here, we evaluated whether MiHA mismatches are associated with clinical outcome after partial T cell depleted allo-SCT. Patients and Methods: We retrospectively analyzed the impact of MiHA mismatches in a cohort of 327 patients who received a partially T cell-depleted allo-SCT because of a hematological malignancy. MiHA allele genotyping was performed by fluorescence-based competitive allele-specific PCR. Subsequently, a multivariable statistical analysis of immunogenic MiHA disparity rates and association with clinical outcome was performed. In addition, development of MiHA-specific T cell responses was assessed by dual-color tetramer staining. Results: Statistical analysis revealed that an autosomal MiHA disparity on DNA level associates with increased relapse-free survival in sibling transplants, especially in patients transplanted for multiple myeloma. In addition, mismatches for the ubiquitous Y chromosome-encoded MiHA resulted in more acute GVHD (grade 3–4), while other MiHA mismatches, either ubiquitous or restricted to hematopoietic cells, were not associated with GVHD. Finally, we demonstrated considerable differences between MiHA in the capability to induce in vivo T cell responses post-transplantation. Conclusion: These data support that autosomal MiHA contribute to the induction of GVT immunity providing a rationale for MiHA-based post-transplantation immunotherapy to prevent and treat persistent and recurrent cancer following allo-SCT. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2165-2165
Author(s):  
Xiangyu Zhao ◽  
Yingjun Chang ◽  
Ling-Ling Xu ◽  
Mingrui Huo ◽  
Xiaosu Zhao ◽  
...  

Abstract Introduction HLA-mismatched/haploidentical stem cell transplantation (SCT) is a feasible therapeutic option for advanced hematologic malignancies patients who lack an HLA-matched related or unrelated donor. The effect of NK alloreactivity in HLA haploidentcial SCT is still under debate and in particular in transplantation for chronic myeloid leukemia (CML) the data are very limited and with conflicting outcome.The goal of this study was to explorethe predictive roles of missing self model in our HLA-mismatched/haploidentical transplantation without T-cell-depletion in vitro in chronic myeloid leukemia patients, and to develop a simple algorithm on the basis of recipients and donor HLA-C and HLA-Bw4 gene content that can be used today to identify HLA-mismatched donors who will associated withbetter prognosis in T cell¨Creplete transplants. Methods We studied the HLA genotype of 78 donor-recipient pairs and the KIR genotype of their donor, who underwent unmanipulatedHLA-mismatched/haploidentical transplantation without T cells depletion in vitro during 2003-2009 in our center. To applythe missing ligand model, the first step was to divide ourdonor-recipient pairs into 2 groups according to the number of KIR ligand indonor and recipient, ie, 3 KIR ligands (“without missing self”) versus fewer than 3(“with missing self”). Meanwhile, to apply the KIR ligand-ligand model, donors who were classified as NK alloreactive against their recipientstermed KIR ligand mismatched donors throughout, possessedHLA class I KIRligand(s) which were missing in the recipients. Results Among the 78 pairs of donor-recipients, 65 and 13 recipients receivedHLA¨Cmismatched/haploidentical transplants from “with missing self (R-L mismatch)” and “without missing self (R-L match)” donors, respectively. Using Ligand-ligand model, 59 and 19 recipients received haploidentical transplantation from “KIR ligand matched (L-L match)” and “KIR ligand mismatched (L-L mismatch)” donors, respectively. In contrast to Perugia's KIR ligand-ligand mismatched model or Handgretinger's KIR missing self model between donor-recipient pairs, we found that the 10-year disease free survival(DFS) rate were higher in patients received transplantation from “without missing self (R-L match)” donorscompared with those from “with missing self (R-L mismatch)” (92.3±7.4% vs. 55.2%±6.2%, p=0.024, Figure1A) , especially in high risk CML patients (100% vs. 37.2%±8.6%, p=0.029, Figure1B). When combined the above missing self model and Ligand-ligand model together, patients were subgrouped as receiving graft from “without missing self and without KIR ligand mismatch (R-L match and L-L match)” (n=13), “with missing self and without KIR ligand mismatch (R-L mismatch and L-L match)” (n=47), and “with missing self and with KIR ligand mismatch (R-L mismatch and L-L mismatch)” (n=18), respectively. Cox regression model showed the 10-yearDFSwas best predicted by the combination of missing self model and Ligand-ligand modelbetween recipients and donors pairs (HR 2.205(1.113-4.368), p=0.023, Figure1C). Meanwhile, donor KIR 2DS5 positive associated with higher DFS post-transplantation (84.2±8.4% vs. 56.8±7.7%, p=0.045) in CML patients. However, donor KIR haplotype B have no effect on DFS and overall survival after allogeneic hematopoietic stem cell transplantation for multiple myeloma Conclusions These data indicate thatpoor prognosis after transplantation is associated with the missing self and KIR ligand mismatch in recipients and T cell alloreaction may play apredominant role in this model.Based on recipients and donor HLA-C and HLA-Bw4 gene content, it could befeasible to identify HLA-mismatched donors who will predict the better prognosis in CML patients post T cell-replete transplant. Disclosures: No relevant conflicts of interest to declare.


2008 ◽  
Vol 45 (13) ◽  
pp. 3631-3638 ◽  
Author(s):  
Arnold van der Meer ◽  
Nicolaas P.M. Schaap ◽  
Anton V.M.B. Schattenberg ◽  
Bram van Cranenbroek ◽  
Henk J. Tijssen ◽  
...  

Blood ◽  
2003 ◽  
Vol 101 (2) ◽  
pp. 469-472 ◽  
Author(s):  
Mehmet Uzunel ◽  
Jonas Mattsson ◽  
Mats Brune ◽  
Jan-Erik Johansson ◽  
Johan Aschan ◽  
...  

The kinetics of minimal residual disease (MRD) and chimerism were studied in 15 patients with chronic myeloid leukemia (CML) receiving nonmyeloablative stem cell transplantation (NST) and in 10 patients receiving conventional stem cell transplantation (CST). All NST patients showed T-cell mixed chimerism (MC) while granulocyte and B-cell MC occurred in 80% and 60% of the NST patients, respectively. In CST patients, T-cell MC was detected in 5 patients, of whom 3 were mixed only during the first month. MRD was detected in all NST patients. During the first 3 months the median BCR-ABL/ABL ratio was 0.2% in NST patients compared with 0.01% in CST patients (P < .01). However, 12 months after transplantation, the percentage of reverse transcriptase–polymerase chain reaction (RT-PCR)–positive patients was 20% in NST patients and 50% in CST patients. In conclusion, molecular remission can be induced in most patients after NST, albeit with different kinetics from CST.


2012 ◽  
Vol 2012 ◽  
pp. 1-9 ◽  
Author(s):  
R. Casalegno-Garduño ◽  
C. Meier ◽  
A. Schmitt ◽  
A. Spitschak ◽  
I. Hilgendorf ◽  
...  

Leukemic blasts overexpress immunogenic antigens, so-called leukemia-associated antigens like the receptor for hyaluronan acid-mediated motility (RHAMM). Persistent RHAMM expression and decreasing CD8+T-cell responses to RHAMM in the framework of allogeneic stem cell transplantation or chemotherapy alone might indicate the immune escape of leukemia cells. In the present study, we analyzed the expression of RHAMM in 48 patients suffering from acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Furthermore, we correlated transcripts with the clinical course of the disease before and after treatment. Real-time quantitative reverse transcriptase polymerase chain reaction was performed from RNA of peripheral blood mononuclear cells. T cell responses against RHAMM were assessed by tetramer staining (flow cytometry) and enzyme-linked immunospot (ELISPOT) assays. Results were correlated with the clinical outcome of patients. The results of the present study showed that almost 60% of the patients were RHAMM positive; specific T-cells recognizing RHAMM could be detected, but they were nonfunctional in terms of interferon gamma or granzyme B release as demonstrated by ELISPOT assays. Immunotherapies like peptide vaccination or adoptive transfer of RHAMM-specific T cells might improve the immune response and the outcome of AML/MDS patients.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3118-3118
Author(s):  
Amado J Karduss-Urueta ◽  
Suarez Gloria ◽  
Gonzalez Miguel ◽  
Perez Rosendo ◽  
Pedro Reyes ◽  
...  

Abstract Introduction: T cell replete haploidentical stem cell transplantation with post-transplantation cyclophosphamide (PTCy) has shown encouraging results for the treatment of hematologic malignancies, its main advantage is that almost every patient will have a donor in a timely manner. However this technique has been explored mainly in adults and using bone marrow as a cellular source. Here we present our experience using T cell replete haploidentical peripheral blood stem cell transplantation (TCR-Haplo-PBSCT) with PTCy in 21 pediatric patients whit high-risk acute leukemia Methods and Patients: Donors were mobilized with filgrastim 5 mg/kg/BID for five days, the PBSC were collected with one large volume apheresis procedure. The conditioning consisted of fludarabine 30 mg/m2/day for 5 days, oral busulfan 4-8 mg/kg/split in 1-2 days and, one day before transplant, total body irradiation 400 cGy divided in two fractions (Flu Bu TBI) or fludarabine 150 mgs/m2 split in 5 days, melphalan 100-140 mgs/m2, one day and TBI 200-400 cGy on day - 1 (Flu Mel TBI). All patients were given PTCy 50 mg/kg/day on D+3 and D+4, followed by ciclosporin and mycophenolate starting on day + 5. In all cases filgrastim was administered after transplant beginning on d + 6. After a signed informed consent, 21 patients who needed an urgent transplant, were allografted; median age was 11 years (range 1-16), 10 were girls, the diagnosis were: acute lymphoblastic leukemia 11 patients, acute myeloid leukemia 9, and blastic phase of chronic myeloid leukemia one. 19% were in first remission (CR1), 43% in second (CR2), and 39% in third or with refractory disease(CR3). Results: 17 patients were given Flu Bu TBI conditioning while 4 received Flu Mel TBI combination All the donors shared 4 out of 8 alleles with the recipient; in 62% of the cases the donor was the Mother in 19% the Father and in other 19% one sibling. A median of 16 million of CD34+ cells/kg was infused. The engraftment rate was 100%, median time to achieve 500 neutrophil or more was 15 days (range 14-20), 1 patient out of 21 died without platelet recovery, the remaining had a self- sustained platelet count of 20.000 or more at a median of 14 days (range 10-21). Chimerism at day + 100 was available in 19 cases; all of them had full donor hematopoiesis. The median follow-up is 11 months (range 3-28), the cumulative incidence of graft versus host disease (GVHD) acute grade II-IV and chronic extensive was 23.8% and 25% respectively. Six patients have died, the causes were; pneumonia (n:1) and relapse of leukemia(n:5). In table 1 is presented the overall survival (OS) and event free survival (EFS) for the whole group and discriminated according remission Table 1.Whole groupCR1CR2CR3OS month 1277.4% ± 10100%100%47.3% ± 18.8EFS month 1271.5% ± 10.9100%100%43.8% ± 18.8OS month 2469.6% ± 11.6100%80% ± 17.947.3% ± 18.8EFS month 2463.6% ± 12.3100%83% *22 months21.9% ±18.1 Conclusion: The use of TCR-Haplo-PBSCT with PTCy and a medium intensity conditioning for treating pediatric high risk acute leukemia is promising; it is associated with very good engraftment rate, low transplantation related mortality and an acceptable incidence of GVHD despite the use of peripheral blood. This protocol produces a remarkable leukemia free survival rate, especially in patients in CR1 and CR2. This approach could be a good alternative for children with high-risk leukemia and without suitable matched donors. It deserve further studies Disclosures No relevant conflicts of interest to declare.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 17572-17572
Author(s):  
F. Vari ◽  
T. Rossetti ◽  
D. Munster ◽  
D. Hart

17572 Background: Despite advances in therapy for multiple myeloma, such as the use of proteosome inhibitors or stem cell transplantation, it is usually incurable. One promising therapeutic approach is to eradicate residual disease after stem cell transplantation by immune therapy. Method: The CMRF-56 monoclonal antibody detects an antigen expressed on blood dendritic cells (BDC) that is expressed after a short in vitro culture period. With clinical trail applications in mind, we have developed a procedure for isolation of CMRF-56+ BDC using CliniMACS. Cell culture, antibody staining and immunoselection occur in a closed system, compatible with regulatory requirements for cell therapy. Simultaneously, our team has developed of a bioprocess for production of the murine biotinylated CMRF-56 antibody for clinical trials. The bioprocess incorporates components that remove contaminating DNA, host cell proteins, viral particles and endotoxin. Results: Positive immunoselection using this antibody produces a BDC population containing some contaminating CMRF-56 positive B cells and monocytes. CliniMACS selection of PBMC from an apheresis produces enough BDC for a vaccination program with an average recovery of 1%, purity of 38%, yield of 48% and viability of 70%. These CMRF-56 dendritic cell preparations have been used to generate strong T cell responses to influenza matrix peptide and the putative tumour antigen MART-1. In order to discover and validate additional antigens capable of inducing effective immune responses against multiple myeloma, we have tested numerous of potential target antigens. It has been possibe to generate moderate specific immune responses against HM1.24 and mucin 1 peptides in normal HLA A2 donors. Using the CMRF-56 dendritic cell preparation as antigen presenting cells, it is possible to generate strong cytotoxic T cell responses against a lysate of the myeloma cell line U266. Indicating the potential usefulness of a whole cell lysate in generating anti-myeloma T cells for immunotherapy. Conclusions: These studies have provided the necessary pre-clinical data to commence a Phase I Clinical Trial in 2006 using CMRF-56 BDC preparations loaded with myeloma and control antigens in myeloma patients with minimal residual disease. No significant financial relationships to disclose.


Sign in / Sign up

Export Citation Format

Share Document