• Some 10.1% of adults with non–Langerhans cell histiocytosis have a concomitant myeloid neoplasm with each often harboring distinct mutations.

  • The presence of distinct kinase mutations in histiocytosis and myeloid neoplasms resulted in discordant responses to targeted therapy.

Erdheim-Chester disease (ECD) is a rare non–Langerhans cell histiocytosis that most commonly affects adults and is driven by a high frequency of mutations in BRAF, MAP2K1, and kinases promoting MAPK signaling. Because of the relative rarity of ECD, key clinical features of the disease may not be well defined. Across a multi-institutional cohort of 189 patients with ECD and ECD overlapping with Langerhans cell histiocytosis (so-called mixed histiocytosis [MH]), we identified an unexpected and heretofore undescribed frequent occurrence of myeloid neoplasms among patients with ECD and MH. Some 10.1% (19/189) of patients with ECD have an overlapping myeloid neoplasm, most commonly occurring as a myeloproliferative neoplasm (MPN), myelodysplastic syndrome (MDS), or mixed MDS/MPN overlap syndrome (including chronic myelomonocytic leukemia). Consistent with this, molecular analysis frequently detected hallmark driver mutations of myeloid neoplasms (such as JAK2V617F and CALR mutations) coexisting with those characteristic of histiocytosis (such as BRAFV600E and MAP2K1 mutations). Histiocytosis patients diagnosed with a concomitant myeloid malignancy were significantly older at diagnosis and more commonly presented with MH than those without a myeloid malignancy. In some cases, the presence of distinct kinase mutations in the histiocytosis and myeloid neoplasm resulted in discordant and adverse responses to kinase-directed targeted therapies. These data highlight the clinical importance of evaluating adults with histiocytosis for a concomitant myeloid neoplasm.

Erdheim-Chester disease (ECD) is a form of histiocytosis characterized by tissue infiltration with foamy histiocytes that are CD68+, CD163+, CD1a, and Langerin (CD207). Prior to 2012, there was a long-standing debate as to whether ECD represented a clonal hematopoietic disorder vs an inflammatory disease related to aberrant immune activation. However, since 2010, a series of genomic studies have uncovered BRAFV600E mutations in 55% to 70% of ECD as well as Langerhans cell histiocytosis (LCH) patients, providing evidence that these diseases represent clonal disorders driven by activated MAPK signaling.1,2  Subsequently, activating mutations in MAP2K1,3-6 ARAF,6,7  and fusions in kinases including BRAF6,8  were found in the majority of BRAFV600-wild-type ECD and LCH patients. PI3KCA9  and N/KRAS mutations are more frequent in ECD than in LCH. Despite the distinct clinical and histologic presentations of LCH and ECD, the previous studies identify a similar constellation of genomic alterations across both disorders. Moreover, nearly 20% of patients with ECD have a diagnosis of both ECD and LCH simultaneously (so-called mixed histiocytosis [MH]) where both lesions may contain the BRAFV600E mutation.10  For these reasons, ECD and LCH have been grouped together in the same class of conditions in a recent revision of histiocytosis classifications.11 

Although there have been great molecular advances in ECD and LCH over the last 7 years, our knowledge of the natural history of ECD and MH and their associated clinical and prognostic features remains incomplete. Nevertheless, despite great recent progress in the understanding of histiocytosis, the cellular origins of LCH, ECD, and MH are not completely understood.

In hopes of clarifying the clinical and molecular features of ECD and MH and facilitating translational research in these disorders, we assembled ECD patient data across 2 institutions in Europe and the United States with a specific research focus on ECD. Using these data, we have uncovered an unexpected high frequency of concomitant clinically diagnosed myeloid neoplasms in patients diagnosed with ECD and MH. Here we describe the clinical, pathological, and molecular features of the hematological disorders associated with ECD. Diagnosis of a concurrent myeloid neoplasm and histiocytosis had important therapeutic consequences for patients. These findings have clear clinical relevance in addition to potentially enlightening our understanding of the etiology of ECD and its disease classification.

Study design

We performed an international 2-center retrospective study of patients with biopsy-proven ECD who were referred at least once to the Internal Medicine Department of Pitié-Salpêtrière Hospital (Paris, France) or Memorial Sloan Kettering Cancer Center (MSKCC; New York, NY) between November 1981 and November 2016. A total of 189 cases of ECD were reviewed (39 from the MSKCC and 150 from the Pitié-Salpêtrière Hospital; 12 of which were also studied by whole exome sequencing previously6 ). Written informed consent was obtained from patients according to Helsinki convention, and this study received approval from the Ethics Committee Ile-de-France III and the Institutional Review Board at MSKCC.

Patients

ECD was diagnosed according to published criteria: (1) tissue biopsy demonstrating typical pathological signs of ECD (infiltration with foamy histiocytes and CD68+CD1a on immunohistochemical staining); (2) symmetric skeletal uptake on 99mTc bone scintigraphy or 18-fluorodeoxyglucose positron emission tomographic (18F-FDG PET) or magnetic resonance imaging scan with the involvement of at least 1 other organ typically affected in ECD (xanthelasma, perinephric infiltration, “coated aorta,” pericardial infiltration, right atrial pseudotumor, or brain or dural infiltration). Patients with MH were included as well. Bone marrow (BM) aspirations and biopsies were performed according to standard of care when a complete blood count abnormality (anemia, thrombocytopenia, polycythemia, thrombocytosis, or monocytosis) was noted and not explained by nonmalignant causes such as iron, vitamin B12, or folic acid deficiencies or inflammatory syndrome. Hematological malignancies (myeloid neoplasms, lymphoma, and myeloma) were diagnosed according to the 2016 revision to the World Health Organization (WHO) classification of hematological malignancies.12,13  For each patient, clinical parameters (age at diagnosis, sex, and main organ involvement), laboratory parameters (blood count, electrolytes, liver function tests, and C-reactive protein), and outcome (treatments and death) were obtained from medical records.

Molecular analyses

For patients from Pitié-Salpêtrière Hospital, BRAFV600E mutational status was evaluated as previously described.2 JAK2V617F mutational status was evaluated in BM aspirate, peripheral blood (PB), or tissue biopsy using the TaqMan method with an Applied 7500 automaton. For patients with available material, targeted deep sequencing of the exonic regions of 24 genes (ASXL1, ATRX, BCOR, BCORL1, CBL, CEBPA, CSF3R, DNMT3A, ETV6, EZH2, FLT3, GATA2, JAK2, KDM6A, KIT, NRAS, RUNX1, SETBP1, SF3B1, STAG2, TET2, TP53, WT1, and ZRSR2) was performed on flow-sorted CD14+ cells (for patients with chronic myelomonocytic leukemia [CMML]) or CD14CD15+CD16+ polymorphonuclear cells (for patients with classic myeloproliferative neoplasms [MPNs]) as well as T cells (CD3+CD19CD14CD34) for germ-line control. This specific gene panel was used as it includes the most frequently mutated genes in myeloid neoplasms. Targeted regions were polymerase chain reaction amplified using an AmpliSeq (Life Technologies), and polymerase chain reaction products sequenced with a MiSeq (Illumina, San Diego, CA) instrument with a mean depth of 1000X (range 590-1800). We used a variant allele frequency minimal threshold of 10% to detect mutation.

For patients from MSKCC, genomic analysis of histiocytosis tissue biopsy and PB mononuclear cells was performed using the MSKCC IMPACT (Integrated Mutation Profiling of Actionable Cancer Targets) assay as previously described.14 

Statistical analysis

Continuous variables were expressed as median and range, and categorical variables as numbers and percentages. Differences between groups of patients were tested with the Mann-Whitney U test for continuous data, and by Fischer exact test or the χ2 test for categorical data. Survival analyses were performed with Kaplan-Meier curves and log-rank test. We used SAS version 9.0 (SAS Institute) and GraphPad Prism 5 for analyses.

Frequent occurrence of myeloid neoplasms in patients with ECD

We reviewed 189 cases of ECD, including MH. Associated hematological disorders (excluding another histiocytosis) were observed in 23 patients (12.2%). Apart from patients with lymphoproliferative or autoimmune disorders (2 patients had lymphoma, 2 had immune thrombocytopenic purpura, 1 had myeloma, and 1 developed an acute lymphoblastic leukemia [ALL] in the context of a primary myelofibrosis), hematological diseases associated with ECD were myeloid neoplasms (10.1%) (Figure 1; supplemental Table 1 and supplemental Figure 1, available on the Blood Web site). This frequency was higher in the United States (15.3%) than the French (8.6%) cohort (P = .22). Among these 19 patients, 8 had CMML; 4, ET; 2, MDSs; 2, primary myelofibrosis; 2, AML (1 secondary to MDS and 1 to PV); and 1, PV. One patient also developed an ALL in the course of his MPN. Seven patients were diagnosed with myeloid neoplasm before the ECD diagnosis (median 4 years between the 2 diagnoses, range 1-22 years), 6 were diagnosed simultaneously, and 6 were diagnosed with myeloid neoplasm after a diagnosis of histiocytosis (median 1 year, range 1-4 years).

Figure 1.

Distribution of myeloid neoplasms in patients with concomitant non-LCH and genetic analysis of both disorders. (A) Pie chart demonstrating proportion of non-LCH patients with concomitant myeloid neoplasm and types of myeloid neoplasms diagnosed. ET, essential thrombocytosis; MDS, myelodysplastic syndrome; MF, primary myelofibrosis; PV, polycythemia vera; sAML, secondary acute myeloid leukemia transformed from antecedent hematological malignancy. (B) Genetic analysis of non-LCH and concomitant myeloid neoplasm. Each patient is noted by a column. Patients had clinical diagnosis of ECD or an overlap of ECD plus LCH or ECD/LCH plus Rosai Dorfman disease (RDD) based on tissue biopsy and clinical evaluation in addition to a form of WHO-classified myeloid. Mutations identified in histiocytosis tissue lesion biopsy alone in each patient are noted in the middle boxes, and those mutations detected in PB or BM mononuclear cells are noted in bottom boxes.

Figure 1.

Distribution of myeloid neoplasms in patients with concomitant non-LCH and genetic analysis of both disorders. (A) Pie chart demonstrating proportion of non-LCH patients with concomitant myeloid neoplasm and types of myeloid neoplasms diagnosed. ET, essential thrombocytosis; MDS, myelodysplastic syndrome; MF, primary myelofibrosis; PV, polycythemia vera; sAML, secondary acute myeloid leukemia transformed from antecedent hematological malignancy. (B) Genetic analysis of non-LCH and concomitant myeloid neoplasm. Each patient is noted by a column. Patients had clinical diagnosis of ECD or an overlap of ECD plus LCH or ECD/LCH plus Rosai Dorfman disease (RDD) based on tissue biopsy and clinical evaluation in addition to a form of WHO-classified myeloid. Mutations identified in histiocytosis tissue lesion biopsy alone in each patient are noted in the middle boxes, and those mutations detected in PB or BM mononuclear cells are noted in bottom boxes.

Close modal

ECD patients with a myeloid neoplasm were also more likely to have a diagnosis of an overlap histiocytosis (ECD associated with LCH [n = 6] and Rosai Dorfman disease [n = 2]) than those patients with ECD and no concomitant myeloid neoplasm (P = .02; Table 1). In addition, patients with ECD plus a concomitant myeloid neoplasm were significantly older at ECD diagnosis (68 vs 56.5 years; P = .0005) and had a lower survival (82 vs 364 months; P = .001) than ECD patients without a myeloid neoplasm (supplemental Figure 2). Deaths were mainly because of cardiac insufficiency or infections, but 2 patients died of hematological disease (patient #1 died of ALL, and patient #11 died of an AML secondary to MDS).

Table 1.

Comparison of clinical and biological characteristics of ECD patients with or without concomitant myeloid neoplasm

VariablesMyeloid neoplasm (n = 19)Others (n = 170)P
MH, % 6 (31.5) 18 (10.6) .02 
Male sex, % 17 (89.4) 115 (68.8) .06 
Age, y 68 (60-73) 56.5 (46-66) .0005 
BRAFV600E mutation in ECD or MH, % 12 (63.2) 89 (52.3) .37 
JAK2V617F mutation in MPN/MDS, % 7 (36.8) 0 (0) <.0001 
Elevated CRP, % 15 (78.9) 130 (76.4) 1.00 
Bone scintigraphy or PET uptake, % 14 (73.7) 155 (91.1) .018 
Number of sites 3 (2-4) 3 (2-5) .21 
Coated aorta, % 8 (42.1) 66 (38.8) .78 
Pericardial infiltration, % 6 (31.5) 42 (24.7) .51 
Right auricular pseudotumor, % 6 (31.5) 57 (33.5) .86 
Xanthelasmas, % 2 (10.5) 41 (24.1) .25 
Exophthalmos, % 4 (21.0) 36 (21.1) 1.00 
CNS infiltration, % 2 (10.5) 68 (40.0) .01 
Pituitary infiltration, % 2 (10.5) 35 (20.6) .37 
Perirenal infiltration, % 14 (73.6) 94 (55.9) .12 
IFN treatment, % 13 (68.4) 110 (64.7) .74 
BRAF inhibitor treatment, % 7 (36.8) 30 (17.6) .045 
Death, % 8 (42.1) 33 (19.4) .002 
Survival, mo 82 (40-99) 364 (129-364) .001 
VariablesMyeloid neoplasm (n = 19)Others (n = 170)P
MH, % 6 (31.5) 18 (10.6) .02 
Male sex, % 17 (89.4) 115 (68.8) .06 
Age, y 68 (60-73) 56.5 (46-66) .0005 
BRAFV600E mutation in ECD or MH, % 12 (63.2) 89 (52.3) .37 
JAK2V617F mutation in MPN/MDS, % 7 (36.8) 0 (0) <.0001 
Elevated CRP, % 15 (78.9) 130 (76.4) 1.00 
Bone scintigraphy or PET uptake, % 14 (73.7) 155 (91.1) .018 
Number of sites 3 (2-4) 3 (2-5) .21 
Coated aorta, % 8 (42.1) 66 (38.8) .78 
Pericardial infiltration, % 6 (31.5) 42 (24.7) .51 
Right auricular pseudotumor, % 6 (31.5) 57 (33.5) .86 
Xanthelasmas, % 2 (10.5) 41 (24.1) .25 
Exophthalmos, % 4 (21.0) 36 (21.1) 1.00 
CNS infiltration, % 2 (10.5) 68 (40.0) .01 
Pituitary infiltration, % 2 (10.5) 35 (20.6) .37 
Perirenal infiltration, % 14 (73.6) 94 (55.9) .12 
IFN treatment, % 13 (68.4) 110 (64.7) .74 
BRAF inhibitor treatment, % 7 (36.8) 30 (17.6) .045 
Death, % 8 (42.1) 33 (19.4) .002 
Survival, mo 82 (40-99) 364 (129-364) .001 

CNS, central nervous system; CRP, C-reactive protein; IFN, interferon-α.

Molecular analysis of patients with ECD and concomitant myeloid neoplasm

Given the unexpected high frequency of myeloid neoplasms associated with histiocytosis, we next sought to examine the molecular features of histiocytosis lesions and the coexisting myeloid neoplasms in these individuals. Many of these patients harbored kinase alterations characteristic of both ECD and myeloid neoplasms. Among the 19 patients with concomitant myeloid neoplasm and ECD, 12 (63.2%) harbored the BRAFV600E mutation in ECD tissue biopsy material, and 7 (36.8%) were positive for JAK2V617F in PB and/or BM. Interestingly, 4 patients (23.5%) had both BRAFV600E and JAK2V617F mutations simultaneously (Figure 2). Similarly, 1 patient with ET had a CALR mutation as well as the BRAFV600E mutation, and another had a MAP2K1 mutation in the histiocytic disease associated with a JAK2V167F mutated ET. Unlike BRAFV600E or MAP2K1 mutations, which were detected only in histiocytosis lesions, mutations in NRAS could be found in both histiocytosis and myeloid neoplasms, as exemplified by a patient who had the same NRAS mutation in ECD lesions from perirenal tissue as well as BM and PB following a diagnosis of CMML. In addition to harboring mutations in JAK2 and CALR, patients with myeloid neoplasm-associated histiocytosis also carried additional mutations in transcriptional regulatory genes common in myeloid neoplasms but rare in ECD,6  such as mutations in TET2, ASXL1, IDH2, U2AF1, and TP53 (Figure 1).

Figure 2.

Effect of targeted therapies on non-LCH and concomitant myeloid neoplasm. (A-E) Effect of vemurafenib on a 75-year-old patient with BRAFV600E-mutant ECD and concomitant JAK2V617F/IDH2R140Q-mutant MDS/MPN. (A) Absolute monocytes (orange line; left y-axis) and urinary BRAFV600E cell-free DNA quantitation (red line; right y-axis) pre- and postvemurafenib therapy (shaded area represents period of vemurafenib treatment). (B) 18F-FDG PET scan pre- (left) and postvemurafenib (right) with corresponding fused computed tomography/18F-FDG PET below. (C) Hematoxylin and eosin–stained biopsies of femoral bone revealing characteristic xanthogranulomatous lesion of ECD within a fibrotic background (histiocytes were CD68+ by immunohistochemistry [not shown]). Original magnification ×400. (D) Evidence of myeloid neoplasm because of the presence of dysplastic myeloid cells (hypogranulation and pseudo–Pelger-Huet cell) in BM aspirate (left; original magnification ×400), increased number of CD34+ cells (middle; original magnification ×100), and hematoxylin and eosin stain revealing hypercellular marrow with dysplastic megakaryocytes (right; original magnification ×200). (E-G) Effect of MEK inhibitor therapy on monocytosis and PB counts on the 66-year-old patient with NRASQ61R-mutant ECD and CMML described in Figure 1. 18F-FDG PET (F) and fused computed tomography/18F-FDG PET (G) pre- and 2 months posttrametinib treatment in this same patient.

Figure 2.

Effect of targeted therapies on non-LCH and concomitant myeloid neoplasm. (A-E) Effect of vemurafenib on a 75-year-old patient with BRAFV600E-mutant ECD and concomitant JAK2V617F/IDH2R140Q-mutant MDS/MPN. (A) Absolute monocytes (orange line; left y-axis) and urinary BRAFV600E cell-free DNA quantitation (red line; right y-axis) pre- and postvemurafenib therapy (shaded area represents period of vemurafenib treatment). (B) 18F-FDG PET scan pre- (left) and postvemurafenib (right) with corresponding fused computed tomography/18F-FDG PET below. (C) Hematoxylin and eosin–stained biopsies of femoral bone revealing characteristic xanthogranulomatous lesion of ECD within a fibrotic background (histiocytes were CD68+ by immunohistochemistry [not shown]). Original magnification ×400. (D) Evidence of myeloid neoplasm because of the presence of dysplastic myeloid cells (hypogranulation and pseudo–Pelger-Huet cell) in BM aspirate (left; original magnification ×400), increased number of CD34+ cells (middle; original magnification ×100), and hematoxylin and eosin stain revealing hypercellular marrow with dysplastic megakaryocytes (right; original magnification ×200). (E-G) Effect of MEK inhibitor therapy on monocytosis and PB counts on the 66-year-old patient with NRASQ61R-mutant ECD and CMML described in Figure 1. 18F-FDG PET (F) and fused computed tomography/18F-FDG PET (G) pre- and 2 months posttrametinib treatment in this same patient.

Close modal

Effect of targeted therapies on coexisting ECD and myeloid neoplasm

Although BRAF inhibition has resulted in remarkable clinical responses for patients with BRAFV600E-mutant histiocytosis,15-18  there is a well-described risk of paradoxical activation of cytokine signaling in cells bearing kinase mutations other than BRAFV600E upon exposure to RAF inhibitors.19-22  BRAF inhibitors (vemurafenib [n = 6] or dabrafenib [n = 1]) were given to 7/19 patients with BRAFV600E-mutant ECD and coexisting myeloid neoplasm here. In 3 cases, vemurafenib treatment resulted in an increase in blood counts, which led to treatment discontinuation. For example, patient #14 was treated with vemurafenib for BRAFV600E-mutant ECD with a partial metabolic response at 2.3 months and substantial reduction in BRAFV600E mutant allele burden as measured by analysis of urinary cell-free DNA23  (Figure 2A-B). However, vemurafenib was discontinued at this time point because of a significant increase in monocyte count in conjunction with development of abdominal pain because of a new splenic infarct. Although PET scan at this point revealed resolution of FDG-avid femur lesions characteristic of ECD, there was a paradoxical increased in FDG avidity in all other major bones of the body (Figure 2A-C). This constellation of findings prompted a BM aspirate and biopsy that led to a diagnosis of JAK2V617F/IDH2R140Q-mutant MDS/MPN overlap disorder (Figure 2D). The patient’s monocytosis returned to normal levels following vemurafenib discontinuation further suggesting that paradoxical stimulation of JAK2-mutant cells in response to RAF inhibition was the basis for the patient’s adverse response to vemurafenib (Figure 2).

In contrast to the previous cases where distinct activating kinase mutations complicated use of a single kinase inhibitor, in other instances where the histiocytosis and myeloid neoplasm shared the same kinase mutation, use of targeted therapeutics resulted in beneficial response across both conditions. For example, patient #13 had NRAS-mutated ECD associated with a CMML-1 with the same NRAS mutation. MEK inhibition with cobimetinib in this patient led to a complete metabolic response by PET scan after 2 months in addition to improving monocyte and platelet counts (Figure 2E-G).

This study identifies a high prevalence (10.1%) of myeloid neoplasms among adults with non-LCH. This frequency of myeloid neoplasms is much higher than that encountered general population which ranges between 0.7 and 17.1 per 100 000 people in Europe, depending on the subtype.24  Although prior case reports and case series25-29  have noted additional hematological malignancies in patients with histiocytosis of the L group,11  the high frequency of myeloid neoplasms in a large population of adult histiocytosis patients has not previously been recognized, likely because of the relative rarity of ECD. Nonetheless, knowledge of the presence of an associated myeloid neoplasm in ECD patients has important implications for clinical management of adult histiocytosis patients as well as the classification and biological understanding of these disorders. The difference in prevalence of myeloid neoplasms in histiocytosis patients between MSKCC (15.3%) and Pitié-Salpêtrière Hospital (8.6%) could be attributable to an institution bias, because Pitié-Salpêtrière Hospital is not a referral center for neoplastic diseases. Although this might result in a skewed estimation of the prevalence of myeloid neoplasms among histiocytosis patients, the prevalence at either center remains high.

Histiocytosis patients with an associated myeloid neoplasm seemed to be significantly older than those without a myeloid neoplasm. One hypothesis for this observation could be the well-described association between aging and clonal hematopoiesis with an increased frequency of myeloid neoplasms because of acquisition of somatic mutations in genes commonly mutated in myeloid neoplasms.30-32  Moreover, the lower survival rate observed in histiocytosis patients with coexisting myeloid neoplasms might be attributable to the older age of those patients and not to a more severe disease.

From a clinical standpoint, the data presented here suggest that patients with ECD should be carefully evaluated at diagnosis for a coexisting myeloid neoplasm. We propose that any histiocytosis patient with a complete blood count abnormality that cannot be explained by a nonmalignant cause should undergo a BM aspiration and biopsy with histomorphologic, flow cytometric, and genetic analysis according to standard of care for myeloid malignancies. Evaluation of a concomitant myeloid neoplasm in a patient with non-LCH will be particularly important before initiation of targeted therapies for refractory ECD. As illustrated here, the myeloid neoplasm may be diagnosed only after initiation of therapy for ECD, and knowledge of both neoplasms would have influenced therapeutic decision making. Because of the paradoxical activation of RAS signaling mediated by RAF inhibitors, use of RAF inhibitors may uncover or enhance growth of a malignancy driven by a mutation other than BRAFV600E19-22  (including mutations in NRAS, KRAS, or JAK2) as illustrated here.

In the recently revised 2016 WHO classification, ECD and LCH are classified with lymphoid neoplasms.12  This is largely based on several case reports describing individual patients with lymphoid neoplasms and a clonally related secondary malignant histiocytosis.33-37  However, the high frequency of myeloid neoplasms in patients with ECD or MH suggests a nonfortuitous biological association of both conditions. Although it is well established that myeloid neoplasms such as classic MPNs, MDS, and AML originate from hematopoietic stem cells,38-41  the cell(s) of origin of LCH and non-LCH conditions are far less clearly delineated. A common nonconstitutional trisomy 21 was recently reported in an adult patient with both LCH and mixed phenotype T/myeloid acute leukemia,42  suggesting a common origin of both tumors. Recent data suggest that the BRAFV600E mutation is detectable in CD34+ hematopoietic progenitors in at least a portion of pediatric LCH patients.43  Moreover, blood monocytes harboring the same mutations as pathological histiocytes have been reported in LCH43  and non-LCH.44  At the same time, the discovery of patients with BRAFV600E/JAK2 wild-type ECD lesions with a concomitant BRAF wild-type/JAK2V617F mutant clonal hematopoietic disorder suggests that the 2 mutations must arise at distinct points of hematopoietic development in many patients. Further efforts to functionally characterize the precise cellular origin of both conditions are therefore warranted.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

B.H.D. is supported by the American Society of Hematology Senior Research Training Award for Fellows and the New York State Council on Graduate Medical Education Empire Clinical Research Investigator Program Fellowship. E.L.D. and O.A.-W. are supported by grants from the Erdheim-Chester Disease Global Alliance and the Histiocytosis Association. O.A.-W. is supported by grants from the Edward P. Evans Foundation; the Department of Defense Bone Marrow Failure Research Program (BM150092 and W81XWH-12-1-0041); the National Institutes of Health, National, Heart, Lung, and Blood Institute (R01 HL128239) and a National Cancer Institute K08 Clinical Investigator Award (1K08CA160647-01); the Josie Robertson Investigator Program; a Damon Runyon Clinical Investigator Award; an award from the Starr Foundation (I8-A8-075); the Leukemia and Lymphoma Society; and the Pershing Square Sohn Cancer Research Alliance. J.-F.E. is supported by grants from Association pour la Recherche et l'Enseignement en Pathologie.

Contribution: M.P., E.L.D., J.-F.E., D.R.-W., B.H.D., N.O., A.D., O.A.B., N.D., and Z.H.-R. performed the experiments and analyzed data; M.P., F.C.-A., E.L.D., N.G., C.S., G.A.U., R.R., J.-E.K., T.S., F.C., C.B., B.H., Z.A., and J.-F.E. collected patient material and clinical data; and M.P., E.L.D., J.-F.E., O.A.-W., and J.H. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Omar Abdel-Wahab, 1275 York Ave, New York, NY 10065; e-mail: abdelwao@mskcc.org; and Julien Haroche, Service de Médecine Interne 2, Institut E3M, Hôpital de la Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, France; e-mail: julien.haroche@aphp.fr.

1.
Badalian-Very
G
,
Vergilio
J-A
,
Degar
BA
, et al
.
Recurrent BRAF mutations in Langerhans cell histiocytosis
.
Blood
.
2010
;
116
(
11
):
1919
-
1923
.
2.
Haroche
J
,
Charlotte
F
,
Arnaud
L
, et al
.
High prevalence of BRAF V600E mutations in Erdheim-Chester disease but not in other non-Langerhans cell histiocytoses
.
Blood
.
2012
;
120
(
13
):
2700
-
2703
.
3.
Brown
NA
,
Furtado
LV
,
Betz
BL
, et al
.
High prevalence of somatic MAP2K1 mutations in BRAF V600E-negative Langerhans cell histiocytosis
.
Blood
.
2014
;
124
(
10
):
1655
-
1658
.
4.
Chakraborty
R
,
Hampton
OA
,
Shen
X
, et al
.
Mutually exclusive recurrent somatic mutations in MAP2K1 and BRAF support a central role for ERK activation in LCH pathogenesis
.
Blood
.
2014
;
124
(
19
):
3007
-
3015
.
5.
Nelson
DS
,
van Halteren
A
,
Quispel
WT
, et al
.
MAP2K1 and MAP3K1 mutations in Langerhans cell histiocytosis
.
Genes Chromosomes Cancer
.
2015
;
54
(
6
):
361
-
368
.
6.
Diamond
EL
,
Durham
BH
,
Haroche
J
, et al
.
Diverse and targetable kinase alterations drive histiocytic neoplasms
.
Cancer Discov
.
2016
;
6
(
2
):
154
-
165
.
7.
Nelson
DS
,
Quispel
W
,
Badalian-Very
G
, et al
.
Somatic activating ARAF mutations in Langerhans cell histiocytosis
.
Blood
.
2014
;
123
(
20
):
3152
-
3155
.
8.
Chakraborty
R
,
Burke
TM
,
Hampton
OA
, et al
.
Alternative genetic mechanisms of BRAF activation in Langerhans cell histiocytosis
.
Blood
.
2016
;
128
(
21
):
2533
-
2537
.
9.
Héritier
S
,
Saffroy
R
,
Radosevic-Robin
N
, et al
.
Common cancer-associated PIK3CA activating mutations rarely occur in Langerhans cell histiocytosis
.
Blood
.
2015
;
125
(
15
):
2448
-
2449
.
10.
Hervier
B
,
Haroche
J
,
Arnaud
L
, et al
;
French Histiocytoses Study Group
.
Association of both Langerhans cell histiocytosis and Erdheim-Chester disease linked to the BRAFV600E mutation
.
Blood
.
2014
;
124
(
7
):
1119
-
1126
.
11.
Emile
JF
,
Abla
O
,
Fraitag
S
, et al
;
Histiocyte Society
.
Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages
.
Blood
.
2016
;
127
(
22
):
2672
-
2681
.
12.
Swerdlow
SH
,
Campo
E
,
Pileri
SA
, et al
.
The 2016 revision of the World Health Organization classification of lymphoid neoplasms
.
Blood
.
2016
;
127
(
20
):
2375
-
2390
.
13.
Arber
DA
,
Orazi
A
,
Hasserjian
R
, et al
.
The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia
.
Blood
.
2016
;
127
(
20
):
2391
-
2405
.
14.
Cheng
DT
,
Mitchell
TN
,
Zehir
A
, et al
.
Memorial Sloan Kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology
.
J Mol Diagn
.
2015
;
17
(
3
):
251
-
264
.
15.
Haroche
J
,
Cohen-Aubart
F
,
Emile
J-F
, et al
.
Dramatic efficacy of vemurafenib in both multisystemic and refractory Erdheim-Chester disease and Langerhans cell histiocytosis harboring the BRAF V600E mutation
.
Blood
.
2013
;
121
(
9
):
1495
-
1500
.
16.
Haroche
J
,
Cohen-Aubart
F
,
Emile
JF
, et al
.
Reproducible and sustained efficacy of targeted therapy with vemurafenib in patients with BRAF(V600E)-mutated Erdheim-Chester disease
.
J Clin Oncol
.
2015
;
33
(
5
):
411
-
418
.
17.
Héritier
S
,
Jehanne
M
,
Leverger
G
, et al
.
Vemurafenib use in an infant for high-risk Langerhans cell histiocytosis
.
JAMA Oncol
.
2015
;
1
(
6
):
836
-
838
.
18.
Hyman
DM
,
Puzanov
I
,
Subbiah
V
, et al
.
Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations
.
N Engl J Med
.
2015
;
373
(
8
):
726
-
736
.
19.
Su
F
,
Viros
A
,
Milagre
C
, et al
.
RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors
.
N Engl J Med
.
2012
;
366
(
3
):
207
-
215
.
20.
Poulikakos
PI
,
Zhang
C
,
Bollag
G
,
Shokat
KM
,
Rosen
N
.
RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF
.
Nature
.
2010
;
464
(
7287
):
427
-
430
.
21.
Callahan
MK
,
Rampal
R
,
Harding
JJ
, et al
.
Progression of RAS-mutant leukemia during RAF inhibitor treatment
.
N Engl J Med
.
2012
;
367
(
24
):
2316
-
2321
.
22.
Abdel-Wahab
O
,
Klimek
VM
,
Gaskell
AA
, et al
.
Efficacy of intermittent combined RAF and MEK inhibition in a patient with concurrent BRAF- and NRAS-mutant malignancies
.
Cancer Discov
.
2014
;
4
(
5
):
538
-
545
.
23.
Hyman
DM
,
Diamond
EL
,
Vibat
CR
, et al
.
Prospective blinded study of BRAFV600E mutation detection in cell-free DNA of patients with systemic histiocytic disorders
.
Cancer Discov
.
2015
;
5
(
1
):
64
-
71
.
24.
Visser
O
,
Trama
A
,
Maynadié
M
, et al
;
RARECARE Working Group
.
Incidence, survival and prevalence of myeloid malignancies in Europe
.
Eur J Cancer
.
2012
;
48
(
17
):
3257
-
3266
.
25.
Edelbroek
JR
,
Vermeer
MH
,
Jansen
PM
, et al
.
Langerhans cell histiocytosis first presenting in the skin in adults: frequent association with a second haematological malignancy
.
Br J Dermatol
.
2012
;
167
(
6
):
1287
-
1294
.
26.
Egeler
RM
,
Neglia
JP
,
Aricò
M
,
Favara
BE
,
Heitger
A
,
Nesbit
ME
.
Acute leukemia in association with Langerhans cell histiocytosis
.
Med Pediatr Oncol
.
1994
;
23
(
2
):
81
-
85
.
27.
Aricò
M
,
Comelli
A
,
Bossi
G
,
Raiteri
E
,
Piombo
M
,
Egeler
RM
.
Langerhans cell histiocytosis and acute leukemia: unusual association in two cases
.
Med Pediatr Oncol
.
1993
;
21
(
4
):
271
-
273
.
28.
Egeler
RM
,
Neglia
JP
,
Aricò
M
, et al
;
The LCH-Malignancy Study Group of the Histiocyte Society
.
The relation of Langerhans cell histiocytosis to acute leukemia, lymphomas, and other solid tumors
.
Hematol Oncol Clin North Am
.
1998
;
12
(
2
):
369
-
378
.
29.
Wollina
U
,
Kaatz
M
,
Krönert
C
, et al
.
Cutaneous Langerhans cell histiocytosis with subsequent development of haematological malignancies. Report of two cases
.
Acta Dermatovenerol Alp Panonica Adriat
.
2006
;
15
(
2
):
79
-
84
.
30.
Jaiswal
S
,
Fontanillas
P
,
Flannick
J
, et al
.
Age-related clonal hematopoiesis associated with adverse outcomes
.
N Engl J Med
.
2014
;
371
(
26
):
2488
-
2498
.
31.
Genovese
G
,
Kähler
AK
,
Handsaker
RE
, et al
.
Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence
.
N Engl J Med
.
2014
;
371
(
26
):
2477
-
2487
.
32.
Busque
L
,
Patel
JP
,
Figueroa
ME
, et al
.
Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis
.
Nat Genet
.
2012
;
44
(
11
):
1179
-
1181
.
33.
Feldman
AL
,
Arber
DA
,
Pittaluga
S
, et al
.
Clonally related follicular lymphomas and histiocytic/dendritic cell sarcomas: evidence for transdifferentiation of the follicular lymphoma clone
.
Blood
.
2008
;
111
(
12
):
5433
-
5439
.
34.
West
DS
,
Dogan
A
,
Quint
PS
, et al
.
Clonally related follicular lymphomas and Langerhans cell neoplasms: expanding the spectrum of transdifferentiation
.
Am J Surg Pathol
.
2013
;
37
(
7
):
978
-
986
.
35.
Shao
H
,
Xi
L
,
Raffeld
M
, et al
.
Clonally related histiocytic/dendritic cell sarcoma and chronic lymphocytic leukemia/small lymphocytic lymphoma: a study of seven cases
.
Mod Pathol
.
2011
;
24
(
11
):
1421
-
1432
.
36.
Chen
W
,
Jaffe
R
,
Zhang
L
, et al
.
Langerhans cell sarcoma arising from chronic lymphocytic lymphoma/small lymphocytic leukemia: lineage analysis and BRAF V600E mutation study
.
N Am J Med Sci
.
2013
;
5
(
6
):
386
-
391
.
37.
Ratei
R
,
Hummel
M
,
Anagnostopoulos
I
, et al
.
Common clonal origin of an acute B-lymphoblastic leukemia and a Langerhans’ cell sarcoma: evidence for hematopoietic plasticity
.
Haematologica
.
2010
;
95
(
9
):
1461
-
1466
.
38.
Jamieson
CH
,
Gotlib
J
,
Durocher
JA
, et al
.
The JAK2 V617F mutation occurs in hematopoietic stem cells in polycythemia vera and predisposes toward erythroid differentiation
.
Proc Natl Acad Sci USA
.
2006
;
103
(
16
):
6224
-
6229
.
39.
Tehranchi
R
,
Woll
PS
,
Anderson
K
, et al
.
Persistent malignant stem cells in del(5q) myelodysplasia in remission
.
N Engl J Med
.
2010
;
363
(
11
):
1025
-
1037
.
40.
Pang
WW
,
Pluvinage
JV
,
Price
EA
, et al
.
Hematopoietic stem cell and progenitor cell mechanisms in myelodysplastic syndromes
.
Proc Natl Acad Sci USA
.
2013
;
110
(
8
):
3011
-
3016
.
41.
Ho
TC
,
LaMere
M
,
Stevens
BM
, et al
.
Evolution of acute myelogenous leukemia stem cell properties after treatment and progression
.
Blood
.
2016
;
128
(
13
):
1671
-
1678
.
42.
Yohe
SL
,
Chenault
CB
,
Torlakovic
EE
,
Asplund
SL
,
McKenna
RW
.
Langerhans cell histiocytosis in acute leukemias of ambiguous or myeloid lineage in adult patients: support for a possible clonal relationship
.
Mod Pathol
.
2014
;
27
(
5
):
651
-
656
.
43.
Berres
ML
,
Lim
KP
,
Peters
T
, et al
.
BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups [published correction appears in J Exp Med. 2015;212(2):281]
.
J Exp Med
.
2014
;
211
(
4
):
669
-
683
.
44.
Emile
JF
,
Diamond
EL
,
Hélias-Rodzewicz
Z
, et al
.
Recurrent RAS and PIK3CA mutations in Erdheim-Chester disease
.
Blood
.
2014
;
124
(
19
):
3016
-
3019
.

Author notes

*

M.P., E.L.D., F.C.-A., O.A.-W., and J.H. contributed equally to this study.

Sign in via your Institution