Plasmacytoid dendritic cells (pDC) are the main producers of a key T-cell–stimulatory cytokine, IFNα, and critical regulators of antiviral immunity. Chronic myeloid leukemia (CML) is caused by BCR-ABL, which is an oncogenic tyrosine kinase that can be effectively inhibited with ABL-selective tyrosine kinase inhibitors (TKI). BCR-ABL–induced suppression of the transcription factor interferon regulatory factor 8 was previously proposed to block pDC development and compromise immune surveillance in CML. Here, we demonstrate that pDCs in newly diagnosed CML (CML-pDC) develop quantitatively normal and are frequently positive for the costimulatory antigen CD86. They originate from low-level BCR-ABL–expressing precursors. CML-pDCs also retain their competence to maturate and to secrete IFN. RNA sequencing reveals a strong inflammatory gene expression signature in CML-pDCs. Patients with high CML-pDC counts at diagnosis achieve inferior rates of deep molecular remission (MR) under nilotinib, unless nilotinib therapy is combined with IFN, which strongly suppresses circulating pDC counts. Although most pDCs are BCR-ABL–negative in MR, a substantial proportion of BCR-ABL+ CML-pDCs persists under TKI treatment. This could be of relevance, because CML-pDCs elicit CD8+ T cells, which protect wild-type mice from CML. Together, pDCs are identified as novel functional DC population in CML, regulating antileukemic immunity and treatment outcome in CML.

Significance: CML-pDC originates from low-level BCR-ABL expressing stem cells into a functional immunogenic DC-population regulating antileukemic immunity and treatment outcome in CML. Cancer Res; 78(21); 6223–34. ©2018 AACR.

BCR-ABL1 is a fusion oncogene, which arises from the t(9;22)(q34;q11) chromosomal translocation in a hematopoietic stem cell (1, 2). Chronic myeloid leukemia (CML) seems to be sensitive to immune therapy, such as with IFNα, which stimulates CML-specific T-cell responses (3, 4). Mounting of a specific T-cell response requires, first, recognition through the T-cell receptor of peptide–MHC class I complexes on the surface of antigen-presenting dendritic cells (DC). However, there is conflicting evidence regarding the capability of BCR-ABL–expressing leukemic CML stem cell–derived DCs (5–9) to trigger CML-specific T-cell immunity.

Little is known about the origin, regulation, and function of a specialized type of DCs, plasmacytoid DCs (pDC) in CML. Normal pDCs are the main source of a key Th1 immune-stimulatory cytokine, IFN (10–12). pDCs regulate innate and adaptive immune responses (12, 13) and contribute to immune activation (13), but also tolerance (14, 15). Untreated patients with CML were reported to have reduced pDC counts (16–18), which might involve BCR-ABL–mediated suppression of interferon regulatory factor 8 (IRF8, ICSBP) gene expression (19–23), a supposed pDC-fate–defining transcription factor (24, 25). Although BCR-ABL–induced Irf8 suppression was linked to impaired pDC development based on a murine model of CML (20), recent genetic in vivo evidence demonstrated that pDC development is Irf8-independent (26), essentially requiring only E2-2 (Tcf-4) and Zeb-2 (27, 28). However, Irf8 is clearly of key importance for the function of pDCs—the most prominent being IFN production (24–26). In a translational study of the multicenter tyrosine kinase inhibitor (TKI) discontinuation trial, EUROSKI, we recently observed unusual high counts of mature (CD86+) pDC in patients with CML in deep molecular remission (MR). Their abundance was associated with CD8+ T-cell exhaustion and predictive of a significantly lower chance of sustaining treatment-free remission (TFR) after TKI stop (29).

Here, we characterized the origin, genetic, and biological characteristics of pDCs in untreated CML (CML-pDCs) and corroborated specific aspects of CML-pDC development and function using a CML mouse model. Furthermore, we prospectively investigated qualitative and quantitative pDC changes under nilotinib versus nilotinib plus IFN therapy within a large multicenter CML trial. We find that CML-pDCs emerge from low-level BCR-ABL+ stem cells, produce inflammatory cytokines, and retain important functional properties, such as IFN secretion, maturation, and elicitation of CML-specific immunity. Data support for the first time that CML-pDCs are a source of inflammation in CML and likely involved in the regulation of treatment response and control CML-specific immunity.

Mice and cell lines

WT C57BL/6J mice, male at 6 to 10 weeks of age and female mice at 12 to 14 weeks of age, were used. Animal experiments were performed according to German law and approved by the regional board “Regierungspräsidium Giessen” (animal proposal # V54-19c20-15(1)MR20-36 Nr.07/2010 and # V54-19c2015h01MR Nr47/2014). 32D cell line was purchased from the “Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH” (DSMZ), and Phönix eco cell line was obtained from Allele Biotechnology. Cells were tested periodically and confirmed as Mycoplasma free by the PCR-based method (myco: 5′-gggacgaaacaggattagataccct-3′ and 5′-tgcaccatctgtcactccgttaacctc-3′), and authentication was not conducted, unless by morphology check due to microscope. Both cell lines were maintained according to the supplier's recommendations and not cultured more than 2 months.

Plasmids

The retroviral vectors pMIGp210 (encoding the BCR-ABLp210 and gfp), referred to as BCR-ABL, and pMIG (control, encoding only gfp) were a kind gift from Dr. W. Pear (The University of Pennsylvania, Philadelphia, PA; ref. 30). Using this vector as basis, the [BCR-ABL]-2A-[IRF8]-2A-[GFP] plasmid (BARF) was constructed, coexpressing BCR-ABL and Irf8 and gfp. Retroviral particles were generated with Phoenix eco cell line as described previously (31).

Generation of pDCs, immunization, and adoptive T-cell transfer

Bone marrow of wild-type mice was harvested, enriched for pDCs (pDC isolation kit II, Miltenyi), and spinoculated with either BCR-ABL– or BARF-containing retroviral particles. During spinoculation, pDCs were cultured in RPMI Medium 1640 (Gibco) and supplemented with 10% FCS and 1% penicillin/streptomycin, 20 mmol/L HEPES, 1 mmol/L sodiumpyruvat, 50 μmol/L mercaptoethanol (all Gibco), and 100 ng/mL FLT3-ligand (FLT3-L; Roche). GFP-positive pDCs were sorted and then i.v. injected into the tail vein of wild-type recipient mice (1,000–3,000 cells). pDC injections were repeated weekly in each mouse, for 3 times.

CD8+ T cells were enriched at a purity of up to 80% from spleens of pDC-immunized animals, and 2.5 × 106 of these were adoptively transferred i.p. the day after CML stem cell transplantation of independent recipient animals. To control for engraftment, blood samples from mice tail vain were analyzed weekly for proportions of GFP+ cells.

Western blotting

Western blotting of 32D cells and primary murine cells was performed as previously described. The following primary antibodies were used: anti-IRF8 goat, clone C-19 and anti-c-Abl mouse, clone 24-11 (both Santa Cruz Biotechnology) and anti-Actin mouse, clone AC-15 (Sigma-Aldrich). The secondary antibodies were horseradish peroxidase–conjugated polyclonal rabbit anti-goat and polyclonal goat anti-mouse antibodies (both Dako).

Primary CML blood samples

Peripheral blood samples of patients with primary diagnosis of CML or during follow-up were received through the immunologic substudy of the German multicenter CML-V study. In accordance to the Declaration of Helsinki, all patients gave written informed consent to participate in this substudy of CML-V or EUROSKI. We confirm that both studies were approved by the local Ethic Committees [NCT01657604 (CML-V) and NCT01596114 (EUROSKI)]. Primary blood mononuclear cells (PBMC) were isolated by Biocoll density gradient separation (density 1.077 g/mL, Biochrom).

Flow cytometry analysis and cell sorting

Flow cytometry was performed on the LSRII (BD Biosciences), and data were analyzed using the FlowJo software (FlowJo LLC). For fluorescence-activated cell sorts, cell populations were enriched prior to sorting using magnetic bead enrichment (Miltenyi). Sorting was performed using the MoFlo XDP (Beckman Coulter). For analysis of maturation status, the PBMCs were cultured for at least 18 hours with and without 2 μmol/L CpG 2006 in RPMI Medium 1640 (Gibco) and supplemented with 10% FCS and 1% penicillin/streptomycin, prior to antibody staining. Two four-color stainings were performed for each, untreated and treated cells, which included FcR blocking, BDCA-2 (PE), CD123 (FITC), and either HLA-DR (APC) + CD86 (TRI-COLOR) or CD80 (APC) + CD40 (TRI-COLOR). Due to very low cell numbers, isotype controls for APC- and TRI-COLOR stainings were performed on CpG-treated cells and used for both untreated and treated cultures. A list of used antibodies and enrichment kits is attached (Supplementary Tables S1 and S2). For details on flow cytometry (sample preparation, gating, and abundance of populations), see Supplementary Table S3.

Intracellular IFN staining

For intracellular staining of primary human pDCs, enriched cells (Supplementary Table S2) were cultured in 100 μL RPMI Medium 1640 (Gibco) and supplemented with 10% FCS and 1% penicillin/streptomycin, in 96-well U-bottom plates for 5 hours with and without 5 μmol/L CpG 2216 (TIB MOLBIOL). After 2-hour incubation, 1 μL Golgi-Plug (BD Biosciences) was added into each well. Then the cells were stained for surface markers and afterward fixed with 100 μL Medium A for 15 minutes at room temperature (dark). This was followed by permeabilization with 100 μL Medium B (both Thermo Fisher Scientific) and simultaneous staining with the intracellular IFNα antibody, FITC-conjugated (clone 225.C, Chromaprobe) for 20 minutes at 4°C. The cells were resuspended in 2% formaldehyde (Fischer) and analyzed on the LSRII.

CML transduction/transplantation model

Bone marrow of wild-type mice was harvested by flushing femura and tibiae. Mononuclear cells were obtained after density-gradient centrifugation (density 1.086 g/mL, Pancoll, PAN-Biotech). Stem cells for transplantation were enriched from bone marrow mononuclear cells using immunomagnetic column separation (lineage depletion) of lineage-positive cells (Lineage Cell Depletion Kit, Miltenyi) and then enrichment of Sca-1+ cells (easy sep mouse SCA1-positive selection kit, stem cell). Isolated stem cells had a purity of 85% to 95%, were precultured for 18 hours in IMDM (Gibco, Thermo Fisher Scientific), and supplemented with 20% FCS, 1% penicillin/streptomycin (both PAN Biotech), 50 μmol/L Mercaptoethanol (Gibco), 10 ng/mL recombinant murine IL3, 50 ng/mL recombinant IL6, and 50 ng/mL recombinant murine SCF (all Immunotools). Cells were transduced with either pMIG, BCR-ABL-, or BARF-retroviral particles by spinoculation (in the presence of 5 μg/mL polybrene) for 2 consecutive days. GFP positivity was assessed by flow cytometry. Note that 3 × 104 GFP+ cells were injected intravenously via tail vein into sublethally (7 Gy) irradiated female recipient mice. Animal experiments were performed according to German law and approved by the regional board “Regierungspräsidium Giessen” (animal proposal # V54-19c20-15(1)MR20-36 Nr.07/2010 and # V54-19c2015h01MR Nr47/2014). For details regarding animal experimentation (sample size calculation, data exclusion, replication, randomization, and blinding), see Supplementary Table S4.

BCR-ABL FISH

BCR-ABL FISH was performed as previously described (32).

Quantitative real-time PCR

Murine RNA was isolated and reverse transcribed using RNeasy Micro Kit and Omniscript RT Kit (both Qiagen) according to the manufacturer's protocol. RNA of human samples was isolated and transcribed using TriFast (peqlab) and SuperScript III reverse transcriptase (Thermo Fisher Scientific). Real-time quantitative PCR data for IRF8 gene expression were performed using the ΔΔCt method, with GAPDH as the housekeeping gene. Samples were analyzed in triplicates using QuantiTect SYBR Green Mix (Qiagen). The following primer pairs were used: mouse gapdh: 5′-catggccttccgtgttccta-3′ and 5′-cctgcttcaccaccttcttgat-3′; mouse irf8: 5′-tgccggcaagcaggattaca-3′ and 5′-ccacgtggctggttcagctt-3′; human gapdh: 5′-ctcctccacctttgacgctg-3′ and 5′-accaccctgttgctgtagcc-3′; human irf8: 5′-gtcccaactggacatttccg-3′ and 5′-cattcacgcagccagcag-3′. Quantitation of BCR-ABL levels in patients was performed following procedures and definitions of molecular response as previously described. Results were reported analogous to the European LeukemiaNet criteria as a normalized transcript number of the international scale, IS (33). Absolute BCR-ABL transcript numbers were assessed relative to GUS expression.

For competitive BCR-ABL and gfp PCR, we used genomic DNA of either spleen or chloroma tissue. For amplification, we used DreamTaq DNA Polymerase (Thermo Fisher Scientific), and the following 2 primer pairs: ba-1 5′-ttcagaagcttctccctggcatccgt-3′ and a-1 5′-ggtaccaggagtgtttctccagactg-3′, generating a 487 bp fragment, and gfp-for 5′-cgtaaacggccacaagttca-3′ and gfp-rev 5′-tcttgtagttgccgtcgtcc-3′, generating a 260 bp product.

Cytology

Cytological smears were stained by the panoptic method of Pappenheim and analyzed using the Olympus BH-2 microscope (Olympus), oil objective, ×100. Images were done with the DHS picture database software (Dietermann & Heuser Solution GmbH).

PCR amplification and sequencing of mutations

Next-generation sequencing of ASXL1 from genomic DNA of CD34+ enriched cells was performed as described previously (34). For conventional sequencing, gDNA from sorted pDCs and CD34+/CD38+ cells was preamplified with GoTaq DNA Polymerase (Promega) using the following primers: human ASXL1 mut2: 5′-cacttacaaaagaccagagcca-3′ and 5′-ctggatggagggagtcaaaa-3′. The resulting 237 bp DNA product was cleaned up by the NucleoSpin Gel and PCR Clean-up Kit (Machery and Nagel) and then sequenced with the human ASXL1 mut2 reverse primer by Seqlab. In a patient harboring the BCR-ABL F359I mutation, cDNA of FACS-sorted pDCs and PBMCs was used in PCR with Phusion Green High Fidelity DNA-Polymerase (Thermo Fisher Scientific). The primers of ABL-A, 5′-acagcattccgctgaccatcaataag-3′ and 5′-atggtccagaggatcgctctct-3′, amplified a 1,718 bp fragment. Using this fragment, a nested PCR was performed with the primers pair: human BCR-ABL F359I 5′-catgacctacgggaacctcc-3′ and 5′-ggccaaaatcagctaccttc-3′. A 200 bp PCR product was sequenced with the BCR-ABL F359I forward primer.

Sequences were analyzed for mutations using the BLAST Align software (www.ncbi.nlm.nih.gov/BLAST/) and are shown as electropherogram.

mRNA sequencing

Total RNAs were isolated from sorted pDCs of healthy donors and the first diagnostic patients with CML with RNeasy Micro Kit (Qiagen), according to the manufacturer's protocol. RNA libraries were generated with the NEBNext Ultra RNA Library Prep Kit for Illumina (New England Biolabs Inc.), according to the manufacturer's protocol. The libraries were sequenced on an Illumina HiSeq 2000, requested Read Length HiSeq 2 × 75 bp with paired end.

Statistical methods

Data were analyzed with GraphPad Prism 6 software (GraphPad Software, Inc.). Statistical significance of data was analyzed by the unpaired t test or ANOVA, after testing for normal distribution, unless indicated otherwise. Significance of survival difference was analyzed by the Mantel–Cox test. P value is shown as: ns, nonsignificant, P > 0.05; *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Software

Flow cytometry data were acquired with BD FACSDIVA software (Version 6.1.3; BD Biosciences) and analyzed by FlowJo software (either on windows version 7.6 or Mac version 9.5) from FlowJo LLC.

The Summit software (version 4.1; Cytomation) was used for acquiring cell sorts.

For Pappenheim image acquisition and processing, the DHS picture database software was used (Dietermann & Heuser Solution GmbH).

Statistical analysis was performed with GraphPad Prism 6 software (GraphPad Software, Inc.).

CML-pDCs are BCR-ABL–positive, express IRF8, and develop at normal frequency

Patients with untreated CML were previously reported to have strongly reduced pDC counts (16–18). However, here we show that patients with treatment-naïve CML only have lower pDC proportions in the peripheral blood (% pDC in white blood cells; Fig. 1A and B), whereas their absolute pDC numbers (pDCs per μL) are not reduced (Fig. 1C). Moreover, we found that CML-pDCs are BCR-ABL–positive (BCR-ABL+) by FISH (median: 81%; range: 57%–100%, n = 8; Fig. 1D). They also express normal IRF8 levels when compared with normal donor pDCs (Fig. 1E). Thus, human data do not show a link between BCR-ABL, IRF8 expression, and pDC counts in CML.

Figure 1.

CML-pDC: frequency, BCR-ABL–positive origin, and IRF8 expression. A, Representative staining of PBMCs for BDCA-2+/CD123+ pDCs in a patient with untreated chronic phase CML versus a normal donor. B, Percentage of pDCs in peripheral blood of normal donors versus patients with untreated CML within the CML-V study. C, Absolute pDCs counts (pDCs/μL) of cohorts as in B. D,BCR-ABL interphase FISH of patients with untreated CML. Top, representative FISH staining of pB CML-PBMC and CML-pDCs (yellow, BCR-ABL fusion signal); bottom, quantitation of BCR-ABL FISH-positive cells and pDCs frequency in peripheral blood (pB; analyzed cell nuclei: 20 to up to 154). E,IRF8 mRNA expression of sorted human BDCA-2+/CD123+ pDCs from normal donor (n = 3) and patients with untreated CML (n = 10). Asterisks indicate significance level, ****, P < 0.0001 or ns, nonsignificant, P > 0.05 according to Mann–Whitney test (B and C) or unpaired t test (E).

Figure 1.

CML-pDC: frequency, BCR-ABL–positive origin, and IRF8 expression. A, Representative staining of PBMCs for BDCA-2+/CD123+ pDCs in a patient with untreated chronic phase CML versus a normal donor. B, Percentage of pDCs in peripheral blood of normal donors versus patients with untreated CML within the CML-V study. C, Absolute pDCs counts (pDCs/μL) of cohorts as in B. D,BCR-ABL interphase FISH of patients with untreated CML. Top, representative FISH staining of pB CML-PBMC and CML-pDCs (yellow, BCR-ABL fusion signal); bottom, quantitation of BCR-ABL FISH-positive cells and pDCs frequency in peripheral blood (pB; analyzed cell nuclei: 20 to up to 154). E,IRF8 mRNA expression of sorted human BDCA-2+/CD123+ pDCs from normal donor (n = 3) and patients with untreated CML (n = 10). Asterisks indicate significance level, ****, P < 0.0001 or ns, nonsignificant, P > 0.05 according to Mann–Whitney test (B and C) or unpaired t test (E).

Close modal

CML-pDCs develop from low-level BCR-ABL–expressing stem cells

To study the origin of CML-pDCs and a previously proposed dependence of CML-pDC development on Irf8, we asked whether a block of CML-pDC transdifferentiation from leukemic stem cells is caused by BCR-ABL–mediated Irf8 suppression in vivo. The murine transduction/transplantation (tt)-CML model was employed (35). In the tt-CML mouse model, leukemogenesis depends on the engraftment of BCR-ABL–transformed LinSca1+c-Kit+ cells (LSK; refs. 36, 37). BCR-ABL+ hematopoiesis can be tracked using GFP expression as a marker. Irf8 was strongly reduced by BCR-ABL+ in CML stem cells (CML-LSK), common lymphoid (c-Kit+Sca-1+IL7-receptor alpha+GFP+; CLP), and myeloid progenitors (c-Kit+Sca-1CD16/32CD34+GFP+; CMP; Supplementary Fig. S1A), but, in contrast to patients with CML (Fig. 1C), CML mice showed a severe pDC loss (Supplementary Fig. S1B). Moreover, restoring IRF8 expression in BCR-ABL+ CML stem cells in vivo using a BCR-ABL/Irf8 coexpression construct (BARF; Fig. 2A) did not rescue CML-pDC maturation, suggesting that the CML-pDC maturation block is causally linked to BCR-ABL overexpression, but not IRF8 suppression (Fig. 2B). This was further supported by the finding that BCR-ABL caused an expansion of CML-LSK, T cells (CD3+), B cells (B220+), and myeloid cells (Gr1+), but selectively blocked maturation of pDCs irrespective of restored Irf8 levels (Fig. 2C). Of note, the few GFP+ pDCs, which transdifferentiated from GFP+ stem cells, consistently expressed much lower BCR-ABL levels (GFP) than the remaining leukemic bulk in the same animals (Fig. 2D). Likewise, human CML-pDCs expressed in median 3.5-fold lower BCR-ABL mRNA (range: 1.2–6.8) than the leukemic bulk (Fig. 2E). This supports that high BCR-ABL expression thresholds, not IRF8 loss, suppress CML-pDC maturation. We concluded that the reason of maintained CML-pDC maturation in human CML as opposed to CML mice is clonal BCR-ABL expression heterogeneity (31, 38, 39). CML-pDCs likely originate from stem cells with lower-level BCR-ABL expression, which is rare in tt-CML mice. Indeed, a heterogeneous evolutional trajectory of CML bulk cells and CML-pDCs was supported genetically in 2 patients with CML. One patient carried an ABL-resistance mutation, detectable only in the sorted CML bulk, but not in the sorted pDC fraction (Fig. 2F). The other patient had an ASXL1 mutation that was detectable in sorted CML-CD34+ cells, but not in CML-pDCs (Fig. 2G).

Figure 2.

pDCs show low-level BCR-ABL expression. A, Left, retroviral BARF-coexpression construct; middle, Western blotting of BARF-transduced 32D cl cell line; right, Western blotting of protein lysates from CMLBCR-ABL and CMLBARF mice (spleen cells). B, Left, absolute number of mPDCA-1+/B220+ pDCs developing from GFP+ hematopoiesis in transplanted mice for spleen; right, bone marrow. Note that one proportion of CMLBARF mice succumb on a myeloproliferative CML-like disease, referred to as CML-BARF(lethal), and another proportion survives (CML-BARF(surviving)). C, Extent of BCR-ABL–positive (GFP+) hematopoiesis in bone marrow in the primitive LSC fraction and distribution in various terminally differentiated compartments for CMLBARF (lethal), CMLBCR-ABL, and control mice. The indicated compartments were gated as LinSca1+c-Kit+ (LSK), GR-1+, CD3+, B220+, and mPDCA1+/B220+ (pDCs) and then analyzed for percentage of GFP+ cells. D, FACS histograms illustrating mean fluorescence intensity (MFI) of GFP (BCR-ABL) of BCR-ABL+ pDCs versus BCR-ABL+ total cells of six CML mice (n = 15). E, Fold reduction of absolute BCR-ABL transcript numbers measured by real-time PCR and expressed as a BCR-ABL/GUS ratio in sorted CML-pDCs compared with the denominator total white blood cells of the same patient, respectively. F, Left, FACS analysis BDCA-2+/CD123+ pDC frequency and BCR-ABLIS in pB of CML patient #059-505 at different time points of nilotinib treatment; right, Sanger sequencing electropherogram of cDNA from sorted pDCs and PBMC of a de novo emerging F359I nilotinib resistance mutation in BCR-ABL. G, Left, FACS-sorting strategy of patient #013-501; right, Sanger sequencing electropherogram of cDNA from sorted CD34 cells for an ASXL1 frame shift mutation and the absence of it in sorted pDCs fraction. Asterisks indicate significance level, *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; or ns, nonsignificant, P > 0.05 according to one-way ANOVA with Tukey multiple comparison test (B) or two-way ANOVA with Tukey multiple comparison test (C), unpaired t test (D), or paired t test (E).

Figure 2.

pDCs show low-level BCR-ABL expression. A, Left, retroviral BARF-coexpression construct; middle, Western blotting of BARF-transduced 32D cl cell line; right, Western blotting of protein lysates from CMLBCR-ABL and CMLBARF mice (spleen cells). B, Left, absolute number of mPDCA-1+/B220+ pDCs developing from GFP+ hematopoiesis in transplanted mice for spleen; right, bone marrow. Note that one proportion of CMLBARF mice succumb on a myeloproliferative CML-like disease, referred to as CML-BARF(lethal), and another proportion survives (CML-BARF(surviving)). C, Extent of BCR-ABL–positive (GFP+) hematopoiesis in bone marrow in the primitive LSC fraction and distribution in various terminally differentiated compartments for CMLBARF (lethal), CMLBCR-ABL, and control mice. The indicated compartments were gated as LinSca1+c-Kit+ (LSK), GR-1+, CD3+, B220+, and mPDCA1+/B220+ (pDCs) and then analyzed for percentage of GFP+ cells. D, FACS histograms illustrating mean fluorescence intensity (MFI) of GFP (BCR-ABL) of BCR-ABL+ pDCs versus BCR-ABL+ total cells of six CML mice (n = 15). E, Fold reduction of absolute BCR-ABL transcript numbers measured by real-time PCR and expressed as a BCR-ABL/GUS ratio in sorted CML-pDCs compared with the denominator total white blood cells of the same patient, respectively. F, Left, FACS analysis BDCA-2+/CD123+ pDC frequency and BCR-ABLIS in pB of CML patient #059-505 at different time points of nilotinib treatment; right, Sanger sequencing electropherogram of cDNA from sorted pDCs and PBMC of a de novo emerging F359I nilotinib resistance mutation in BCR-ABL. G, Left, FACS-sorting strategy of patient #013-501; right, Sanger sequencing electropherogram of cDNA from sorted CD34 cells for an ASXL1 frame shift mutation and the absence of it in sorted pDCs fraction. Asterisks indicate significance level, *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; or ns, nonsignificant, P > 0.05 according to one-way ANOVA with Tukey multiple comparison test (B) or two-way ANOVA with Tukey multiple comparison test (C), unpaired t test (D), or paired t test (E).

Close modal

CML-pDCs display constitutive inflammatory signaling

In order to obtain insights into the activation- and functional status of CML-pDCs, we compared the gene expression profiles of CML-pDCs and normal donor pDCs using RNA sequencing (Fig. 3A). Gene expression profiles of CML-pDCs (n = 6) differed significantly from normal pDCs (n = 3; Fig. 3B; Supplementary Table S5). With a median sequencing depth of 127.2 × 106 mapped reads (CML-pDC samples) and 154.1 × 106 mapped reads in normal pDC samples, 3,109 genes were found to be significantly regulated (log2FC ≥ 1) in CML-pDCs (Fig. 3B; Supplementary Data File S1). The majority of these differentially regulated genes (63.5%) were upregulated (Fig. 3C). Significantly more of these genes (20.5%) were strongly upregulated in CML-pDCs, whereas only a minority (5.8%) were strongly downregulated. Gene set enrichment analysis (40) showed enrichment in CML-pDCs of nine classes of genes involved in antigen capturing, processing, and presentation by MHC class I and II, as well as IFN, IL, and Toll-like receptor (TLR) signaling (Fig. 3D). The leading-edge subsets of genes in these nine classes were commonly overlapping between 2, 3, 4, or more classes, consistent with their critical importance (Fig. 3D; Supplementary Data File S2).

Figure 3.

Inflammatory signaling in CML-pDC. A, Sort strategy and Pappenheim staining of sorted human normal and untreated CML BDCA-2+/CD123+ pDCs for mRNA sequencing. B, Hierarchical clustering of the set of genes detected as significantly differentially expressed between CML-pDCs and normal donor pDCs. Hierarchical clustering parameters: Euclidean distance, average linkage. C, Pie chart illustrating extent of differential expression between CML-pDCs and normal donor pDCs. D, Top, leading edge analysis of the nine immune-related GSEA classes, which are highlighted in bold (bottom), reveals subsets of genes that contribute most to the enrichment result. Bottom, all significantly enriched immune-related GSEA classes in CML-pDCs versus normal donor pDCs.

Figure 3.

Inflammatory signaling in CML-pDC. A, Sort strategy and Pappenheim staining of sorted human normal and untreated CML BDCA-2+/CD123+ pDCs for mRNA sequencing. B, Hierarchical clustering of the set of genes detected as significantly differentially expressed between CML-pDCs and normal donor pDCs. Hierarchical clustering parameters: Euclidean distance, average linkage. C, Pie chart illustrating extent of differential expression between CML-pDCs and normal donor pDCs. D, Top, leading edge analysis of the nine immune-related GSEA classes, which are highlighted in bold (bottom), reveals subsets of genes that contribute most to the enrichment result. Bottom, all significantly enriched immune-related GSEA classes in CML-pDCs versus normal donor pDCs.

Close modal

CML-pDCs are functional, frequently CD86+, and overexpress inflammatory cytokines

IFN secretion is a key function of activated normal pDCs and Irf8-dependent (26, 27). mRNA sequencing shows that CML-pDCs constitutively express higher basal levels of IFNs than normal donor pDCs in an IRF8-dependent manner (Fig. 4A). IFN secretion is also promptly induced upon in vitro stimulation with the TLR9 ligand (oligonucleotide CpG 2216; Fig. 4B). In contrast, healthy donor-derived pDCs do not express or secrete IFN unless stimulated with CpG (Fig. 4A and B). In addition, also other cytokines, which are implemented in the elicitation of T-cell responses (e.g. ILs, CXCL9, 10, 11, CCL 22) or CML stem cell survival (TNFα, TGFβ; refs. 37, 39, 41), are overexpressed in CML-pDCs. TGFβ is strongly expressed also in normal pDCs (Fig. 4C). Intriguingly, although normal pDCs mature and then upregulate MHC class II and T-cell costimulatory molecules (CD80, CD86, and CD40) in response to CpG-induced TLR signaling (Fig. 4D, top), CML-pDCs are frequently spontaneously mature/activated (CD86+, CD40+) and thus resemble a recently described subpopulation of normal BDCA-2+CD123+ pDCs, so-called Axl+Siglec-6+ DCs (AS-DCs) or pre-DCs (Figs. 3A and 4D, bottom; Supplementary Fig. S2; refs. 42, 43) and display activated TNF, JAK, and NF-κB signaling (Supplementary Fig. S3), suggesting a constitutive autocrine stimulation of CML-pDCs by secretion of TNF, IFNs, and ILs. Only a minority of patients with de novo CML (exemplary shown pat. # 009-501) do not show spontaneous maturation and CD86, CD80, or CD40 expression (Supplementary Fig. S2). However, also immature CML-pDCs (pat. #009-501) retain their IFN secretion capacity and mature upon CpG treatment (Supplementary Fig. S2).

Figure 4.

CML-pDCs are functional and express high amounts of inflammatory cytokines. A, Heat map, based on RNA-seq data [transcripts per million (TPM)], illustrating the expression of IFNα subtypes in untreated CML-pDC and normal donor pDCs. Samples are ordered for IRF8 expression, and individual sample ID is mentioned at the bottom. B, Left and middle, representative FACS plots showing intracellular IFNα staining of MACS-enriched pDCs with and without TLR-9 engagement by in vitro CpG treatment. Exemplary shown are two normal donors (left) and patients with untreated CML (middle); right, statistical analysis of proportion of IFN-secreting pDCs. C, Heat maps, based on RNA-seq data (TPM), illustrating expression of different cytokines in untreated CML-pDC and normal donor pDCs. Individual sample ID is mentioned at the bottom. D, FACS histogram of activation/maturation markers of pDC (gated as CD123+/BDCA-2+) with and without CpG treatment for a normal donor (top plot) and a CML-patient # 001-509 (bottom plot). Shown are the indicated specific antibodies (gray curves) in comparison with isotype controls (black curves). Same isotype controls were used for untreated and CpG-treated cultures. Asterisks indicate significance level, *, P < 0.05 or **, P < 0.01 according to two-tailed unpaired t test (B).

Figure 4.

CML-pDCs are functional and express high amounts of inflammatory cytokines. A, Heat map, based on RNA-seq data [transcripts per million (TPM)], illustrating the expression of IFNα subtypes in untreated CML-pDC and normal donor pDCs. Samples are ordered for IRF8 expression, and individual sample ID is mentioned at the bottom. B, Left and middle, representative FACS plots showing intracellular IFNα staining of MACS-enriched pDCs with and without TLR-9 engagement by in vitro CpG treatment. Exemplary shown are two normal donors (left) and patients with untreated CML (middle); right, statistical analysis of proportion of IFN-secreting pDCs. C, Heat maps, based on RNA-seq data (TPM), illustrating expression of different cytokines in untreated CML-pDC and normal donor pDCs. Individual sample ID is mentioned at the bottom. D, FACS histogram of activation/maturation markers of pDC (gated as CD123+/BDCA-2+) with and without CpG treatment for a normal donor (top plot) and a CML-patient # 001-509 (bottom plot). Shown are the indicated specific antibodies (gray curves) in comparison with isotype controls (black curves). Same isotype controls were used for untreated and CpG-treated cultures. Asterisks indicate significance level, *, P < 0.05 or **, P < 0.01 according to two-tailed unpaired t test (B).

Close modal

ABL-TKI blocks CML-pDC development and suppresses pDC numbers in remission

In vivo treatment with the BCR-ABL–specific TKI nilotinib reduced CML-pDC counts within months of therapy (Fig. 5A). This suggests an antiproliferative effect of TKI on CML-pDC precursors. A more potent reduction in pDC counts occurred under cotreatment with nilotinib and IFN within the CML-V trial (Fig. 5A). This combined therapy also reduced the number of mature (CD86+) pDC more efficacious than nilotinib alone. This could be clinically important, because CD86+pDCs were previously linked to a lower TFR probability (29). Of note, there is a rapid decline of the CD86+pDC counts after 6 to 12 months under nilotinib and more pronounced with nilotinib plus IFN. CD86+pDC counts were comparably low in patients with long-term imatinib treatment (median, 7.5 years) within the EUROSKI TKI discontinuation trial (Fig. 5A and B). More than 10 CML-pDCs/μL at diagnosis—corresponding to the median number of pDC detected in MR (Fig. 5A)—were associated with a reduced probability to achieve a MR4 after 12 months of therapy with nilotinib (Fig. 5C). Interestingly, if patients with CML-pDC counts > 10/μL were randomized to receive nilotinib plus IFN, an inferior molecular response was no longer seen (Fig. 5C). PDCs from patients in MR were BCR-ABL–negative by FISH (n = 8/8), which enabled to analyze approximately only 100 pDCs per patient (Fig. 5D). However, when using absolute quantification and a standardized, international scale BCR-ABLIS PCR (sensitivity 5 × 10−5), a median of 7,700 BCR-ABL copies (range, 470–17,000) were detected in every patient. Thus, numbers of residual CML-pDCs remain detectable by PCR in all 18 analyzed patients—even in very deep MR of CML (Fig. 5E). This suggests that a significant proportion of BCR-ABL+ CML-pDCs is TKI-insensitive.

Figure 5.

Remission pDC counts are suppressed and impaired CML-pDC development. A, Absolute pDC counts (pDCs/μL) in peripheral blood of patients with untreated CML compared with nilotinib or nilotinib + PEG IFN treatment for 6 to 12 months within the CML-V study, or versus long-term imatinib-treated patients from EUROSKI study. Dotted line indicates mean level of absolute pDC in normal donors. B, Absolute CD86+pDCs counts (pDCs/μL) of cohorts as in A. Dotted line indicates mean level of absolute CD86+pDC in normal donors. C, Analysis for achieving a BCR-ABLIS of ≤ 0.01 % after 12 months of therapy according to their absolute pDC counts (pDC/μL) and their therapy-arm within CML-V study. White numbers, patients who achieved BCR-ABLIS of ≤ 0.01%; black numbers, total patients. D,BCR-ABL interphase FISH of remission pDCs from deep MR of CML. Top, shown are representative cell pictures; bottom, quantitation of BCR-ABL FISH-positive cells and pDCs' frequency in peripheral blood (pB; analyzed cell number: 100, except R56, 89 cells). E,BCR-ABL (BCR-ABL/ABL ratio) expression levels in pDCs of remission patient in comparison with peripheral blood from the same sample are shown. Asterisks indicate significance level, *, P < 0.05; **, P < 0.01; ****, P < 0.0001; or ns, nonsignificant, P > 0.05 according to Kruskal–Wallis test with Dunn multiple comparison (A and B) or Fisher exact test (C) or unpaired t test (E).

Figure 5.

Remission pDC counts are suppressed and impaired CML-pDC development. A, Absolute pDC counts (pDCs/μL) in peripheral blood of patients with untreated CML compared with nilotinib or nilotinib + PEG IFN treatment for 6 to 12 months within the CML-V study, or versus long-term imatinib-treated patients from EUROSKI study. Dotted line indicates mean level of absolute pDC in normal donors. B, Absolute CD86+pDCs counts (pDCs/μL) of cohorts as in A. Dotted line indicates mean level of absolute CD86+pDC in normal donors. C, Analysis for achieving a BCR-ABLIS of ≤ 0.01 % after 12 months of therapy according to their absolute pDC counts (pDC/μL) and their therapy-arm within CML-V study. White numbers, patients who achieved BCR-ABLIS of ≤ 0.01%; black numbers, total patients. D,BCR-ABL interphase FISH of remission pDCs from deep MR of CML. Top, shown are representative cell pictures; bottom, quantitation of BCR-ABL FISH-positive cells and pDCs' frequency in peripheral blood (pB; analyzed cell number: 100, except R56, 89 cells). E,BCR-ABL (BCR-ABL/ABL ratio) expression levels in pDCs of remission patient in comparison with peripheral blood from the same sample are shown. Asterisks indicate significance level, *, P < 0.05; **, P < 0.01; ****, P < 0.0001; or ns, nonsignificant, P > 0.05 according to Kruskal–Wallis test with Dunn multiple comparison (A and B) or Fisher exact test (C) or unpaired t test (E).

Close modal

BCR-ABL does not inhibit the in vivo T-cell priming function of pDCs

To experimentally assess the functional competence and biological relevance of persisting BCR-ABL+ pDCs in vivo, wild-type C57BL/6J mice were vaccinated with BCR-ABL+ pDCs or different types of control pDCs. Vaccination occurred intravenously weekly for 3 successive weeks using 1 to 3 × 103in vitro–generated control pDCs (pDCwt), BCR-ABL+ pDCs (pDCBCR-ABL), BARF+ pDCs (pDCBARF), or gfp+ Mig-control pDCs (pDCMig), respectively (Fig. 6A).

Figure 6.

CML-pDCs can induce protective T-cell immunity in CML. A, pDC immunization mouse model. pDC generation: in vitro generation of BCR-ABL+ pDCs by retroviral transduction of enriched pDCs with either BCR-ABL, BARF, or empty Mig-vector; pDC immunization: injection of sorted BARF+pDCs, BCR-ABL+ pDCs, untransduced pDCs, or Mig-vector–transduced pDCs into WT mice once weekly for three successive weeks; adoptive T-cell transfer: isolation and injection of CD8+ cytotoxic T cells from immunized mice into BCR-ABL–transplanted mice on day +1. B, Representative FACS plots of kinetics of GFP (BCR-ABL)–positive cell proportions in blood of CML mice after adoptive T-cell transfer from pDCBCR-ABL-, pDCBARF-, pDCwt-, or pDCMig-immunized mice at indicated time points. C, Summary of GFP-positivity kinetics in blood from surviving pDCBCR-ABL mice, surviving pDCBARF, moribund pDCwt mice, and surviving pDCMig over time. D, Survival curves of CML mice after adoptive T-cell transfer from pDCBCR-ABL-, pDCBARF-, pDCwt-, and pDCMig-immunized mice. P value was assessed by the Mantel–Cox test.

Figure 6.

CML-pDCs can induce protective T-cell immunity in CML. A, pDC immunization mouse model. pDC generation: in vitro generation of BCR-ABL+ pDCs by retroviral transduction of enriched pDCs with either BCR-ABL, BARF, or empty Mig-vector; pDC immunization: injection of sorted BARF+pDCs, BCR-ABL+ pDCs, untransduced pDCs, or Mig-vector–transduced pDCs into WT mice once weekly for three successive weeks; adoptive T-cell transfer: isolation and injection of CD8+ cytotoxic T cells from immunized mice into BCR-ABL–transplanted mice on day +1. B, Representative FACS plots of kinetics of GFP (BCR-ABL)–positive cell proportions in blood of CML mice after adoptive T-cell transfer from pDCBCR-ABL-, pDCBARF-, pDCwt-, or pDCMig-immunized mice at indicated time points. C, Summary of GFP-positivity kinetics in blood from surviving pDCBCR-ABL mice, surviving pDCBARF, moribund pDCwt mice, and surviving pDCMig over time. D, Survival curves of CML mice after adoptive T-cell transfer from pDCBCR-ABL-, pDCBARF-, pDCwt-, and pDCMig-immunized mice. P value was assessed by the Mantel–Cox test.

Close modal

CD8+ cytotoxic T cells from these vaccinated mice (TcBCR-ABL, TcBARF, Tcwt, and TcMig) were sorted and adoptively transferred into other recipients: tt-CML-mice (Fig. 6A). These mice succumbed from GFP+ leukemia, unless they had received TcBCR-ABL or TcBARF, which potently suppressed the outgrowth GFP+ leukemia (Fig. 6B–D). Of note, pDCMig, which do not express BCR-ABL, but only gfp, also primed a leukemia-protective T-cell response. Presumably, GFP is the target of this T-cell response, because leukemic relapse after TcMig transfer was gfp-negative and BCR-ABL–positive, suggesting an immune escape through loss of gfp expression. In contrast, mice relapsing despite TcBCR-ABL or TcBARF lymphocyte infusions were never gfp-negative (Supplementary Fig. S4). This would be in line with an immune escape, supporting that BCR-ABL+ pDCs could be functional regulators of CML immunity. However, it cannot be ruled out that GFP-derived neoantigens or BCR-ABL–dependent leukemia-associated antigens are also released from GFP+ dying cells and then cross-presented by the recipient's own professional DCs such as cDCs, but also pDCs.

BCR-ABL–induced IRF8 suppression has previously been causally linked to pDC loss, pDC dysfunction, and a lack of immune surveillance in CML (16–18, 20, 44). Our results do not support this concept. First, while acknowledging that BCR-ABL overexpression suppresses Irf8 in primitive murine hematopoiesis and abrogates pDC development from BCR-ABL+ LSCs in mice (Fig. 2B and C; Supplementary Fig. S1; refs. 20, 45), there is no causal link between IRF8 loss and pDC loss in vivo. Secondly, there is no pDC loss in human CML, although CML-pDCs are of BCR-ABL–positive origin (Figs. 1C and 5A).

Data rather support that modeling human CML-pDC development in tt-CML mice is flawed by BCR-ABL overexpression, which is a prerequisite for LSC transformation in mice and potently abrogates pDC transdifferentiation from LSC (Fig. 2D). In contrast, CML-stem cell evolution is characterized by clonal heterogeneity (38, 39, 46), including BCR-ABL expression variability (31). Data imply, therefore, that pDC development in human CML is maintained from low-level BCR-ABL–expressing precursors, which are not progenies of the CML LSC clone producing the bulk of leukemia cells (Fig. 2F and G). Here, we showed that in spite of their BCR-ABL+ origin, CML-pDCs retain important functional pDC-properties, namely, IFN secretion and maturation capacity. At the same time, CML-pDCs aberrantly express multiple inflammatory cytokines and chemokines. Autocrine constitutive activation of JAK-STAT signaling in CML-pDCs could be a consequence (Supplementary Fig. S3), possibly leading to their peculiar mature phenotype in steady state (Fig. 4D; Supplementary Fig. S2). Very recently, a subpopulation of BDCA-2+ CD123+Axl+Siglec-6+ pDCs (AS-DC) also referred to as pre-DCs have been identified in normal individuals. These cells are very potent CD4+ and CD8+ T-cell stimulators. This raises the intriguing possibility that BCR-ABL leads to an expansion of BDCA-2+CD123+ CML-pDCs, which are AS-DC-/pre–DC-like and hence, unlike BDCA-2+CD123+ conventional pDCs, T-cell stimulatory (42, 43).

Via paracrine effects, CML-pDC–derived TNF and tumor-derived growth factor (TGF) might also promote CML stem cell survival (47, 48), limit TKI response, and contribute to CML-stem cell persistence (39). Supporting this, we noticed that higher CML-pDC counts at diagnosis were associated with an inferior depth of nilotinib response. However, when patients with high CML-pDCs at diagnosis were treated with a nilotinib/IFN combination within the randomized German CML-V study, total pDCs including CD86+pDCs were strongly reduced, and an association between high CML-pDC counts at diagnosis and inferior depth of response was no longer seen. Based on our recent results showing that CD86+pDCs are associated with lower probability of TFR after TKI cessation (29), this implies that an IFN plus nilotinib-based induction therapy leads to a higher rate of TFR, which is the primary endpoint of the CML-V study (NCT01657604). To study this, it will be tested whether TFR risk is associated with the number of CD86+pDCs and exhausted T cells measured at diagnosis and prior to TKI stop.

Chronic inflammation has detrimental effects during tumorigenesis. It promotes tumor evolution and is immune suppressive. We showed here that low-level BCR-ABL–expressing CML stem cells might trigger chronic inflammation by transdifferentiation into CML-pDCs as a source for many inflammatory chemokines and cytokines. Indeed, there is precedence that persistent high counts of activated CD86+pDCs during chronic infection are immune suppressive. Mechanistically, pDCs can induce immune-suppressive regulatory T cells (15, 49) and attenuate natural killer– and T-cell functions (50).

In contrast, in the early (preleukemic) evolution of CML, low frequencies of BCR-ABL+ pDCs might be tumor-suppressive. When low numbers of BCR-ABL+ pDC were injected into wild-type mice, they acted as vaccines and induced antileukemic T cells, which indeed protected from leukemia in vivo, when adoptively transferred into CML mice. This suggests that CML-CD86+pDCs—like their normal AS-pDC counterparts—are capable of priming T-cell responses to endogenously expressed antigens (12, 51, 52). Because a significant proportion of CML-pDCs are TKI-insensitive and persist during TKI-induced MR (Fig. 7), we propose that they elicit anti-CML CD8+ T-cell responses in MR via exploiting a restored and less exhausted remission T-cell repertoire (53, 54). It will now be important to validate that BDCA-2+CD123+CD86+ CML-pDCs correspond to the CD86+AS-DC/pre-DC population (42, 43). Beyond the scope of CML, it is tempting to speculate that other oncogenic driver mutations, which lead to an increased stem cell fitness and stem cell expansion such as in benign clonal hematopoiesis (55) or myelodysplastic syndrome, cause the emergence of a higher CD86+AS-DCs frequency. This hypothesis can be tested. Myeloid driver mutation–expressing stem cells, via maturation into CD86+pDCs, might supposedly engage extrinsic tumor suppression before cell-intrinsic tumor-suppressive signaling cascades such as ARF/TP53 can sense oncogenic stress and become activated. Oncogenic stress-sensing tumor-suppressive pathways usually require high-level oncogenic signaling output (56–58), suggesting that CML-pDC–induced extrinsic tumor suppression can be effective long before intrinsic tumor suppression is engaged.

Figure 7.

Proposed roles of inflammatory CML-pDCs in immunity and treatment response of CML. Hematopoietic stem cells acquire the BCR-ABL translocation and initially express low-level BCR-ABL and thus retain the ability to transdifferentiate into CML-pDCs. These cells aberrantly express inflammatory chemokines, cytokines, and CML-stem cell–protective TNFα and TGFβ. Whereas limited numbers of inflammatory pDCs may trigger antileukemic immunity during the early phase of CML evolution and again in deep MR under TKI-based treatment, high CML-pDC counts (expanding with BCR-ABL+ stem cell mass) drive chronic inflammation, which stimulates CML-stem cell persistence and immune suppression.

Figure 7.

Proposed roles of inflammatory CML-pDCs in immunity and treatment response of CML. Hematopoietic stem cells acquire the BCR-ABL translocation and initially express low-level BCR-ABL and thus retain the ability to transdifferentiate into CML-pDCs. These cells aberrantly express inflammatory chemokines, cytokines, and CML-stem cell–protective TNFα and TGFβ. Whereas limited numbers of inflammatory pDCs may trigger antileukemic immunity during the early phase of CML evolution and again in deep MR under TKI-based treatment, high CML-pDC counts (expanding with BCR-ABL+ stem cell mass) drive chronic inflammation, which stimulates CML-stem cell persistence and immune suppression.

Close modal

Together, qualitative (pDC-activation and BCR-ABL-status) and quantitative (high versus low pDC counts) factors govern the outcome of pDC biology in CML, which can be tumor-suppressive or -promoting. Further studies are warranted to eventually exploit oncogene-expressing pDCs as leukemia vaccines.

S. Saussele reports receiving commercial research grant from Novartis and BMS, and has received honoraria from the speakers' bureau of Novartis, BMS, Incyte, Pfizer, and Roche. A. Hochhaus reports receiving commercial research grant from Novartis, BMS, Incyte, and Pfizer. No potential conflicts of interest were disclosed by the other authors.

Conception and design: S. Inselmann, Y. Wang, M. Huber, A. Hochhaus, A. Burchert

Development of methodology: S. Inselmann, Y. Wang, S. Liebler, T. Ernst, C. Brendel, D. Pavlinic, A. Burchert

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Inselmann, S. Saussele, L. Fritz, C. Schütz, M. Huber, T. Ernst, S. Botschek, C. Brendel, D. Pavlinic, V. Benes, A. Hochhaus, A. Burchert

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Inselmann, Y. Wang, S. Saussele, L. Fritz, S. Botschek, R.A. Calogero, E.T. Liu, A. Neubauer, A. Hochhaus, A. Burchert

Writing, review, and/or revision of the manuscript: S. Inselmann, S. Saussele, E.T. Liu, A. Neubauer, A. Hochhaus, A. Burchert

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Wang, D. Cai, C. Brendel, V. Benes, A. Neubauer, A. Burchert

Study supervision: A. Hochhaus, A. Burchert

We thank C. Haferlach (MLL Munich) and D. Haase (University Hospital Göttingen) for performing BCR-ABL FISH. We thank C. Fabisch for coordinating the CML-V study. We thank Prof. Dr. Zink and Prof. Dr. Engenhart-Cabillic for help in performing animal radiation. We thank S. Hühn for excellent help with patient sample preparation, cell sorting, and mice work. We thank S. Sopper (University Innsbruck) and D. Wolf (University Bonn) for helpful discussions.

This work was supported by the “Deutsche Forschungsgemeinschaft “(DFG), Klinische Forschergruppe 210 “Genetics of Drug resistance in Cancer”, and the Deutsche José Carreras Leukämiestiftung (AR12/12) and the “Anneliese Pohl Stiftung.” The CML Study V has been supported by Novartis Oncology and MSD Sharp & Dohme GmbH. Scientific subprojects related to this study have also been supported by EUTOS.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Rowley
JD
. 
Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining
.
Nature
1973
;
243
:
290
3
.
2.
Ben-Neriah
Y
,
Daley
GQ
,
Mes-Masson
AM
,
Witte
ON
,
Baltimore
D
. 
The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene
.
Science
1986
;
233
:
212
4
.
3.
Molldrem
JJ
,
Lee
PP
,
Wang
C
,
Felio
K
,
Kantarjian
HM
,
Champlin
RE
, et al
Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia
.
Nat Med
2000
;
6
:
1018
23
.
4.
Burchert
A
,
Wölfl
S
,
Schmidt
M
,
Brendel
C
,
Denecke
B
,
Cai
D
, et al
Interferon-alpha, but not the ABL-kinase inhibitor imatinib (STI571), induces expression of myeloblastin and a specific T-cell response in chronic myeloid leukemia
.
Blood
2003
;
101
:
259
64
.
5.
Eibl
B
,
Ebner
S
,
Duba
C
,
Böck
G
,
Romani
N
,
Erdel
M
, et al
Dendritic cells generated from blood precursors of chronic myelogenous leukemia patients carry the Philadelphia translocation and can induce a CML-specific primary cytotoxic T-cell response
.
Genes Chromosomes Cancer
1997
;
20
:
215
23
.
6.
Heinzinger
M
,
Waller
CF
,
den Berg von
A
,
Rosenstiel
A
,
Lange
W
. 
Generation of dendritic cells from patients with chronic myelogenous leukemia
.
Ann Hematol
1999
;
78
:
181
6
.
7.
Mumprecht
S
,
Claus
C
,
Schürch
C
,
Pavelic
V
,
Matter
MS
,
Ochsenbein
AF
. 
Defective homing and impaired induction of cytotoxic T cells by BCR/ABL-expressing dendritic cells
.
Blood
2009
;
113
:
4681
9
.
8.
Eisendle
K
,
Lang
A
,
Eibl
B
,
Nachbaur
D
,
Glassl
H
,
Fiegl
M
, et al
Phenotypic and functional deficiencies of leukaemic dendritic cells from patients with chronic myeloid leukaemia
.
Br J Haematol
2003
;
120
:
63
73
.
9.
Dong
R
,
Cwynarski
K
,
Entwistle
A
,
Marelli-Berg
F
,
Dazzi
F
,
Simpson
E
, et al
Dendritic cells from CML patients have altered actin organization, reduced antigen processing, and impaired migration
.
Blood
2003
;
101
:
3560
7
.
10.
Siegal
FP
,
Kadowaki
N
,
Shodell
M
,
Fitzgerald-Bocarsly
PA
,
Shah
K
,
Ho
S
, et al
The nature of the principal type 1 interferon-producing cells in human blood
.
Science
1999
;
284
:
1835
7
.
11.
Cella
M
,
Jarrossay
D
,
Facchetti
F
,
Alebardi
O
,
Nakajima
H
,
Lanzavecchia
A
, et al
Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon
.
Nat Med
1999
;
5
:
919
23
.
12.
Swiecki
M
,
Colonna
M
. 
The multifaceted biology of plasmacytoid dendritic cells
.
Nat Rev Immunol
2015
;
15
:
471
85
.
13.
Reizis
B
,
Colonna
M
,
Trinchieri
G
,
Barrat
F
,
Gilliet
M
. 
Plasmacytoid dendritic cells: one-trick ponies or workhorses of the immune system?
Nat Rev Immunol
2011
;
11
:
558
65
.
14.
Hadeiba
H
,
Lahl
K
,
Edalati
A
,
Oderup
C
,
Habtezion
A
,
Pachynski
R
, et al
Plasmacytoid dendritic cells transport peripheral antigens to the thymus to promote central tolerance
.
Immunity
2012
;
36
:
438
50
.
15.
Ochando
JC
,
Homma
C
,
Yang
Y
,
Hidalgo
A
,
Garin
A
,
Tacke
F
, et al
Alloantigen-presenting plasmacytoid dendritic cells mediate tolerance to vascularized grafts
.
Nat Immunol
2006
;
7
:
652
62
.
16.
Mohty
M
,
Isnardon
D
,
Vey
N
,
Brière
F
,
Blaise
D
,
Olive
D
, et al
Low blood dendritic cells in chronic myeloid leukaemia patients correlates with loss of CD34+/CD38– primitive haematopoietic progenitors
.
Br J Haematol
2002
;
119
:
1
4
.
17.
Mohty
M
,
Jourdan
E
,
Mami
NB
,
Vey
N
,
Damaj
G
,
Blaise
D
, et al
Imatinib and plasmacytoid dendritic cell function in patients with chronic myeloid leukemia
.
Blood
2004
;
103
:
4666
8
.
18.
Boissel
N
,
Rousselot
P
,
Raffoux
E
,
Cayuela
J-M
,
Maarek
O
,
Charron
D
, et al
Defective blood dendritic cells in chronic myeloid leukemia correlate with high plasmatic VEGF and are not normalized by imatinib mesylate
.
Leukemia
2004
;
18
:
1656
61
.
19.
Hao
S
,
Ren
R
. 
Expression of interferon consensus sequence binding protein (ICSBP) is downregulated in Bcr-Abl-induced murine chronic myelogenous leukemia-like disease, and forced coexpression of ICSBP inhibits Bcr-Abl-induced myeloproliferative disorder
.
Mol Cell Biol
2000
;
20
:
1149
61
.
20.
Watanabe
T
,
Hotta
C
,
Koizumi
S-I
,
Miyashita
K
,
Nakabayashi
J
,
Kurotaki
D
, et al
The transcription factor IRF8 counteracts BCR-ABL to rescue dendritic cell development in chronic myelogenous leukemia
.
Cancer Res
2013
;
73
:
6642
53
.
21.
Gerber
JM
,
Gucwa
JL
,
Esopi
D
,
Gurel
M
,
Haffner
MC
,
Vala
M
, et al
Genome-wide comparison of the transcriptomes of highly enriched normal and chronic myeloid leukemia stem and progenitor cell populations
.
Oncotarget
2013
;
4
:
715
28
.
22.
Diaz-Blanco
E
,
Bruns
I
,
Neumann
F
,
Fischer
JC
,
Graef
T
,
Rosskopf
M
, et al
Molecular signature of CD34(+) hematopoietic stem and progenitor cells of patients with CML in chronic phase
.
Leukemia
2007
;
21
:
494
504
.
23.
Schmidt
M
,
Nagel
S
,
Proba
J
,
Thiede
C
,
Ritter
M
,
Waring
JF
, et al
Lack of interferon consensus sequence binding protein (ICSBP) transcripts in human myeloid leukemias
.
Blood
1998
;
91
:
22
9
.
24.
Schiavoni
G
,
Mattei
F
,
Sestili
P
,
Borghi
P
,
Venditti
M
,
Morse
HC
, et al
ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8alpha(+) dendritic cells
.
J Exp Med
2002
;
196
:
1415
25
.
25.
Tsujimura
H
,
Tamura
T
,
Ozato
K
. 
Cutting edge: IFN consensus sequence binding protein/IFN regulatory factor 8 drives the development of type I IFN-producing plasmacytoid dendritic cells
.
J Immunol
2003
;
170
:
1131
5
.
26.
Sichien
D
,
Scott
CL
,
Martens
L
,
Vanderkerken
M
,
Van Gassen
S
,
Plantinga
M
, et al
IRF8 transcription factor controls survival and function of terminally differentiated conventional and plasmacytoid dendritic cells, respectively
.
Immunity
2016
;
45
:
626
40
.
27.
Cisse
B
,
Caton
ML
,
Lehner
M
,
Maeda
T
,
Scheu
S
,
Locksley
R
, et al
Transcription factor E2-2 is an essential and specific regulator of plasmacytoid dendritic cell development
.
Cell
2008
;
135
:
37
48
.
28.
Scott
CL
,
Soen
B
,
Martens
L
,
Skrypek
N
,
Saelens
W
,
Taminau
J
, et al
The transcription factor Zeb2 regulates development of conventional and plasmacytoid DCs by repressing Id2
.
J Exp Med
2016
;
213
:
897
911
.
29.
Schütz
C
,
Inselmann
S
,
Sausslele
S
,
Dietz
CT
,
Mu Ller
MC
,
Eigendorff
E
, et al
Expression of the CTLA-4 ligand CD86 on plasmacytoid dendritic cells (pDC) predicts risk of disease recurrence after treatment discontinuation in CML
.
Leukemia
2017
;
31
:
829
36
.
30.
Pear
WS
,
Miller
JP
,
Xu
L
,
Pui
JC
,
Soffer
B
,
Quackenbush
RC
, et al
Efficient and rapid induction of a chronic myelogenous leukemia-like myeloproliferative disease in mice receiving P210 bcr/abl-transduced bone marrow
.
Blood
1998
;
92
:
3780
92
.
31.
Kumari
A
,
Brendel
C
,
Hochhaus
A
,
Neubauer
A
,
Burchert
A
. 
Low BCR-ABL expression levels in hematopoietic precursor cells enable persistence of chronic myeloid leukemia under imatinib
.
Blood
2012
;
119
:
530
9
.
32.
Schoch
C
,
Schnittger
S
,
Bursch
S
,
Gerstner
D
,
Hochhaus
A
,
Berger
U
, et al
Comparison of chromosome banding analysis, interphase- and hypermetaphase-FISH, qualitative and quantitative PCR for diagnosis and for follow-up in chronic myeloid leukemia: a study on 350 cases
.
Leukemia
2002
;
16
:
53
9
.
33.
Cross
NCP
,
White
HE
,
Colomer
D
,
Ehrencrona
H
,
Foroni
L
,
Gottardi
E
, et al
Laboratory recommendations for scoring deep molecular responses following treatment for chronic myeloid leukemia
.
Leukemia
2015
;
29
:
999
1003
.
34.
Schmidt
M
,
Rinke
J
,
Schäfer
V
,
Schnittger
S
,
Kohlmann
A
,
Obstfelder
E
, et al
Molecular-defined clonal evolution in patients with chronic myeloid leukemia independent of the BCR-ABL status
.
Leukemia
2014
;
28
:
2292
9
.
35.
Daley
GQ
,
Van Etten
RA
,
Baltimore
D
. 
Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome
.
Science
1990
;
247
:
824
30
.
36.
Huntly
BJP
,
Shigematsu
H
,
Deguchi
K
,
Lee
BH
,
Mizuno
S
,
Duclos
N
, et al
MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors
.
Cancer Cell
2004
;
6
:
587
96
.
37.
Naka
K
,
Hoshii
T
,
Muraguchi
T
,
Tadokoro
Y
,
Ooshio
T
,
Kondo
Y
, et al
TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia
.
Nature
2010
;
463
:
676
80
.
38.
Warfvinge
R
,
Geironson
L
,
Sommarin
MNE
,
Lang
S
,
Karlsson
C
,
Roschupkina
T
, et al
Single-cell molecular analysis defines therapy response and immunophenotype of stem cell subpopulations in CML
.
Blood
2017
;
129
:
2384
94
.
39.
Giustacchini
A
,
Thongjuea
S
,
Barkas
N
,
Woll
PS
,
Povinelli
BJ
,
Booth
CAG
, et al
Single-cell transcriptomics uncovers distinct molecular signatures of stem cells in chronic myeloid leukemia
.
Nat Med
2017
;
23
:
692
702
.
40.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
P Natl Acad Sci Usa
2005
;
102
:
15545
50
.
41.
Nievergall
E
,
Reynolds
J
,
Kok
CH
,
Watkins
DB
,
Biondo
M
,
Busfield
SJ
, et al
TGF-α and IL-6 plasma levels selectively identify CML patients who fail to achieve an early molecular response or progress in the first year of therapy
.
Leukemia
2016
;
30
:
1263
72
.
42.
See
P
,
Dutertre
C-A
,
Chen
J
,
Günther
P
,
McGovern
N
,
Irac
SE
, et al
Mapping the human DC lineage through the integration of high-dimensional techniques
.
Science
2017
;
356
:
eaag3009
.
43.
Villani
A-C
,
Satija
R
,
Reynolds
G
,
Sarkizova
S
,
Shekhar
K
,
Fletcher
J
, et al
Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors
.
Science
2017
;
356
:
eaah4573
.
44.
Schürch
CM
,
Riether
C
,
Ochsenbein
AF
. 
Dendritic cell-based immunotherapy for myeloid leukemias
.
Front Immunol
2013
;
4
:
496
.
45.
Inselmann
S
,
Liebler
S
,
Brendel
C
,
Koschmieder
S
,
Neubauer
A
,
Burchert
A
. 
BCR-ABL-induced transcriptional repression of the interferon regulatory factor 8 (IRF-8/ICSBP) leads to depletion of plasmocytoid dendritic cells (PDC), which may contribute to leukemogenesis in a murine model of chronic myeloid leukemia
.
Blood
2012
;
120
:
36
.
46.
Valent
P
,
Bonnet
D
,
De Maria
R
,
Lapidot
T
,
Copland
M
,
Melo
JV
, et al
Cancer stem cell definitions and terminology: the devil is in the details
.
Nat Rev Cancer
2012
;
12
:
767
75
.
47.
Naka
K
,
Hoshii
T
,
Muraguchi
T
,
Tadokoro
Y
,
Ooshio
T
,
Kondo
Y
, et al
TGF-β-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia
.
Nature
2010
;
463
:
676
80
.
48.
Gallipoli
P
,
Pellicano
F
,
Morrison
H
,
Laidlaw
K
,
Allan
EK
,
Bhatia
R
, et al
Autocrine TNF-α production supports CML stem and progenitor cell survival and enhances their proliferation
.
Blood
2013
;
122
:
3335
9
.
49.
Moseman
EA
,
Liang
X
,
Dawson
AJ
,
Panoskaltsis-Mortari
A
,
Krieg
AM
,
Liu
Y-J
, et al
Human plasmacytoid dendritic cells activated by CpG oligodeoxynucleotides induce the generation of CD4+CD25+ regulatory T cells
.
J Immunol
2004
;
173
:
4433
42
.
50.
Martinet
J
,
Dufeu-Duchesne
T
,
Bruder Costa
J
,
Larrat
S
,
Marlu
A
,
Leroy
V
, et al
Altered functions of plasmacytoid dendritic cells and reduced cytolytic activity of natural killer cells in patients with chronic HBV infection
.
Gastroenterology
2012
;
143
:
1586
8
.
51.
Fonteneau
J-F
,
Gilliet
M
,
Larsson
M
,
Dasilva
I
,
Münz
C
,
Liu
Y-J
, et al
Activation of influenza virus-specific CD4+ and CD8+ T cells: a new role for plasmacytoid dendritic cells in adaptive immunity
.
Blood
2003
;
101
:
3520
6
.
52.
Salio
M
,
Palmowski
MJ
,
Atzberger
A
,
Hermans
IF
,
Cerundolo
V
. 
CpG-matured murine plasmacytoid dendritic cells are capable of in vivo priming of functional CD8 T cell responses to endogenous but not exogenous antigens
.
J Exp Med
2004
;
199
:
567
79
.
53.
Molldrem
JJ
,
Lee
PP
,
Kant
S
,
Wieder
E
,
Jiang
W
,
Lu
S
, et al
Chronic myelogenous leukemia shapes host immunity by selective deletion of high-avidity leukemia-specific T cells
.
J Clin Invest
2003
;
111
:
639
47
.
54.
Hughes
A
,
Clarson
J
,
Tang
C
,
Vidovic
L
,
White
DL
,
Hughes
TP
, et al
CML patients with deep molecular responses to TKI have restored immune effectors, decreased PD-1 and immune suppressors
.
Blood
2017
;
129
:
blood–2016–10–745992–1176
.
55.
Busque
L
,
Patel
JP
,
Figueroa
ME
,
Vasanthakumar
A
,
Provost
S
,
Hamilou
Z
, et al
Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis
.
Nat Genet
2012
;
44
:
1179
81
.
56.
Burchert
A
. 
Maintaining low BCR-ABL signaling output to restrict CML progression and enable persistence
.
Curr Hematol Malig Rep
2014
;
9
:
9
16
.
57.
Murphy
DJ
,
Junttila
MR
,
Pouyet
L
,
Karnezis
A
,
Shchors
K
,
Bui
DA
, et al
Distinct thresholds govern Myc's biological output in vivo
.
Cancer Cell
2008
;
14
:
447
57
.
58.
Junttila
MR
,
Karnezis
AN
,
Garcia
D
,
Madriles
F
,
Kortlever
RM
,
Rostker
F
, et al
Selective activation of p53-mediated tumour suppression in high-grade tumours
.
Nature
2010
;
468
:
567
71
.