Indoleamine 2,3-Dioxygenase-1 Expressing Dendritic Cell Populations Are Associated with Tumor-Induced Immune Tolerance & Aggressive Disease Biology in Chronic Myelomonocytic Leukemia

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4344-4344
Author(s):  
Abhishek A. Mangaonkar ◽  
Kaaren K. Reichard ◽  
April Chiu ◽  
Matthew T Howard ◽  
Rebecca L King ◽  
...  

Abstract Introduction: Chronic myelomonocytic leukemia (CMML) is a chronic myeloid malignancy associated with monocytosis, autoimmunity (~30%) & an inherent risk for leukemic transformation. Bone marrow (BM) dendritic cell (DC) populations occur in ~30% of patients, with a poorly defined biological & prognostic role. The malignant immune microenvironment is regulated by indoleamine 2,3-dioxygenase-1 (IDO-1) expressing DCs, which modulate regulatory T (Treg) cells & block their conversion into proinflammatory T helper (Th17)-like cells. IDO-1 is a known immune checkpoint & functions by catabolizing tryptophan, an amino acid essential for T cell function. We hypothesized that distinct IDO-1 expressing DC populations in CMML modulate Tregs & contribute towards immune tolerance & aggressive disease biology. Methods: Primary diagnostic CMML peripheral blood mononuclear cells (PBMC) & BM biopsy specimens were obtained after Mayo Clinic IRB approval. A DC population was defined on H&E stained biopsy sections as focal collections (>10) of cells with characteristic elongated nuclei & cytoplasmic extensions. Transcriptomic & protein expression studies assessing IDO-1 expression were done by previously described methods. In addition, IHC expression of PD-1, PD-L1 & CTLA-4 was also done. IDO-1 promoter methylation studies with DIP-seq were performed. The impact on immune tolerance was assessed using mass cytometry (CyTOF). Results: Cohort: Twenty eight patients with CMML were included in the study, median age 70 (range: 51-80) years; 71% males. Eleven (39%) patients had coexisting autoimmune conditions. Of these, 8 (73%) had detectable DC populations either at diagnosis, or during the course of their disease. At a median follow-up of 46 (95% CI 27, 84) months, there were 14 (50%) deaths & 9 (32%) leukemic transformations.IHC results: Nine (32%) patients were identified to have a DC population at CMML diagnosis. CD123 & TCL1 staining was performed in 5 (56%) patients, with 3 being positive for both, & 2 positive for CD123 only (additional IHC studies ongoing). IDO1 expression by IHC was documented in all 9 (100%) cases (Fig 1A & 1B), while rare populations of PD-1, PD-L1 & CTLA-4 lymphocytes were also seen in all cases. Due to the low DC burdens (median cellularity ≤ 5%) & uniform staining intensity, IHC-based grading was not done. Samples at serial time-points, post-HMA therapy & at the time of blast transformation, were available in 5 & 3 patients respectively. Among the patients who did not have DC populations at diagnosis, 5 (42%) developed them post-HMA therapy, while 3 (50%) developed them at the time of LT. The development of DC populations was associated with loss of response to HMA (50%) & disease progression (50%).Transcriptomic analysis: RNA expression data was available on 7 (25%) patients, of whom only 1 (14%) had DC populations at diagnosis. The IDO-1 RPKM value in the former was higher than the mean pooled value in the latter group (330 versus 74, p=0.05).Methylation studies: DIP-seq was performed on 12 (43%) cases from the primary IHC cohort. Qualitative analysis of IDO-1 promoter hypomethylation was conducted & confirmed in all 9 (100%) cases with 5-mC & 5-hmC marks compared to input as displayed in figure 1C.Immune profiling: CyTOF was performed on 4 CMML samples (3 with IDO-1 expressing DC populations at diagnosis) from the primary IHC cohort & compared to a normal PBMC control. Results confirmed an increase in DC populations (fig 1D& 1E), & reduced % of Th17-like T cells in CMML samples compared to control (1.1 versus 5.07, p=0.05, fig 1F).Clinical correlates & survival analysis: With the exception that CMML patients with DC populations had a higher frequency of NRAS (P=0.007) mutations, the two groups were comparable for cytogenetic & molecular abnormalities. The median OS for the cohort was 45 (95% CI 29, 84) months. CMML patients with IDO-1 expressing DC populations at diagnosis had a shorter median OS, in comparison to those without (median OS 30 vs 45, p=0.03, Kaplan-Meier analysis in fig 1G). Conclusions: In conclusion, we demonstrate that DC populations are seen in ~30% of patients with CMML with a uniform expression of IDO-1 & limited expression of PD-1, PD-L1 & CTLA-4. CMML patients with BM DC populations have a higher frequency of NRAS mutations & DC IDO-1 expression is associated with tumor induced immune tolerance. Additional IHC, genomic & preclinical studies with IDO-1 inhibitors are ongoing. Figure 1. Figure 1. Disclosures Al-Kali: Novartis: Research Funding.

2020 ◽  
Author(s):  
Abhishek A. Mangaonkar ◽  
Kaaren K. Reichard ◽  
Moritz Binder ◽  
Giacomo Coltro ◽  
Terra L. Lasho ◽  
...  

AbstractSystemic immune tolerance is not well-characterized in chronic myelomonocytic leukemia (CMML). Due to the presence of clonal plasmacytoid dendritic cells (pDC) in CMML, and the established association of lymph node indoleamine 2,3-dioxygenase-1 (IDO1)-positive (+) DC populations (IDC) with systemic immune tolerance in other malignant contexts, we sought to determine the association of IDO1 expression and bone marrow (BM) DC populations with systemic T-cell compartment changes using primary CMML patient samples (BM, plasma, and peripheral blood mononuclear cells) via immunohistochemistry (IHC), liquid chromatography-mass spectrometry (LC-MS), and time-of-flight mass cytometry (CyTOF). Our results highlight that aggregate BM IDC (CD123 and/or CD11c positive) occur in 33% CMML patients at any disease time-point (IHC), correlate with accentuated tryptophan catabolism (LC-MS, increased kynurenine level, median 4.7 versus 3 microM, P=0.049*), systemic regulatory T-cell expansion (CyTOF, %parent cell type, 14.5 versus 4.9%, P=0.04*) and play a role in disease progression, as evidenced by a higher rate of transformation to acute myeloid leukemia (41 versus 13%, P=0.002**), when compared to CMML patients without BM IDC. Our data also highlight a perturbed immune system in CMML with specific systemic immune signatures, particularly type 1, IL-17 producing helper T, CD4 terminal effector and natural killer cell suppression.Key PointsAggregate IDO1+ dendritic cell populations occur in the CMML bone marrow microenvironment, and their presence correlates with disease progression.Systemic immune microenvironment signatures in CMML indicate an altered T- and natural killer (NK)-cell balance. Specifically, suppression of type 1 helper T (Th1), IL-17 producing helper T (Th17), CD4 terminal effector and NK cells.IDO1+ bone marrow dendritic cell populations in CMML are associated with a T-cell compartment shift towards a regulatory T cell phenotype.


Leukemia ◽  
2017 ◽  
Vol 31 (5) ◽  
pp. 1238-1240 ◽  
Author(s):  
L Brunetti ◽  
V Di Battista ◽  
A Venanzi ◽  
G Schiavoni ◽  
M P Martelli ◽  
...  

Blood ◽  
2012 ◽  
Vol 119 (11) ◽  
pp. 2612-2614 ◽  
Author(s):  
Norio Shiba ◽  
Daisuke Hasegawa ◽  
Myoung-ja Park ◽  
Chisato Murata ◽  
Aiko Sato-Otsubo ◽  
...  

Abstract Familial platelet disorder with a propensity to develop acute myeloid leukemia (FPD/AML) is a rare autosomal dominant disease characterized by thrombocytopenia, abnormal platelet function, and a propensity to develop myelodysplastic syndrome (MDS) and AML. So far, > 20 affected families have been reported. Recently, a second RUNX1 alteration has been reported; however, no additional molecular abnormalities have been found so far. We identified an acquired CBL mutation and 11q-acquired uniparental disomy (11q-aUPD) in a patient with chronic myelomonocytic leukemia (CMML) secondary to FPD with RUNX1 mutation but not in the same patient during refractory cytopenia. This finding suggests that alterations of the CBL gene and RUNX1 gene may cooperate in the pathogenesis of CMML in patients with FPD/AML. The presence of CBL mutations and 11q-aUPD was an important “second hit” that could be an indicator of leukemic transformation of MDS or AML in patients with FPD/AML.


2017 ◽  
Author(s):  
Artem Kaznatcheev ◽  
David Robert Grimes ◽  
Robert Vander Velde ◽  
Vincent Cannataro ◽  
Etienne Baratchart ◽  
...  

Acquired therapy resistance to cancer treatment is a common and serious clinical problem. The classic U-shape model for the emergence of resistance supposes that: (1) treatment changes the selective pressure on the treatment-naive tumour; (2) this shifting pressure creates a proliferative or survival difference between sensitive cancer cells and either an existing or de novo mutant; (3) the resistant cells then out-compete the sensitive cells and – if further interventions (like drug holidays or new drugs or dosage changes) are not pursued – take over the tumour: returning it to a state dangerous to the patient. The emergence of ruxolitinib resistance in chronic myelomonocytic leukemia (CMML) seems to challenge the classic model: we see the global properties of resistance, but not the drastic change in clonal architecture expected with the selection bottleneck. To study this, we explore three population-level models as alternatives to the classic model of resistance. These three effective models are designed in such a way that they are distinguishable based on limited experimental data on the time-progression of resistance in CMML. We also propose a candidate reductive implementation of the proximal cause of resistance to ground these effective theories. With these reductive implementations in mind, we also explore the impact of oxygen diffusion and spatial structure more generally on the dynamics of CMML in the bone marrow concluding that, even small fluctuations in oxygen availability can seriously impact the efficacy of ruxolitinib. Finally, we look at the ability of spatially distributed cytokine signaling feedback loops to produce a relapse in symptoms similar to what we observe in the clinic.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 611-611 ◽  
Author(s):  
Anna Jankowska ◽  
Hideki Makishima ◽  
Ramon V. Tiu ◽  
Hadrian Szpurka ◽  
Yun Huang ◽  
...  

Abstract Abstract 611 Chronic myelomonocytic leukemia (CMML), a myelodysplastic/myeloproliferative overlap neoplasm, is characterized by monocytic proliferation, cytomorphologic dysplasia and frequent progression to acute myelogeneous leukemia (AML). The molecular basis of CMML is poorly defined, although somatic mutations in a number of genes have recently been identified in a proportion of patients. Single nucleotide polymorphisms array (SNP-A) technologies have improved the definition of shared regions of loss of heterozygosity (LOH), including uniparental disomy (UPD) and facilitated discovery of new mutations c-CBL, TET2, and EZH2 which can occur in a homozygous configuration in the areas of UPD. Other mutations such as ASXL1 have been found in heterozygous form. In myeloid malignancies we have also identified mutations in UTX, which like EZH2 and ASXL1, are involved in modification of histone methylation. Based on these findings we hypothesized that defining the mutational spectrum of CMML would help in the molecular characterization of this disease and have diagnostic and prognostic significance. Within this spectrum, we stipulated that various genes involved in epigenetic regulation may be especially affected by mutations in CMML. Here we present results of broad molecular screen in a group of 63 patients with CMML (32 CMML-1, 15 CMML-2 and 16 CMML-derived sAML) which included SNP-A karyotyping and mutational screen for IDH1/2, RAS, TET2, ASXL1, c-CBL, JAK2, UTX and EZH2. First, we aligned all lesions that were detected by SNP-A. In addition to microdeletions involving 4q24 and 11q23.3, we detected recurrent areas of somatic UPD involving chromosomes 1, 4, 7 and 11 and the corresponding homozygous mutations in RAS (UPD1p, N=1), EZH2 (UPD7q N=3), c-CBL (UPD11q, N=4), TET2 (UPD4q, N=6), and UTX genes (UPDXq, N=1). When all patients were sequenced, TET2, ASXL1, c-CBL, IDH1/2, RAS, JAK2, UTX and EZH2 mutations were found in 48%, 24%, 14%, 5%, 11%, 2%, 6% and 8% of patients, respectively. In 78% of patients, >1 mutation was found. Concomitant second and third mutations were found in 34% and 5% of patients, respectively. The most frequently observed combinations included TET2 and ASXL1 (14%) and TET2 and c-CBL (6%). Only 22% of patients had no alterations in analyzed genes. Novel UTX and EZH2 mutations were present either alone or in combination with other mutations. Study of potential functional consequences of the foregoing gene mutations revealed an association of TET2 mutations with consistently low levels of 5-hydroxymethylcytosine (5-hmC), quantitated by dot blot assay, while c-CBL mutations were associated with aberrant phospho-STAT5 staining. Loss of H3K27-me3 in cases with EZH2 mutations but not controls, and an increase in UTX mutant case was identified as measured by ELISA and western blot. When we tested for association of different mutations with pathomorphologic features, specific clinical features were not identified, except for an association of TET2 and c-CBL mutations with more advanced age (p=.0004 and p=.02, respectively), RAS mutation with increased blasts (p=.03) and UTX with dysplastic megakaryocytes (p=.03). Splenomegaly was noted more frequent in c-CBL mutants than any other patient group. No differences in OS and EFS were observed between mutant and wt cases. There is a trend toward better OS in TET2 mutants compared to WT in the good cytogenetic risk group (17 vs 8 mo, p=.07) but worse outcomes in TET2 mutants in the intermediate cytogenetic risk group (OS 2 vs. 16 mo, p=.001; EFS 2 vs. 9 mo, p=.04). As expected, patients who have accumulated more mutations have a trend toward inferior outcomes compared to those with single mutations but better than those who are WT (>1 mutations vs 1 mutation vs WT, 16 vs 18 vs 9 mo, p=.07 in low risk CMML). In summary, our study identified the presence of a wide spectrum of mutations in CMML with various combinations, including the newly discovered mutations in UTX and EZH2 genes. Our results suggest that molecular abnormalities affecting various pathways can lead to a clinically indistinguishable phenotype. It is possible that these mutations are secondary in nature but work in conjunction with a yet unidentified founder defect. The abundance of mutations in factors known or hypothesized to be involved in epigenetic regulation in CMML provide important implications for future research into the development of effective therapies for this disease. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document