scholarly journals Transcriptome Based Projection of Single Cells to Uncover Development and Heterogeneity of Abnormal Hematopoietic Cells

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2520-2520
Author(s):  
Parashar Dhapola ◽  
Mikael Sommarin ◽  
Mohamed Eldeeb ◽  
Amol Ugale ◽  
David Bryder ◽  
...  

Single-cell transcriptomics (scRNA-Seq) has accelerated the investigation of hematopoietic differentiation. Based on scRNA-Seq data, more refined models of lineage determination in stem- and progenitor cells are now available. Despite such advances, characterizing leukemic cells using single-cell approaches remains challenging. The conventional strategies of scRNA-Seq analysis map all cells on the same low dimensional space using approaches like tSNE and UMAP. However, when used for comparing normal and leukemic cells, such methods are often inadequate as the transcriptome of the leukemic cells has systematically diverged, resulting in irrelevant separation of leukemic subpopulations from their healthy counterpart. Here, we have developed a new computational approach bundled into a tool called Nabo (nabo.readthedocs.io) that has the capacity to directly compare cells that are otherwise unalignable. First, Nabo creates a shared nearest neighbor graph of the reference population, and the heterogeneity of this population is subsequently defined by performing clustering on the graph and calculating a low dimensional representation using t-SNE or UMAP. Nabo then calculates the similarity of incoming cells from a target population to each cell in the reference graph using a modified Canberra metric. The reference cells with higher similarity to the target cells obtain higher mapping scores. The built-in classifier is used to assign each target cell a reference cluster identity. We tested Nabo's accuracy on control datasets and found that Nabo's performance in terms of accuracy and robustness of projection is comparable to state-of-art methods. Moreover, Nabo is a generalized domain adaptation algorithm and hence can perform classification of target cells that are arbitrarily dissimilar to reference cells. Nabo could identify the cell-identity of sorted CD19+ B cells, CD14+ monocytes and CD56+ by projecting these unlabeled cells onto labelled peripheral blood mononuclear cells with an average specificity higher than 0.98. The general applicability of Nabo was demonstrated by successfully integrating pancreatic cells, sequenced in three different studies using different sequencing chemistries with comparable or better accuracy than existing methods. Also, it was conclusively demonstrated that Nabo can predict the identity of human HSPC subpopulations to the same accuracy as can be achieved by established cell-surface markers. Having Nabo at hand, we aimed to uncover the heterogeneity of hematopoietic cells from different stages of AML. Nabo showed that AML cells lacked the heterogeneity of normal CD34+ cells and were devoid of cells with HSC gene signature. A large patient-to-patient variability was found where leukemic cells mapped to distinct stages of myeloid progenitors. To ask whether this variability could reflect differences in leukemia-initiating cell identity, we induced leukemia in murine granulocyte-monocyte-lymphoid progenitors (GMLPs) using an inducible model for MLL-ENL-driven AML. On projection, more than 70% of MLL-ENL-activated cells mapped to a distinct Flt3+ subpopulation present within healthy GMLPs. Statistical validity of this projection was verified using two novel null models for testing cell projections: 1) ablated node model, wherein the mapping strength of target cells are evaluated after removal of high mapping score source nodes, and 2) high entropy features model, which rules out the background noise effect. By separating Flt3+ and Flt3- cells prior to activation of the fusion gene and performing in vitro replating assays, we could demonstrate that Flt3+ GMLPs contained 3-4 fold more leukemia-initiating cells (1/1.34 cells) than Flt3- GMLPs (1/4.89 cells), indicating that leukemia-initiating cells within GMLPs express Flt3. Taken together, Nabo represents a robust cell projection strategy for relevant analysis of scRNA-Seq data that permits an interpretable inference of cross-population relationships. Nabo is designed to compare disparate cellular populations by using the heterogeneity of one population as a point of reference allowing for cell-type specification even following perturbations that have resulted in large molecular changes to the cells of interest. As such, Nabo has critical implementation for delineation of leukemia heterogeneity and identification of leukemia-initiating cell population. Disclosures No relevant conflicts of interest to declare.

Development ◽  
2001 ◽  
Vol 128 (13) ◽  
pp. 2517-2524 ◽  
Author(s):  
Robert Klapper ◽  
Sandra Heuser ◽  
Thomas Strasser ◽  
Wilfried Janning

In order to reveal syncytia within the visceral musculature of Drosophila melanogaster, we have combined the GAL4/UAS system with the single-cell transplantation technique. After transplantation of single cells from UAS-GFP donor embryos into ubiquitously GAL4-expressing recipients, the expression of the reporter gene was exclusively activated in syncytia containing both donor- and recipient-derived nuclei. In the first trial, we tested the system in the larval somatic musculature, which is already known to consist of syncytia. By this means we could show that most of the larval somatic muscles are generated by clonally non-related cells. Moreover, using this approach we were able to detect syncytia within the visceral musculature – a tissue that has previously been described as consisting of mononuclear cells. Both the longitudinal visceral musculature of the midgut and the circular musculature of the hindgut consist of syncytia and persist through metamorphosis. This novel application of the transplantation technique might be a powerful tool to trace syncytia in any organism using the GAL4/UAS system.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Ayshwarya Subramanian ◽  
Eriene-Heidi Sidhom ◽  
Maheswarareddy Emani ◽  
Katherine Vernon ◽  
Nareh Sahakian ◽  
...  

AbstractHuman iPSC-derived kidney organoids have the potential to revolutionize discovery, but assessing their consistency and reproducibility across iPSC lines, and reducing the generation of off-target cells remain an open challenge. Here, we profile four human iPSC lines for a total of 450,118 single cells to show how organoid composition and development are comparable to human fetal and adult kidneys. Although cell classes are largely reproducible across time points, protocols, and replicates, we detect variability in cell proportions between different iPSC lines, largely due to off-target cells. To address this, we analyze organoids transplanted under the mouse kidney capsule and find diminished off-target cells. Our work shows how single cell RNA-seq (scRNA-seq) can score organoids for reproducibility, faithfulness and quality, that kidney organoids derived from different iPSC lines are comparable surrogates for human kidney, and that transplantation enhances their formation by diminishing off-target cells.


Blood ◽  
1998 ◽  
Vol 91 (6) ◽  
pp. 2197-2207 ◽  
Author(s):  
Edus H. Warren ◽  
Philip D. Greenberg ◽  
Stanley R. Riddell

Abstract Cytotoxic T lymphocytes (CTL) specific for human minor histocompatibility (H) antigens can be isolated from the blood of major histocompatibility complex (MHC)-matched allogeneic bone marrow transplant (BMT) recipients and may play a prominent role in the graft-versus-host (GVH) and graft-versus-leukemia (GVL) reactions (Tsoi et al, J Immunol 125:2258, 1980; Tsoi et al, Transplant Proc 15:1484, 1983; Goulmy et al, Nature 302:159, 1983; Irle et al, Transplantation 40:329, 1985; and Niederwieser et al, Blood 81:2200, 1993). The identification of minor H antigens that are expressed in hematopoietic cells, including leukemic cells, but not in fibroblasts and other tissue types has suggested that such tissue-restricted antigens could potentially serve as targets for T-cell immunotherapy to enhance GVL activity without inducing GVH disease (de Bueger et al, J Immunol 149:1788, 1992; van der Harst et al, Blood 83:1060, 1994; and Dolstra et al, J Immunol 158:560, 1997). To explore the feasibility of this strategy, donor CD3+CD8+ CTL clones specific for recipient minor H antigens were isolated and characterized from allogeneic BMT recipients. CTL clones were obtained from the majority of donor/recipient pairs. Seventeen distinct minor H antigens distinguishable by their MHC-restricting allele, population frequency, and/or distribution of tissue expression were defined by 56 CD3+CD8+ CTL clones isolated from these patients. The MHC-restricting alleles for these CTL clones included HLA-A2 and HLA-B7, which had previously been shown to present minor H antigens to CTL, as well as HLA-A3, -A11, -B8, -B53, and -Cw7, which had not previously been described to present minor H antigens to CTL. Estimated phenotype frequencies for these 17 distinct minor H antigens range from 0.17 to 0.92. In vitro cytotoxicity assays using hematopoietic cells and fibroblasts as target cells showed that 5 of the 17 minor H antigens were expressed in both hematopoietic cells and fibroblasts. However, 12 were presented for CTL recognition only by hematopoietic cells and not by dermal fibroblasts derived from the same donors. These results significantly extend the spectrum of CTL-defined human minor H antigens that could potentially serve as target antigens for cellular immunotherapy to promote GVL activity after allogeneic BMT.


2017 ◽  
Vol 114 (18) ◽  
pp. E3659-E3668 ◽  
Author(s):  
Ann Wiegand ◽  
Jonathan Spindler ◽  
Feiyu F. Hong ◽  
Wei Shao ◽  
Joshua C. Cyktor ◽  
...  

Little is known about the fraction of human immunodeficiency virus type 1 (HIV-1) proviruses that express unspliced viral RNA in vivo or about the levels of HIV RNA expression within single infected cells. We developed a sensitive cell-associated HIV RNA and DNA single-genome sequencing (CARD-SGS) method to investigate fractional proviral expression of HIV RNA (1.3-kb fragment of p6, protease, and reverse transcriptase) and the levels of HIV RNA in single HIV-infected cells from blood samples obtained from individuals with viremia or individuals on long-term suppressive antiretroviral therapy (ART). Spiking experiments show that the CARD-SGS method can detect a single cell expressing HIV RNA. Applying CARD-SGS to blood mononuclear cells in six samples from four HIV-infected donors (one with viremia and not on ART and three with viremia suppressed on ART) revealed that an average of 7% of proviruses (range: 2–18%) expressed HIV RNA. Levels of expression varied from one to 62 HIV RNA molecules per cell (median of 1). CARD-SGS also revealed the frequent expression of identical HIV RNA sequences across multiple single cells and across multiple time points in donors on suppressive ART consistent with constitutive expression of HIV RNA in infected cell clones. Defective proviruses were found to express HIV RNA at levels similar to those proviruses that had no obvious defects. CARD-SGS is a useful tool to characterize fractional proviral expression in single infected cells that persist despite ART and to assess the impact of experimental interventions on proviral populations and their expression.


Blood ◽  
1990 ◽  
Vol 75 (10) ◽  
pp. 1941-1946 ◽  
Author(s):  
H Ema ◽  
T Suda ◽  
Y Miura ◽  
H Nakauchi

Abstract To characterize human hematopoietic progenitors, we performed methylcellulose cultures of single cells isolated from a population of CD34+ cells by fluorescence-activated cell-sorting (FACS) clone-sorting system. CD34+ cells were detected in bone marrow (BM) and peripheral blood (PB) cells at incidences of 1.0% and 0.01% of total mononuclear cells, respectively. Single cell cultures revealed that approximately 37% of BM CD34+ cells formed colonies in the presence of phytohemagglutinin-leukocyte conditioned medium and erythropoietin. Erythroid bursts-, granulocyte-macrophage (GM) colony-, and pure macrophage (Mac) colony-forming cells were 10% each in CD34+ cells. Approximately 15% of PB CD34+ cells formed colonies in which erythroid bursts were predominant. CD34+ cells were heterogeneous and fractionated by several antibodies in FACS multicolor analysis. In these fractionated cells, CD34+, CD33+ cells formed GM and Mac colonies 7 to 10 times as often as CD34+, CD33- cells. Most of the erythroid bursts and colonies were observed in the fraction of CD34+, CD13- cells or CD34+, CD33- cells. The expression of HLA-DR on CD34+ cells was not related to the incidence, size, or type of colonies. There was no difference in the phenotypical heterogeneity of CD34+ cells between BM and PB. About 10% of CD34+ cells were able to form G colonies in response to granulocyte colony-stimulating factor (G-CSF) and to form Mac colonies in GM-CSF or interleukin-3 (IL-3). Progenitors capable of generating colonies by stimulation of G-CSF were more enriched in CD34+, CD33+ fraction than in CD34+, CD33- fraction. Thus, single cell cultures using the FACS clone-sorting system provide an accurate estimation of hematopoietic progenitors and an assay system for direct action of colony-stimulating factors.


2011 ◽  
Vol 57 (7) ◽  
pp. 1032-1041 ◽  
Author(s):  
Thomas Kroneis ◽  
Jochen B Geigl ◽  
Amin El-Heliebi ◽  
Martina Auer ◽  
Peter Ulz ◽  
...  

BACKGROUND Analysis of chromosomal aberrations or single-gene disorders from rare fetal cells circulating in the blood of pregnant women requires verification of the cells' genomic identity. We have developed a method enabling multiple analyses at the single-cell level that combines verification of the genomic identity of microchimeric cells with molecular genetic and cytogenetic diagnosis. METHODS We used a model system of peripheral blood mononuclear cells spiked with a colon adenocarcinoma cell line and immunofluorescence staining for cytokeratin in combination with DNA staining with the nuclear dye TO-PRO-3 in a preliminary study to define candidate microchimeric (tumor) cells in Cytospin preparations. After laser microdissection, we performed low-volume on-chip isothermal whole-genome amplification (iWGA) of single and pooled cells. RESULTS DNA fingerprint analysis of iWGA aliquots permitted successful identification of all analyzed candidate microchimeric cell preparations (6 samples of pooled cells, 7 samples of single cells). Sequencing of 3 single-nucleotide polymorphisms was successful at the single-cell level for 20 of 32 allelic loci. Metaphase comparative genomic hybridization (mCGH) with iWGA products of single cells showed the gains and losses known to be present in the genomic DNA of the target cells. CONCLUSIONS This method may be instrumental in cell-based noninvasive prenatal diagnosis. Furthermore, the possibility to perform mCGH with amplified DNA from single cells offers a perspective for the analysis of nonmicrochimeric rare cells exhibiting genomic alterations, such as circulating tumor cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4493-4493 ◽  
Author(s):  
Yoshihiro Hatta ◽  
Minoru Saiki ◽  
Yuko Enomoto ◽  
Shin Aizawa ◽  
Umihiko Sawada ◽  
...  

Abstract Troglitazone and pioglitazone are one of thiazolidinediones that are high affinity ligand for the nuclear receptor called peroxisome proliferator-activated receptor gamma (PPAR-γ). Troglitazone is a potent inhibitor of clonogenic growth of acute myeloid leukemia cells when combined with a retinoid. However, the effect of pioglitazone to neoplastic cells and normal hematopoietic cells has not been studied yet. Adult T-cell leukemia (ATL), prevalent in western Japan, is a highly aggressive malignancy of mature T lymphocyte. Therefore, we studied antitumor effect of pioglitazone against leukemic cells including ATL as well as normal hematopoietic cells. With 300 μM of pioglitazone, colony formation of ATL cell lines (MT1, MT2, F6T, OKM3T, and Su9T01) was completely inhibited. Colony formation of HUT102, another ATL cell line, was 12 % compared to untreated control. Clonogenic cells of other leukemic cell lines (K562, HL60, U937, HEL, CEM, and NALM1) was also inhibited to 0–30% of control. Colony formation of primary leukemic cells from 5 AML patients was decreased to 15 %. However, normal hematopoietic cells were weakly inhibited with 300 μM pioglitazone; 77 % of CFU-GM, 70 % of CFU-E, and 33 % of BFU-E survived. Cell cycle analysis showed that pioglitazone decreased the ratio of G2/M phase in HL60 cells, suggesting the inhibition of cell division. By Western blotting, PPAR-γ protein level was similar in all leukemic cells and normal bone marrow mononuclear cells. Taken together, pioglitazone effectively eliminate leukemic cells and could be used as an antitumor agent in vivo.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3764-3764
Author(s):  
Monica L. Guzman ◽  
Shama Nasim ◽  
Marlene Balys ◽  
Cheryl A Corbett ◽  
Peter A Crooks ◽  
...  

Abstract Abstract 3764 Poster Board III-700 We have recently described the novel and unique anti-leukemic properties of 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8). Indeed, TDZD-8 was shown to eradicate leukemia at the bulk, stem, and progenitor level with rapid kinetics (typically < 2 h) with minimal toxicity to normal hematopoietic cells. Moreover, the cytotoxic activity of this drug is observed only in hematologic malignancies. The precise mechanism of TDZD-8 is not fully appreciated, but the compound has been shown to inhibit NFkappaB, GSK3beta, protein kinase C, FMS-like tyrosine kinase (Flt3), AKT and KDR. In addition, TDZD-8 rapidly depletes free thiols and appears to disrupt membrane integrity. Interestingly, TDZD-8 results in rapid mitochondrial swelling, followed by vacuole formation and accumulation. Despite possessing potent and specific anti-leukemia activity, the clinical utility of TDZD-8 is limited by the need for high concentrations (20 microM) and poor solubility. Therefore, to improve the pharmacological properties of TDZD-8, we initiated efforts to generate derivatives with greater anti-leukemia activity at lower concentrations and with greater water-solubility. To this end, an extensive structure-activity relationship study was carried out to examine the effects on anti-leukemic activity resulting from introduction of substituents at the C-2 and C-4- positions of the thiadiazolidine ring of TDZD-8, and of replacement of the TDZD ring with isosteric scaffolds. In total, we have screened more than 60 new analogs. Preliminary screens were performed using two different AML cell lines, evaluating the LD50 for each of the analogs relative to the parental compound. We found that introducing substitutuents into the main TDZD ring resulted in loss of anti-leukemic activity of the compounds. Moreover, substitution in the benzyl/phenyl ring does not affect anti-leukemia activity of the TDZD analogs. Importantly, we found that N-2 halogenoethyl analogs, exhibit exceptional activity against leukemic cells. Of the halogenoethyl analogs evaluated, the iodoethyl analog TD-361 was the most active compound with an LD50 of 0.49 microM in MV-411 cells. Compounds exhibiting increased anti-leukemia activity were subjected to further testing in phenotypically-defined AML stem/progenitor cells. All analogs demonstrated efficacy in primary AML cells at lower concentrations than TDZD-8. Moreover, these analogs still maintained the rapid kinetics observed with TDZD-8. Finally, we performed colony assays to determine the effect of new analogs on progenitor cells from normal and leukemic cells. These studies demonstrated that the more active TDZD analogs retained the selective ability of TDZD-8 to abate AML progenitor cells without harming normal hematopoietic cells. In summary, we have identified the critical chemical moieties for the observed activity of TDZD-8, and have also discovered analogs with improved anti-leukemia activity. Going forward, the most active derivatives are being optimized for water-solubility and will then be evaluated using primary human AML specimens engrafted into immune deficient mice. Based on evidence to date, we propose that the TDZD family of compounds may represent a new class of drugs for the treatment of leukemia and related hematologic malignancies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4909-4909 ◽  
Author(s):  
Annalisa Chiarenza ◽  
Nunziatina Parrinello ◽  
Piera La Cava ◽  
Eleonora Spina ◽  
Daniele Tibullo ◽  
...  

Abstract Abstract 4909 LENALIDOMIDE IS ABLE TO RESTORE IMMUNE SYSTEM IN MULTIPLE MYELOMA PATIENTS Annalisa Chiarenza, Nunziatina Parrinello, Piera La Cava, Eleonora Spina, Daniele Tibullo, Cesarina Giallongo, Maide Cavalli, Alessandra Romano, Paolo Fiumara, Giuseppe A. Palumbo, Francesco Di Raimondo Background Multiple myeloma (MM) is a malignant plasma-cell proliferative disorder associated with dysfunctional T-cell responses. The immunomodulatory Thal derivative (IMiD) CC-5013 (lenalidomide) appears to be a promising agent for the treatment of myeloma. Although the exact antitumor mechanism of action of lenalidomide is unknown, a number of mechanisms are postulated to be responsible for it's activity (inhibition of angiogenesis, direct antiproliferative and proapoptotic effects on MM cells, suppression of pro-inflammatory cytokines, modulation of myeloma-stromal cells adhesive interactions). In addition, it has been demonstrated that lenalidomide in vitro is able to enhance T cell proliferation and to promotes ADCC. In this study we evaluated if MM patients have a deficit of T-reg (CD4+, CD25+, and FOXP3+) and of T lymphocytes bearing CD200 (a tolerogenic molecule) and the effect of lenalidomide treatment on these parameters. In addition, we investigated whether lenalidomide could improve ex vivo the ADCC against myeloma cells. Materials and methods Eight patients with previously untreated MM (median age 56 years) were treated with lenalidomide plus dexamethasone as first line therapy. Lenalidomide was given orally 25 mg daily on days 1 to 21 of a 28-day cycle. Dexamethasone was given orally 40 mg daily on days 1, 8, 15, 22 of each cycle. All patients were evaluable for response and toxicity. Peripheral blood mononuclear cells (PBMNc) were obtained from MM patients using density gradient centrifugation (Fycoll) under sterile condictions, at the beginning of treatment and after 4 cycles of therapy. The percentage of T-reg (CD4+CD25+FOXP3+) and the expression of CD200 on T- lymphocytes were evaluated by cytometry. Twelve healthy subjects were used as control. Moreover, PBMNc (effector cells, E) were incubated with MM cells line ARH-77 (target cells, T), previously labelled with CFDA,SE (carboxyfluorescein diacetate, succinimidyl ester) as a tracing fluorescent marker, in culture medium (RPMI-1640, 10%FCS, 1%penicillin/streptomycin) at different concentration (T/E ratio 1:20, 1:40). After 18-24 h co-colture cells were analyzed by flow cytometry and MM plasma cells cytotoxicity was calculated as the percentage of positive CFDA,SE/propidium cells. Myeloma cell viability was determined by tripan blue esclusion and apoptosis was also evaluated using Annexin V/propidium assay. Two MM patients treated in first line with a combination of Velcade, Thalidomide and Dexamethasone (VTD) were used as control and the experiments were performed in duplicate. Results MM patients have a significantly lower rate of CD4+/CD25+/FOXP3+ and CD200+/CD3+ than normal (28,3±14,9/mmc and 37,8±24,7 /mmc vs 79,3±27,8 and 79,5± 48,9)(p=0,0001 and p=0,01 respectively). In our study, lenalidomide treatment resulted in an increase both of Treg cells and T-lymphocytes espressing CD200. This improvement is not statistically significant probably due to the low number of patients examined (tab I). More important, we observed that PBMC derived from patients treated with lenalidomide showed an increase ability to kill a target MM cell line compared to PBMC collected at diagnosis (CFDA,SE/propidium cells 11% vs 68%). This effect was more prominent in patients treated with lenalidomide than in MM patients treated with VTD (CFDA,SE/propidium cells 12% vs 39%), Fig.1. Conclusions Our data emphasize the role of lenalidomide in modulating the endogenous tumor-specific immune response and underline the anti-myeloma activity of these new class of drugs. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3032-3032
Author(s):  
Helena Dhamko ◽  
Gabrielle Melanie Siegers ◽  
Julia Schüler ◽  
Armand Keating

Abstract Abstract 3032 Poster Board II-1008 Gamma delta T cells (GDTCs), a small subset of T-lymphocytes (<10%) involved in tumor immune surveillance, are promising candidates for adoptive immunotherapy demonstrated by their ability to elicit cytolytic responses against many tumors. We have isolated and expanded GDTCs as a first step in developing a clinical protocol (Siegers, GM et al., ASH 2008). GDTCs exist in subsets whose specificity and function are determined by receptor rearrangement and tissue localization. The Vdelta2 (Vd2) subset in blood recognizes small phosphate containing non-peptide antigens and has been shown to kill myeloma and Burkitt lymphoma cells, whereas Vdelta1 (Vd1) GDTCs are typically found in tissue mucosae and provide defense against epithelial cancers. Although circulating GDTCs are predominantly of the Vdelta2 (Vd2) subset, we found that in 59% of GDTC cultures derived from the peripheral blood of healthy donors (n=17), the Vdelta1 (Vd1) subset was preferentially expanded, comprising 70.5% ± 14.7% (mean ± standard deviation) as determined by flow cytometry. In the remaining cultures, Vd2 GDTCs comprised 75.9 ± 14.2%. Preferential expansion of Vd1 did not correlate with a higher percentage of this subset in donor blood prior to GDTC isolation. In one expanded culture, Vd1 and Vd2 were equally present (40.3% and 41.3% respectively, on day 17). To determine activation status of Vd1 and Vd2 subsets simultaneously when co-incubated for 3 hours at a 1:5 effector:target ratio (E:T) with EM2eGFPluc, Ph(+) leukemic target cells, exposure of the degranulation-induced marker CD107 was determined by flow cytometry. Assays performed on culture days 10 to 17 (n=8) revealed that only 3.4 ± 2.7% Vd1 cells were activated, whereas Vd2 cells exhibited ten-fold activation with 34.1 ± 4.7% expressing CD107. To further investigate the different cytotoxic potential of these GDTC subsets, we generated 3 Vd2 clones from Donor 1 and 7 clones (3 Vd1 and 4 Vd2) from Donor 2. 3 clones were obtained from 200 Vd1-sorted cells, and 4 clones from 600 Vd2-sorted cells, suggesting superior clonogenicity of Vd1. Indeed, Vd1 clones grew faster than Vd2 from this donor. After 40 days in culture, we obtained 57 ± 37 × 106 Vd1 and 37 ± 23 × 106 Vd2 cells from a single cell on day 0. The enhanced growth of Vd1 explains how this subset predominates in most polyclonal GDTC cultures, despite donors having more Vd2 than Vd1 in their blood (Vd2:Vd1 = 5.7±3.2, n=7). Polyclonal expansion of GDTCs from Donor 2 yielded 11.2 × 106 cells on day 20, from 1.7 × 106 on day 0, a 6.7-fold expansion compared to 107-fold achieved with clones from the same donor. Vd2 clones were screened for their ability to lyse EM2eGFPluc in vitro. In a flow-cytometric assay based on propidium iodide staining, Vd2 clones exhibited cytotoxicities ranging 4.5%-10.6% for a 4-hour co-incubation at 2.6:1 E:T. Clones from Donor 1 were tested again and ranking confirmed in a 4-hour cytotoxicity assay at 10:1 E:T, with a range of 23.5%-35.4% for clones A1, B3 and C6, respectively. When C6 was compared to polyclonal GDTCs from the same donor, it was found to be more cytotoxic (9.0% versus 2.0% at 10:1 for 4 hours). Vd2 clones and polyclonal GDTC from Donor 2 were compared; clone E5 exhibited 10-fold (49.2%) and E3 1.4-fold (7.6%) cytotoxicity of polyclonal GDTCs (5.3%). Published reports describe an increase in Vd1 in B-CLL patients, hence we used MEC1, an EBV-positive B-cell line derived from B-CLL, as a target. At a 1.9:1 ratio over 4 hours, % cytotoxicity ranged 7.0% - 13.8% (D3 most cytotoxic). Vd1 clones were compared with polyclonal GDTC cultures derived from Donors 2 and 3, which exhibited 57% and 52% Vd1, respectively. Clone D3 again proved most cytotoxic at 10:1 E:T over 4 hours, with 40.8% compared to 18.6% (Donor 3) and 6.8% (Donor 2). Immunophenotyping indicates phenotypic stability in clones over time that is not evident in polyclonal populations. We conclude that the increased cytotoxicity, superior expansion potential and extended culture duration as well as phenotypic stability of GDTC clones make them a more attractive therapeutic agent than polyclonal cultures for the treatment of hematological malignancies. Our study reveals the potential importance of selecting specific and potent GDT effector cells for treating Ph(+) and B-CLL leukemias with GDTCs. We next plan to test this approach in our established pre-clinical xenogeneic leukemia mouse model. (Dhamko H was the recipient of an ASH Summer Trainee Research Award). Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document