scholarly journals Allogeneic, CD34 +, Umbilical Cordblood-Derived NK Cell Adoptive Immunotherapy for the Treatment of Acute Myeloid Leukemia Patients with Measurable Residual Disease

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1745-1745
Author(s):  
Michael Heuser ◽  
Astrid Tschan-Plessl ◽  
Felicitas Thol ◽  
Adrian Schwarzer ◽  
Arnold Kloos ◽  
...  

Abstract Although the majority of patients with acute myeloid leukemia (AML) achieve complete remission (CR), most of them relapse due to measurable residual disease (MRD), leading to poorer prognosis. The elimination of MRD before morphological relapse in patients who achieved CR is therefore an important therapeutic strategy to achieve improved long term outcomes in AML. A promising approach is targeting MRD via natural killer (NK) cell adoptive immunotherapy. NK cells can be used without HLA matching (allogeneic), enabling the transfer of high numbers of immune effector cells, which were not exposed to cytotoxic chemotherapy. A phase I dose-escalation trial in ten elderly subjects with AML in morphologic CR after induction chemotherapy showed that a single infusion of an allogeneic NK cell product generated from umbilical cord blood-derived CD34 + cells, following cyclophosphamide/fludarabine (Cy/Flu) pre-conditioning, was safe and well tolerated, with some patients (2 of 4) achieving MRD negativity (Dolstra et al. 2017). Currently, our dose escalation and expansion trial with the "off-the shelf", cryopreserved, allogeneic NK cell product GTA002 (NCT04632316) is ongoing in adults with AML who are in morphologic CR (including CRi) with MRD and who are not proceeding to allogeneic hematopoietic stem cell transplantation. Two patients have been dosed with a single GTA002 infusion of approximately 5 x 10 8 viable NK cells and completed 6- and 3-month follow-up, respectively. Patient #1 is a 73-year-old male, with AML with recurrent genetic abnormalities (including NPM1mut and FLT3-ITD), who initially received azacitidine and gilteritinib, leading to stable disease. Upon progression, the patient received further treatment with decitabine, followed by 3 cycles of azacitidine/venetoclax (aza/ven), which led to CRi with MRD persistence (confirmed by centralized multiparameter flow cytometry (MFC) on bone marrow (BM)). The patient then received cyclophosphamide (Cy) and fludarabine (Flu) as preconditioning regimen from day -6 to day -3 (Cy: 900 mg/m 2; Flu:15 mg/m 2/day; Flu adjusted for renal impairment), followed by a single intravenous infusion of GTA002 on day 0. The patient experienced a serious adverse event (SAE) on day 5 (grade 3 febrile neutropenia) deemed related to Cy/Flu, which fully resolved on day 16. MRD results by MFC on BM showed that patient #1 converted to MRD negativity (<0.1%) on day 0, which was sustained at 1, 2, 3 and 6 months. Interestingly, NPM1 MRD was detectable by next generation sequencing (MRD-NGS) up to month 1 in peripheral blood (PB), but was cleared by month 2, 3 and 6 in PB (<0.01%VAF). In line with the PB results, NPM1 MRD was detectable in BM at month 1 and was cleared at months 3 and 6. Patient #2 is a 78 year old male with a history of MDS with pancytopenia and IDH2, SRSF2 and PTPN11 mutations that evolved into AML with myelodysplasia-related changes. The patient achieved CR after receiving aza/ven, with MRD positivity after 3 cycles, leading to clinical trial inclusion. He was preconditioned with an adjusted dose of Cy/Flu (Cy 300 and Flu 30 mg/m 2/day from day -5 to day -3), with no SAEs reported. Analysis of BM by MFC showed MRD positivity at screening and on day 0, which turned to MRD negativity at month 1, turning positive again at month 2 and 3. Follow up in PB and BM by MRD-NGS showed that the IDH2 and SRSF2 clones persisted after preconditioning and GTA002 infusion, but the PTPN11 clone became undetectable in PB by Day 0 and in BM by month 2 and month 3. Taken together, these cases describe clearance of MRD by MFC and NGS in AML with a single dose infusion of allogeneic, CD34+ cord blood derived NK cells in one patient and clearance of the PTPN11 mutated clone in another one, reconfirming the excitement to further investigate this treatment strategy for disease control. The delayed clearance of MRD could suggest an ongoing immunotherapeutic effect that will be further investigated. Disclosures Heuser: Karyopharm: Research Funding; Daichi Sankyo: Honoraria, Research Funding; Bayer AG: Honoraria, Research Funding; BMS/Celgene: Research Funding; Jazz Pharmaceuticals: Honoraria, Research Funding; BergenBio: Research Funding; Astellas: Research Funding; Novartis: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Roche: Research Funding; Tolremo: Honoraria; AbbVie: Honoraria; Janssen: Honoraria. Thol: Abbvie: Honoraria; Astellas: Honoraria; Novartis: Honoraria; Jazz: Honoraria; Pfizer: Honoraria; BMS/Celgene: Honoraria, Research Funding. Schwarzer: Bantam Pharmaceutical: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Pinkernell: Medigene AG: Ended employment in the past 24 months; Glycostem Therapeutics: Current Employment. Ganser: Jazz Pharmaceuticals: Honoraria; Novartis: Honoraria; Celgene: Honoraria.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3695-3695 ◽  
Author(s):  
Michele Malagola ◽  
Crisitina Skert ◽  
Enrico Morello ◽  
Francesca Antoniazzi ◽  
Erika Borlenghi ◽  
...  

Abstract Background: Although a complete remission (CR) can be achieved in 70-80% of newly diagnosed acute myeloid leukemia (AML) patients, relapses occur in up to the 50% of cases. Thus, minimal residual disease (MRD) monitoring is a major issue for early detection of patients at high-risk of treatment failure and relapse. Aim: to dynamically evaluate WT1 pan-leukemic molecular marker of MRD in patients with AML. Matherial and methods: 107 newly diagnosed AML patients consecutively treated between 2010 and 2013 were monitored with quantitative WT-1 from bone marrow (BM) and peripheral blood (PB) at baseline, after induction, after the first consolidation course, before allogeneic stem cell transplantation (allo-SCT), at the 3rd and the 6th month after transplantation Results: At diagnosis, 104/107 (97%) had increased PB and BM WT1 levels assessed according to the ELN assay. Eighty-eight out of 107 patients (82%) achieved a complete remission (CR) after induction, 30/88 (34%) relapsed during follow up and 24/107 (22%) were addressed to allogeneic stem cell transplantation (allo-SCT). By univariate analysis, PB-WT > 50x10^4/ABL and BM-WT1 > 250x10^4/ABL after induction (PB: p=0.02; BM: p=0.04), after consolidation (PB: p=0.003), at the end of treatment (PB and BM: p=0.001), at 3rd month of follow up (PB and BM: p=0.005) and at 6th month of follow up (PB: p=0.005) were associated with a reduced overall survival (OS). By multivariate analysis, a BM-WT1 > 250 x 10^4/ABL at the end of treatment was significantly associated with a reduced OS. In order to adapt the cut-off of WT1 in our series of patients, we considered WT1 levels as continuous variables and categorized them at approximately the 25th, 50th, and 75th percentile. A cut-off of PB-WT1 > 25x10^4/ABL and BM-WT1 > 125x10^4/ABL at the end of the treatment program was identified as correlated with reduced leukemia-free survival (LFS) and OS (p=0.001). Similarly, and restricting the analysis on the 24 patients allo-transplanted in CR, 8/11 (73%) with pre-transplant PB-WT1 ≥ 5 and 4/13 (31%) with PB-WT1 < 5 relapsed, respectively (p=0.04). The incidence of relapse was higher in AML patients with PB-WT1 ≥ 5 measured at 3rd (56% vs 38%; p=0.43) and 6th month (71% vs 20%; p=0.03) after allo-SCT. Interestingly, 5/5 (100%) patients with pre-transplant PB-WT1 ≥ 5 who never reduced this level at 3rd or 6th month after allo-SCT experienced a disease recurrence. Conclusions: our data, although retrospectively collected, show that WT1 monitoring may be useful to predict the relapse in AML patients. Acknowledgments: This work was supported in part by Banca di Credito Cooperativo di Pompiano e Franciacorta and Lions Club Bassa Bresciana Association. Disclosures Russo: Celgene: Research Funding; Gilead: Research Funding; Novartis: Consultancy.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5294-5294
Author(s):  
Patrick Schlegel ◽  
Chihab Klose ◽  
Christina Kyzirakos ◽  
Ursula J.E. Seidel ◽  
Kai Witte ◽  
...  

Abstract GMP-grade NK cell expansion for clinical purpose has been demonstrated feasible and safe. Here we share our pilot data on posttransplant immunotherapy with ex vivo expanded NK cells to treat minimal residual disease in a pediatric patient with posttransplant relapsed myeloid leukemia. Our patient, a 13 year old boy who underwent 2nd allogeneic stem cell transplantation (haploidentical stem cell transplantation from his mother) due to posttransplant relapsed acute myeloid leukemia. After the 2nd haploidentical stem cell transplantation (SCT) minimal residual disease (MRD) was detected by multiparameter flow cytometry and by two molecular markers CALM-AF10 fusion transcript and a NRAS-mutation. For posttransplant compassionate use immunotherapy by NK cell transfer, NK cells were expanded from untouched isolated PBMCs of the patient post 2nd haploidentical SCT. GMP-grade expansion of the NK cells was done under static conditions in our GMP-facility. Isolated PBMCs were pooled with 100Gy irradiated K562mb15 4-1BBL feeder cells (kindly provided by Dario Campana) in a proportion of 1:20 (NK to K562mb15 4-1BBL). PBMCs and K562mb15 4-1BBL were seeded in conventional cell culture flasks (175cm2) at a density of 1.1E6 cells/ml. Cell culture media contained RPMI1640 supplemented with 10% AB-human serum, 1% L-glutamine and 100IU Proleukine® IL2/ml. Cell culture was monitored daily for cell number, white blood cell differentiation, pH of the cell culture, glucose metabolism, lactate production and microbial sterility testing at the beginning and the end of the expansion period. The cell product was harvested on day 15-17. Fresh isolated PBMCs and the expanded NK cell product were characterized by flow cytometry. NK cells were expanded &gt;1000 fold (3.1 and 3.4 log-fold) in 14-17 days. The product contained a total number of 9.8E9 and 19.9E9 cells, which was 328 and 665E6/kgBW. The expansion protocol supports NK and γδ T cell expansion whereas the number of αβ T cells stays stable. Cytotoxicity assay against various targets revealed excellent cellular cytotoxicity and antibody dependent cellular cytotoxicity. To prevent relapse in our patient with posttransplant MRD positivity, NK cells from the patient post 2nd haploidentical SCT were expanded for cellular immunotherapy. 2 weeks post 1st NK cell transfer (day +170) the patient achieved complete MRD response in the bone marrow. Unfortunately the patient showed detectable MRD one month later. Therefore another NK cell expansion and transfer was done. 2 weeks post 2nd NK cell transfer (day +232) the patient again achieved complete MRD response in the bone marrow and is in complete molecular remission ever since (day +340). The NK cell products were tolerated well. Transient coughing and temporary increase of temperature were registered. Both, in vitro and in vivo effect of the NK cell product were documented. Clinical use of expanded and activated NK cells and γδ T cells can induce molecular remission in posttransplant MRD positive acute myeloid leukemia. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS7053-TPS7053
Author(s):  
Michael Heuser ◽  
Walter M. Fiedler ◽  
Tessa Kerre ◽  
Johan Maertens ◽  
Jakob Passweg ◽  
...  

TPS7053 Background: Acute myeloid leukemia (AML) is a malignant disease with poor long-term prognosis in patients who cannot achieve morphological and molecular remission. Although insights into AML biology and treatment modalities have improved over recent years and even though many patients achieve morphological complete remission (CR), most are still relapsing. These relapses are due to residual leukemia stem cells that can be identified as minimal/measurable residual disease (MRD), with MRD serving as a predictive factor for relapse and mortality. Elimination of MRD in patients having reached CR is seen as essential for optimal and persistent clinical responses. A promising approach is the development of adoptive immunotherapies aimed at directly eradicating tumor cells using T-cells or natural killer (NK) cells. NK cells are part of the body’s innate immune system and play a key role in controlling viral infections and conducting tumor immunosurveillance. Furthermore, NK cells can be applied clinically in an allogeneic setting, enabling the supply of high numbers of immune effector cells, which were not exposed to cytotoxic chemotherapeutics. A proprietary ex vivo expansion and differentiation method in a fully closed, automated manufacturing platform was developed to generate GTA002, an “off-the-shelf” (allogeneic), cryopreserved NK cell preparation, generated from CD34+ hematopoietic stem and progenitor cells derived from umbilical cord blood. The safety and tolerability of the product was already demonstrated in a Phase I trial in elderly patients with AML (PMLA25) (Dolstra et al. 2017). Methods: We are currently conducting a prospective 2-stage, open-label, single arm, multicenter Phase I/IIa trial to evaluate the safety and efficacy of GTA002 in 33 adults with AML who are in CR with MRD and who are not proceeding to allogeneic HSCT (ClinicalTrials.gov Identifier: NCT04632316). Patients enrolled in the clinical trial receive a lymphodepleting conditioning regimen consisting of cyclophosphamide and fludarabine (Cy/Flu) followed by up to 3 NK cell infusions 4 days apart and will be followed up for 12 months. The dose escalation stage of the trial will assess the safety and tolerability of repeat NK cell infusions in a 3+3 design with 3 cohorts and a cumulative dose range of 325 to 3,000 x106 viable NK cells. The expansion stage will evaluate the safety, tolerability and efficacy of NK cell infusions in 24 additional subjects. The primary efficacy endpoint is the cumulative incidence of the MRD response and secondary efficacy endpoints include the duration of the MRD response, event-free survival, overall survival and cumulative incidence of relapse. Enrollment in the first cohort (one single NK cell infusion) started in December 2020. Clinical trial information: EudraCT number 2019-003686-17.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4656-4656
Author(s):  
Veit Bücklein ◽  
Alexandra Stein ◽  
Benjamin Tast ◽  
Thomas Koehnke ◽  
Karsten Spiekermann ◽  
...  

Patients with Acute Myeloid Leukemia (AML) frequently relapse due to chemorefractory AML cells persisting after intensive chemotherapy at levels below the 5% morphological detection threshold (measurable residual disease, MRD). MRD has been established as an important prognostic factor for relapse-free and overall survival, making it highly relevant for post-remission treatment stratification. In contrast to MRD assessment by molecular techniques, multiparameter flow cytometry (MFC)-based MRD measurements are applicable in more than 95% of AML patients, while still offering a sensitivity of 10-4 to 10-5. Current MFC MRD assessment strategies measure 8-10 fluorochromes in parallel, resulting in a high-dimensional data set. However, evaluation of this data is usually performed by scatterplot-based manual, two-dimensional analysis. This leads to loss of information and significant inter-observer variability in MRD diagnostics. We therefore established a computational data analysis strategy for MFC MRD diagnostics, based on the unsupervised FlowSOM algorithm. By comparison with healthy bone marrow (HBM) data, FlowSOM analysis can identify aberrant (sub-)populations of cells, clustered in nodes (according to similarity of their antigen profile). These nodes can be denoted as "nodes of interest" (NOI) to simplify MRD analysis after clustering. Aim of the project was to establish FlowSOM analysis protocols and retrospectively evaluate their prognostic significance in a cohort of 46 patients with known outcomes. Bone marrow samples of these patients were analyzed at aplasia (day 16 after initiation of induction chemotherapy). Only patients with morphological blast clearance at aplasia were included. Healthy reference FlowSOM trees were established by merging flow data of 17 HBM. Analysis protocols were developed to report individual ("any node" approach) and cumulative ("sum node" approach) differences in NOI percentages when comparing HBM and MRD samples. We then performed FlowSOM MRD analyses in a patient subcohort of 19 AML patients. Importantly, for these analyses, we excluded patients who underwent allogeneic stem cell transplantation in first remission (non-HSCT subcohort). Median follow-up time was 8.3 (range 2-40) months for this subcohort. Receiver operating characteristic (ROC) analyses were used to determine optimal threshold values to differentiate relapse (n=5) and non-relapse (n=14) patients within the cohort. For "sum node" analysis strategies (defining MRD levels as cumulative difference of NOI percentages) a threshold of -2.44% was identified, optimized for Youden (Y) index and diagnostic odds ratio (DOR). For the "any node" strategy (defining MRD levels by the maximum difference of any NOI), a threshold of 0.04%, also optimized for the Y-index and DOR, discriminated best between relapse and non-relapse patients. Relapse-free survival (RFS) was significantly shorter for MRD-positive (MRDpos) patients identified by "sum node" analysis (median 8 months vs. not reached, p=0.016) and tended to be shorter for MRDpos patients by "any node" analysis (median 8 months vs. not reached, p=0.1). When applying the thresholds identified in the non-HSCT cohort to the full set of 46 patients (median follow-up interval 10.6 months, range 2-40), median RFS was not reached for the MRD-negative group (both for "sum node" and "any node" analysis), and was 14 ("sum node", p=0.098) and 14 months ("any node", p=0.360) for the MRDpos patients. Median overall survival for MRDpos patients by "sum node" analysis was 27 months, whereas it was not reached for MRD-negative patients. However, this difference did not reach statistical significance (p=0.335), probably due to the small sample size. Taken together, FlowSOM-based analysis strategies seem well suited to identify patients with MRD positivity after intensive induction chemotherapy. MFC MRD positivity at aplasia, defined by FlowSOM-based analysis, is associated with inferior RFS in retrospective analyses of small patient cohorts. Due to the underlying computational, unsupervised data analysis, FlowSOM-based assessment can be a means to harmonize MFC MRD evaluation. These promising results need to be verified in larger cohorts, with inclusion of post-induction assessments, and should be followed by prospective analyses to delineate the diagnostic validity of FlowSOM for AML MRD diagnostics in clinical trials. Disclosures Subklewe: Janssen: Consultancy; Morphosys: Research Funding; Celgene: Consultancy, Honoraria; Gilead: Consultancy, Honoraria, Research Funding; Miltenyi: Research Funding; Oxford Biotherapeutics: Research Funding; Pfizer: Consultancy, Honoraria; AMGEN: Consultancy, Honoraria, Research Funding; Roche: Consultancy, Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 16-17
Author(s):  
Nicholas J. Short ◽  
Shouhao Zhou ◽  
Chenqi Fu ◽  
Donald Berry ◽  
Roland P Walter ◽  
...  

Background: The persistence of measurable residual disease (MRD) has been shown to correlate with higher rates of relapse and worse survival in several leukemias. Individual studies have similarly suggested that the detection of MRD is associated with inferior outcomes in patients with acute myeloid leukemia (AML) undergoing frontline therapy; however, the optimal use of MRD information to risk stratify patients and inform clinical decision-making has been limited in part by the heterogeneity of these reports. To better understand the relationship between MRD and clinical outcomes in AML, we performed a literature-based meta-analysis of published AML studies reporting the association of MRD with survival outcomes. Methods: We conducted independent searches of PubMed, MEDLINE, and Embase for papers published between January 1, 2000 and October 1, 2018 that included the keywords "AML," "acute myeloid leukemia," or "acute myelogenous leukemia," in combination with the keywords "MRD," "minimal residual disease," or "measurable residual disease." Reviews, non-English articles, and studies only reporting outcomes after hematopoietic stem cell transplantation or those with insufficient description of MRD information were excluded. Study sample size, patient age, median follow-up time, MRD detection method, MRD assessment time point(s), AML subtype, specimen source, and survival outcomes were extracted. Meta-analyses of survival probabilities and hazard ratios (HRs) were performed separately for disease-free survival (DFS) and overall survival (OS) using Bayesian hierarchical modeling. Results: Eighty-one publications reporting on 11,151 patients were included in the meta-analysis (17 studies for OS, 20 studies for DFS, and 44 for both). Both OS and DFS were better for those who achieved MRD negativity (Figure 1). At 5 years, the estimated OS was 68% (95% Bayesian credible interval [CrI], 63%-73%) for the MRD negative group versus 34% (95% CrI, 28%-40%) for the MRD positive group. Similarly, the estimated DFS at 5 years for the MRD negative and MRD positive groups were 64% (95% CrI, 59%-70%) and 25% (95% CrI, 20%-32%), respectively. The relative benefit of achieving MRD negativity was comparable for both OS and DFS (average HR, 0.36; 95% CrI, 0.33-0.39 for OS; average HR, 0.37; 95% CrI, 0.34-0.40 for DFS). Achievement of MRD negativity was associated with superior DFS and OS irrespective of age group (adult or pediatric), MRD assessment time point (induction, during consolidation or after consolidation), AML subgroup (core-binding factor [CBF] or non-CBF), or specimen source (bone marrow or peripheral blood) (Figure 2). While most of the MRD detection methods were able to identify a difference in DFS and OS between groups with MRD negativity versus MRD positivity, the MRD effect using cytogenetics/FISH was not significant (average HR, 0.77; 95% CrI, 0.39-1.56 for OS and average HR, 0.65; 95% CrI, 0.34-1.23 for DFS), possibly due to only 2 studies being included in this subgroup analysis. The effect of MRD on survival was more profound in studies reporting outcomes of CBF AML compared to non-CBF AML, with a posterior probability of 0.999 for OS and 0.997 for DFS. Interestingly, across AML subtypes, peripheral blood assessment of MRD better distinguished MRD-positive and MRD-negative groups as compared to bone marrow assessment of MRD, with a posterior probability of 0.918 for OS and 0.999 for DFS. Multivariate analysis was performed to control for possible confounding factors that might influence the subgroup analyses. Overall, multivariate results were consistent with the univariate results. All subgroups showed DFS and OS benefit to achievement of MRD negativity, with the exception of MRD detection by cytogenetics/FISH. Conclusions: In this large-cohort meta-analysis, achievement of MRD negativity was strongly associated with superior DFS and OS in patients with AML, an association that was observed across ages, disease subtypes, time of assessment, specimen source, and most MRD detection methods. Given the robustness of the association of MRD with long-term outcomes across studies, MRD status may serve as a valuable clinical endpoint that may allow for accelerated evaluation of novel therapies in AML. Disclosures Short: Astellas: Research Funding; Takeda Oncology: Consultancy, Honoraria, Research Funding; AstraZeneca: Consultancy; Amgen: Honoraria. Berry:Berry Consultants LLC.: Other: Co-owner. Walter:StemLine: Research Funding; Selvita: Research Funding; Seattle Genetics: Research Funding; Race Oncology: Consultancy; BioLineRx: Consultancy, Research Funding; Astellas: Consultancy; Arog: Research Funding; Argenx: Consultancy; Aptevo: Consultancy, Research Funding; Amphivena: Current equity holder in publicly-traded company; Amgen: Consultancy, Research Funding; Agios: Consultancy, Research Funding; Jazz: Consultancy, Research Funding; ImmunoGen: Research Funding; Genentech: Consultancy; Daiichi: Consultancy; Celgene: Consultancy, Research Funding; Boston Biomedical: Consultancy; BiVictriX: Consultancy; Pfizer: Consultancy, Research Funding; New Link Genetics: Consultancy; Macrogenics: Research Funding; Kite: Consultancy. Kantarjian:Immunogen: Research Funding; Daiichi-Sankyo: Honoraria, Research Funding; BioAscend: Honoraria; Delta Fly: Honoraria; Abbvie: Honoraria, Research Funding; Ascentage: Research Funding; Novartis: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Amgen: Honoraria, Research Funding; Janssen: Honoraria; Oxford Biomedical: Honoraria; Adaptive biotechnologies: Honoraria; Actinium: Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi: Research Funding; Aptitute Health: Honoraria; BMS: Research Funding; Jazz: Research Funding. Ravandi:Orsenix: Consultancy, Honoraria, Research Funding; BMS: Consultancy, Honoraria, Research Funding; Macrogenics: Research Funding; Jazz Pharmaceuticals: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria, Research Funding; Xencor: Consultancy, Honoraria, Research Funding; AstraZeneca: Consultancy, Honoraria; Astellas: Consultancy, Honoraria, Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 624-624
Author(s):  
Antonio Curti ◽  
Loredana Ruggeri ◽  
Sarah Parisi ◽  
Andrea Bontadini ◽  
Elisa Dan ◽  
...  

Abstract Introduction: This is a Phase 1b monocentric clinical trial, aimed to evaluate the safety, feasibility and preliminary clinical efficacy of alloreactive natural killer (NK) cell infusions after immunosuppressive regimen in elderly acute myeloid leukemia (AML) patients, who achieved first complete remission (CR) after induction/consolidation chemotherapy. Methods: Seventeen AML patients, in first morphological CR (3 patients showed molecular disease) (median age 64 years, range 53-73) received highly purified CD56+CD3- NK cells from haploidentical KIR-ligand mismatched donors after fludarabine/cyclophosphamide (Flu/Cy) immunosuppressive chemotherapy, followed by interleukin-(IL-) 2. Results: No signs of NK cell-related toxicity, including graft-versus-host disease, were observed. Myelosuppression was moderate although 1 patient died due to overwhelming bacterial pneumonia and was censored for clinical follow-up. With a median follow-up of 22.5 months (range, 6-68 months), 9/16 evaluable patients (0.56) are alive disease-free, whereas 7/16 (0.44) relapsed with a median time to relapse of 9 months (range, 3-51 months). Among relapsed patients, 2 individuals showed a very prolonged CR phase of 24 and 51 months, respectively, in absence of any concomitant anti-leukemia treatments. Three patients, treated with molecular disease, achieved molecular CR lasting 9 and 4 months in 2 cases and 8+ months in the third patient. These results have been compared to that obtained in a cohort of patients, who achieved CR after induction/consolidation chemotherapy, but did not undergo NK cell immunotherapy due to the absence of a KIR-L mismatched donor (Fig. 1). Disease-free survival (DFS) in the control cohort was 0.39, indicating a trend towards better DFS under the NK therapy (0.56; p=0.07). To correlate the donor NK activity with the clinical response, donor NK cells, as evaluated both as the total number of infused NK cells and as the frequency of alloreactive NK clones, were assessed before NK cell infusion. No statistically significant correlation between the total number of infused NK cells and clinical response was observed. On the contrary, we observed a higher number of donor-derived alloreactive NK cell clones in the group of responders as compared to that of non-responders (Fig. 2A). Accordingly to statistical analysis, a threshold of 8 over 100 NK clones was chosen as the cut-off level discriminating responders versus non-responders (Fig. 2A). Importantly, the infusion of higher number of donor alloreactive NK cells (more than 8/100 clones) was associated with prolonged DFS (Fig.2B). In particular, for patients who received more than 8/100 alloreactive NK cell clones the probability of DFS was 0.81, whereas patients who received less than 8/100 clones had a DFS of 0.14. Such difference was statistically significant (p=0.03) and demonstrates that the number of alloreactive NK cell clones in the donor may discriminate patients receiving NK cell therapy as for response evaluation. Interestingly, the DFS of the group of patients who received less than 8/100 donor alloreactive NK cell clones was comparable to that of the historical control group (Fig. 2B). This finding suggests that the therapeutical effect of the whole procedure, which comprises chemotherapy (Flu/Cy) plus NK immunotherapy, mainly relies on the anti-leukemia activity of alloreactive NK cells. Noteworthy, the increased anti-leukemia effect in patients receiving the higher number of donor alloreactive NK cells was not associated with increased myelosuppression, as shown by comparable data of hematological recovery after NK cell infusion in responders and non-responders.Conclusions: infusion of purified NK cells is feasible in elderly patients with AML as post-CR consolidation strategy and donor NK alloreactivity has a predictive role on the clinical outcome of treated patients. Supported by: Italian Leukemia Association (BolognAIL), Section of Bologna and Fondazione Carisbo, Bologna, ELN, AIRC, PRIN, progetto Regione-Università 2010-12 (L. Bolondi), FP7 NGS-PTL project. Figure 1 Figure 1. Figure 2 Figure 2. Disclosures No relevant conflicts of interest to declare.


Leukemia ◽  
2021 ◽  
Author(s):  
Tanja Božić ◽  
Chao-Chung Kuo ◽  
Jan Hapala ◽  
Julia Franzen ◽  
Monika Eipel ◽  
...  

AbstractAssessment of measurable residual disease (MRD) upon treatment of acute myeloid leukemia (AML) remains challenging. It is usually addressed by highly sensitive PCR- or sequencing-based screening of specific mutations, or by multiparametric flow cytometry. However, not all patients have suitable mutations and heterogeneity of surface markers hampers standardization in clinical routine. In this study, we propose an alternative approach to estimate MRD based on AML-associated DNA methylation (DNAm) patterns. We identified four CG dinucleotides (CpGs) that commonly reveal aberrant DNAm in AML and their combination could reliably discern healthy and AML samples. Interestingly, bisulfite amplicon sequencing demonstrated that aberrant DNAm patterns were symmetric on both alleles, indicating that there is epigenetic crosstalk between homologous chromosomes. We trained shallow-learning and deep-learning algorithms to identify anomalous DNAm patterns. The method was then tested on follow-up samples with and without MRD. Notably, even samples that were classified as MRD negative often revealed higher anomaly ratios than healthy controls, which may reflect clonal hematopoiesis. Our results demonstrate that targeted DNAm analysis facilitates reliable discrimination of malignant and healthy samples. However, since healthy samples also comprise few abnormal-classified DNAm reads the approach does not yet reliably discriminate MRD positive and negative samples.


Sign in / Sign up

Export Citation Format

Share Document