Langerhans cell histiocytosis (LCH) is a clonally derived neoplasm with a highly variable clinical course. Although LCH was once considered a disorder of immune regulation, the identification of activating mutations in the proto-oncogene BRAF-V600E in ∼50%-60% of cases and MEK and ERK phosphorylation in 100% of examined cases, has changed the definition of LCH to a dendritic cell neoplasm with a strong inflammatory component. Current international LCH trials are focused on further improving the outcome of high-risk multisystem LCH patients, by decreasing the reactivation rate, optimizing early salvage regimens, and preventing late sequelae. Anecdotal responses to vemurafenib, a BRAF-V600E inhibitor, have been reported in a few cases of LCH and Erdheim–Chester disease. However, the development of resistance, as well as the potential risks of cutaneous and pancreatic cancers in patients with BRAF-V600E-mutated melanoma treated with single inhibitors, suggest the need for prospective trials with BRAF inhibitors, alone or in combination with other inhibitors of this pathway, for patients with refractory or multiply-relapsed LCH. The recent discovery of somatic mutations in ARAF and in MAP2K1, which lead to activation of the RAS-RAF-MEK –ERK pathway in the setting of wild-type BRAF, as well as the finding that activating mutation in MAP2K1 are relatively insensitive to MEK inhibitors, suggest that a more detailed understanding of this pathway in LCH may be necessary for the development of more effective targeted therapies.

Learning Objectives
  • To gain an understanding of the advantages and disadvantages of current therapeutic options in children and adults with LCH

  • To discuss mutations in genes of the RAS-RAF-MEK-ERK pathway in patients with LCH and the implications for targeted therapies

Langerhans cell histiocytosis (LCH) is a dendritic cell (DC) neoplasm defined by the presence in the lesion of pathologic Langerhans cells (LCH cells) that are positive for CD1a, CD207 (Langerin), and S100. Despite identical histopathologic features, LCH has a diverse clinical behavior ranging from benign single system disease (SS) that can regress spontaneously to multisystem (MS) disease that can be life-threatening or a chronically reactivating form of disease that is not life-threatening but has the potential to result in significant permanent sequelae, such as diabetes insipidus (DI), growth retardation, bone pain, hearing loss, sclerosing cholangitis, and CNS neurodegenerative disease. However, because LCH-associated DI may be the first manifestation of LCH, it is unclear whether therapy to prevent reactivations will entirely prevent the permanent sequelae.

LCH can occur at any age but is more common in children of whom two-thirds have SS-LCH predominantly in bone followed by skin. Children <2 years of age with MS-LCH commonly have involvement of “risk” organs (ROs) today defined as involvement of liver, spleen, and hematopoietic system, with “risk” being the risk of death.

In adults, the mean age at diagnosis is 35 years with 10% being older than 55 years. Sixty-nine percent of adults with LCH in the Histiocyte Society Adult Registry have MS-LCH with skin and lung involvement in 51% and 62%, respectively. Of the 31% of adult patients with SS disease, lung was involved in 51%, most of whom were smokers, followed by bone and skin in 38% and 14%.1  The natural history of LCH in adults is less clear but it is thought that spontaneous regression, even in SS disease, is less likely to occur and that adult patients typically require some form of therapy depending on the extent of disease. Survival of adult patients with MS disease including skin is better than that seen in children, because of the lower number of organs involved.2  But chronicity of disease, such as in skin-LCH, may pose a special problem in adults and this coupled with the greater toxicity of standard therapeutic protocols makes targeted therapies even more interesting in the adult patients.

Earlier pathogenetic theories of LCH suggested an inappropriate activation of epidermal Langerhans cells (LCs). The benign morphology of the pathologic LCs and the lesional expression of inflammatory chemokines and cytokines have historically deemed LCH an immune dysregulatory disorder. Further, the documented examples of spontaneous self-regression, the fact that limited disease responds well to mild treatment, and in particular, the fact that infants with RO+ LCH who respond to therapy have a 50% recurrence rate but the majority of those recurrences are in low risk organs, such as bone with a 100% survival, speaks strongly against a malignant etiology, at least from a clinical point of view. On the other hand, the detection of clonal LCs from nonpulmonary LCH, the telomere length shortening and the elevated expression of non-mutant TP53, c-myc, H-ras, Ki-67, and Bcl-2 were in favor of LCH being a neoplasm,3  although not necessarily a malignancy. This hypothesis was confirmed by the discovery of recurrent somatic activating mutations of the BRAF-V600E gene in 57% of archived LCH lesions.4 BRAF has an important function in the signaling cascade, which usually begins with activation of a receptor tyrosine kinase and proceeds by phosphorylation steps through Ras to Raf to MEK and the extracellular signal-regulated kinase (ERK), which ultimately leads to modulation of gene expression.5  Evidence of MEK and ERK phosphorylation was found in 100% of examined cases in this study, regardless of BRAF mutation status, suggesting that activation of the pathway occurs in most or all LCH.4 

In support of this, additional genetic drivers of ERK pathway activation have been since identified. Nelson et al, using whole-exome sequencing of DNA isolated from purified LCH cells of 3 patients with wild-type BRAF, documented the first somatic, activating mutations (F351L and Q347_A3438del) within the kinase-encoded domain of ARAF, leading to ARAF and MEK constitutive kinase activity,6  which could also be inhibited by vemurafenib, a BRAF inhibitor. Brown et al,7  using next-generation sequencing, found that 11 of 40 (27%) cases showed somatic MAP2K1 mutations that occurred mutually exclusive of BRAF mutations, with 50% of wild-type BRAF cases showing MAP2K1 mutation. Together with the studies by Badalian-Very et al4  and Nelson et al,6  these findings suggest that most LCH patients harbor a somatic activating mutation in critical signaling steps of the RAS-RAF-MEK-ERK pathway, supporting the potential for pursuing targeted therapeutic strategies in patients with wild-type BRAF, as well as those with BRAF mutations. Nelson et al went on to test 30 LCH samples for the presence of additional genetic alterations that might cause ERK pathway activation. In 20 BRAF wild-type samples, they found 3 somatic mutations in MAP2K1 (MEK1) all of which constitutively phosphorylated ERK in in vitro kinase assays. Importantly some of the variants were resistant to the MEK inhibitor trametinib in vitro, which has obvious implications for inhibitor therapy in LCH patients. Within the entire sample set, they also found 3 specimens with mutations in MAP3K1 (MEKK1).6 

These data were the first genetic evidence supporting the notion of LCH as a myeloid neoplasm and were rapidly confirmed by other investigators who found that LCH cells with different degrees of maturation, compatible either with myeloid cell or de-differentiated LC antigens, carry the BRAF-V600E mutation.8  It remains unclear whether LCH derives from a bone marrow precursor or an abnormally reprogrammed LC. Recently, Berres et al reported that BRAF-V600E expression in tissue DCs was associated with a two-fold increased risk of recurrence but did not affect overall survival. However, patients with active high-risk LCH were found to harbor BRAF-V600E in circulating CD11c+ and CD14+ peripheral blood mononuclear cells and in bone marrow CD34+ progenitors, whereas in low-risk LCH patients the mutation was restricted to lesional CD207+ DCs.9  This study suggested that high-risk LCH originates from somatic mutation of a hematopoietic progenitor whereas low-risk LCH originates from somatic mutation of tissue-restricted precursor DCs,9  but this concept remains to be proven.

The BRAF-V600E mutations were also found to be present in 40% of the cases of adult pulmonary LCH4  correlating reasonably well with the ∼30% incidence of monoclonality found in these patients in earlier studies.10  This might suggest 2 types of adult lung LCH, one of which is the traditional polyclonal inflammatory disease of adult smokers,11  or as suggested by Bedalian-Very et al, smoking may induce BRAF V600E mutations at multiple sites throughout the lungs of susceptible smokers resulting in multiple clones.12  The absence of BRAF-V600E from samples of other rare histiocytic disorders, such as Rosai-Dorfman disease (RDD) and juvenile xanthogranuloma adds to the specificity of this mutation to LCH.4  However, BRAF-V600E mutations have been observed in 54% of patients with Erdheim-Chester disease (ECD), another rare histiocytosis, suggesting a possible common origin with LCH.13 

The treatment of LCH depends on the extent and severity of disease at diagnosis. SS disease has an excellent survival rate and can be treated with either observation (isolated skin), curettage/intralesional steroids (unifocal bone), or indomethacin, bisphosphonates or low-dose systemic chemotherapy (multifocal bone).3  MS-LCH, involving 2 or more systems, can be associated with a poor survival when there is RO+ (liver, spleen, bone marrow) involvement. Patients with MS-LCH but without risk organ involvement (RO) have an excellent survival and are usually treated with systemic chemotherapy in an attempt to reduce reactivations and prevent permanent sequelae.3  During the last 2 decades, a more aggressive chemotherapy approach has been used in RO+ MS-LCH with 2 goals, first to decrease mortality and second to reduce the 50% reactivation rate in patients who respond to front-line therapy. The early prospective European trials, conducted in Italy (AIEOP-CNR-HX 83)14  and Germany/Austria (DAL-HX 83/90),15  as well as the Japanese16  and Histiocyte Society randomized trials,17-19  showed that mortality could be reduced for the whole group of patients to <10% with single-agent or multiple-agent chemotherapy, but that multi-agent chemotherapy given for a longer duration (ie, 12 months) reduced mortality in the highest-risk group, reduced the reactivation rate and reduced the diabetes insipidus rate. Other important findings were that the prolongation of induction therapy to 12 weeks in patients who did not achieve a complete response at week 6, and an early switch to salvage therapy for those with progressive disease by week 6 (or earlier if necessary) appears to significantly decrease mortality.16,19  These findings form the basis for the currently open Histiocyte Society trial, LCH-IV, which has seven different strata. Stratum 1 will be testing in a randomized fashion whether prolonging (12 vs 24 months) and intensifying (±6-mercaptopurine) therapy for high-risk patients, and comparing 6 versus 12 month therapy for single-system disease (craniofacial bones or multifocal bone) will further reduce the rate of reactivation and the potentially devastating permanent consequences. Stratum 2 was designed for MS patients without risk organ involvement who fail first-line treatment or who initially respond but have a reactivation, and will include a 6 month re-induction with vincristine/prednisone/cytarabine followed by randomized 18 month maintenance of oral indomethacin versus oral 6-mercaptopurine and methotrexate. Stratum 3 will assess the efficacy of salvage with a cladribine/cytarabine combination in MS-LCH patients with risk organ involvement who fail to respond to first-line treatment. Stratum 4 will study the efficacy of a reduced intensity hematopoietic stem cell transplant as a salvage option for MS-LCH patients with risk organ involvement who fail stratum 1 and stratum 3 therapies. Stratum 5 will prospectively explore the effectiveness of cladribine in tumorous CNS-LCH and whether IVIg or cytarabine will impact the progression of neurodegenerative CNS-LCH. Stratum 6 will describe the natural history of single-system LCH treated by conservative methods (“wait and watch”) or local therapy. Stratum 7 will monitor the long-term outcome and the incidence of late sequelae in all LCH patients. Details of the published Histiocyte Society trials are shown in Table 1.17-19 

Table 1.

Histiocyte society trials in Langerhans cell histiocytosis

Histiocyte society trials in Langerhans cell histiocytosis
Histiocyte society trials in Langerhans cell histiocytosis

At the same time, colleagues at several institutions in the United States are testing in a non-randomized fashion the use of single-agent cytosine arabinoside,20  but a longer follow-up period will be required to fully evaluate this strategy, particularly with respect to the rates of reactivation and permanent sequelae.

Approximately 50% of patients with LCH will be refractory to induction therapy or develop reactivation of disease within 5 years.21  Patients with MS+LCH who progress early or who fail to respond after 2 courses of induction (week 12) have a very poor outcome, and their treatment has been challenging. Nucleoside analogs, such as cladribine and clofarabine, have activity against LCH because of their antiproliferative and immunomodulatory effects but also because of their efficacy against the immature myeloid precursors which have recently been suggested to be the precursor cells for LCH.9  Data on relapsed LCH patients are largely derived from pilot studies, surveys or case series. Low-dose cladribine was effective in achieving a 22% response rate in RO+ patients and 66% in RO− patients refractory to front-line therapy. However, only 3% of all patients had “no active disease” by week 24 and prolonged myelosuppression was a limiting factor.22  Clofarabine, a second-generation nucleoside analog, has shown significant activity as single-agent against disseminated LCH refractory to cladribine or cytarabine, and encouraging results with manageable toxicity have been reported by Abraham et al23  and Simko et al.24  The drug appears to be active in both high-risk and low-risk LCH and the major toxicity of myelosuppression appeared to be manageable with reduced doses and with filgrastim. Prolonged and cumulative cytopenias were not seen, possibly because of the relatively low dose of clofarabine required for therapy of LCH compared to the leukemic population. The authors concluded that the high cost of the drug could be justified by the potential to avoid hematopoietic stem cell transplantation in these refractory patients.24 

Nonetheless, the safety and efficacy of clofarabine in relapsed LCH needs to be established in a larger prospective multicenter trial. For patients with high-risk refractory disease, the most successful published salvage regimen is a combination of higher doses of cladribine plus high-dose cytarabine. In the LCH-S-2005 protocol of the Histiocyte Society, a response rate of 92% was achieved in 27 very high-risk young patients (median age 0.7 months). Four patients relapsed, some of whom were salvaged by HSCT. There were 4 deaths, 2 from toxicity and 2 from disease. This regimen proved to be very effective but is associated with significant toxicity and requires excellent supportive care.25  Rosso et al used the same combination of drugs and treated a series of 9 patients with progressive MS-LCH with a lower dose of cladribine and a much lower dose of cytarabine (100 mg/m2/d for 4 days per cycle). Six patients achieved remission and 1 a partial response, 3 patients reactivated. The overall probability of survival at 3 years was 73% (standard error 16%).26  The studies are not strictly comparable as only 5 of the 9 patients treated in the Rosso series had progressive disease at 6 weeks of LCH therapy, a known very poor prognosticator. However, these results are encouraging and suggest a worthwhile salvage strategy, particularly for sites that do not have the supportive care required for the high-dose therapy. A head-to-head comparison of the 2 strategies would be of interest, but considering the very slow accrual of LCH-S-2005, would be difficult to do.

Allogeneic hematopoietic stem cell transplant (HSCT) appears to be promising in LCH patients who fail multiple salvage regimens. Whether its beneficial effect is because of a graft-versus-LCH or the cytotoxic effect of high-dose therapy remains to be proven. The earlier published experience with myeloablative conditioning (MAC) regimens showed survival rates of ∼50% in highly resistant patients, but a transplant-related mortality (TRM) which was unacceptably high.27  Using reduced-intensity conditioning regimens (RIC), a 78% survival and a low TRM rates have been reported.28  Kudo et al more recently published the Japanese experience of 15 refractory LCH patients treated with HSCT, 5 of whom had RIC regimens. Ten year overall and disease-free survival were 73% (11/15); 80% with myeloablative and 60% with RIC, although small numbers and patient variability precluded any definitive conclusions.29  A recently published study from the United Kingdom retrospectively reviewed 87 LCH patients who underwent allogeneic transplant from 1990-2013. Prior to 2000, the TRM was 55%. Review of the patients transplanted after 2000, however, showed no significant difference in outcome between those receiving RIC compared with those receiving MAC transplants with overall survivals at 3 years of 71% and 77%, respectively (p = 0.89). The relapse rate was slightly higher after RIC transplants but most patients were salvaged with additional chemotherapy and the authors suggested that it was possible that more seriously ill patients might have received RIC. There was also no difference in GVHD rates or severity.30  RIC-HSCT is currently being tested prospectively in the ongoing Histiocyte Society LCH-IV trial for refractory MS-LCH patients who fail more than one salvage regimen.

There is no standard of care treatment for newly diagnosed adults with MS-LCH. The standard pediatric vinblastine/prednisone regimen has been associated with an increased neurotoxicity, more profound myelosuppression and more steroid toxicity in adults compared with children. A recent retrospective study of 58 adult patients with bone LCH showed an advantage for cytarabine monotherapy compared with vinblastine/prednisone and even to cladribine, in terms of response and toxicity.31  As a result, first-line monotherapy with cytarabine, etoposide, or cladribine is considered to be a preferable option in adult LCH in North America.32  Vinblastine/prednisone remains the preferred first-line option in many European centers, however.32  Other systemic therapy used in adult LCH patients, particularly those with skin-LCH, include weekly oral methotrexate, daily oral etoposide, oral azathioprine, and thalidomide.32  More intensive combination regimens, such as methotrexate, doxorubicin, cyclophosphamide, vincristine, prednisone, and bleomycin (MACOP-B), have been shown to be effective,33  but should be reserved for rare aggressive forms of adult LCH.32  Most adult experts recommend starting with cladribine in patients with risk organ or tumorous cerebral involvement, although cytarabine seems reasonable as well.32  Bisphosphonates, such as zoledronic acid, have been shown to be effective in bone LCH in adults,34  however, patients need to be counseled about risk of jaw osteonecrosis, renal and ocular adverse effects.

Adults with reactivated SS or MS-LCH can respond to single-agent cladribine, as was shown in a phase II trial.35  Patients with CNS involvement may benefit from the combination of cladribine and cytarabine, as both these drugs cross the blood–brain barrier.36  In the aggressive forms of adult LCH, RIC-HSCT has been performed successfully as well, but large studies are lacking.37  The increased toxicity to standard front line pediatric therapies and the lack of agreement on the best therapy for adult LCH patients, as well as the failure to accrue patients to the 1 prospective adult trial that has been attempted (Histiocyte Society LCH-A1 study), suggest that targeted therapies may be at least as or even more important in this patient population.

Imatinib mesylate is a potent competitive inhibitor of tyrosine kinases associated with ABL, ARG, KIT, platelet derived growth factor receptors (PDGFRA and PDGFRB), and can inhibit differentiation of CD34+ progenitors into dendritic cells. A recent study showed that a subset of patients with LCH were positive for PDGFRA, and suggested that they could potentially be treated with tyrosine kinase inhibitors.38  Imatinib has been successfully used in few cases of refractory MS-LCH with cerebral and lung involvement,39,40  but not all patients have responded. Imatinib has shown activity also in other histiocytic disorders such as ECD, and RDD, although again with mixed results.39 

BRAF inhibition

The finding of the A/BRAF and MAP2K1 mutations in LCH patients has raised the possibility of targeted therapies in histiocytic disorders, possibly through subsequent deactivation of the proliferative RAS/RAF/MEK/ERK pathway as demonstrated in BRAF-V600E/-driven melanoma treated with the BRAF-V600E inhibitor vemurafenib. The largest published series to date is by Haroche et al, who reported favorable responses to vemurafenib in 8 adult patients with refractory BRAF mutated ECD, 4 of whom had concurrent LCH. The responses were seen despite the reduction in vemurafenib dose to one-half, because of cutaneous adverse events seen in the first 3 patients. Of note, one patient developed squamous cell carcinoma. Importantly, all the others showed sustained response at a median of 10 (range, 10–16) months on verumafinib.41 

Pediatric formulations are being developed, and phase I/II trials are ongoing in pediatrics for the first generation BRAF inhibitor, dabrafenib (NCT01677741). Preliminary results were presented at the American Society of Oncology (ASCO) meeting in June 2014. One child with refractory LCH, who was treated with oral dabrafenib, continued to show stable disease at week 16.42  Anecdotal responses to vemurafenib have been reported in a 2-month-old girl with refractory MS-LCH,43  a 3-year-old boy with progressive CNS neurodegenerative LCH,44  a 45-year-old woman with refractory LCH,45  and a 90-year-old woman with severely symptomatic refractory skin LCH who responded within a few days and achieved complete remission within 6 months of starting vemurafenib.46 

The responses to BRAF-V600E inhibition are interesting for several reasons. First, there is no established standard of care for adults with severe LCH, and children with refractory LCH continue to have poor prognosis, especially those with progressive CNS neurodegeneration. Prospective clinical trials are clearly needed. However, a number of critical factors must be considered in the development of these trials for patients with histiocytic disorders. Treatment of melanoma patients with vemurafenib has been associated with the development of de novo squamous cell carcinoma in as many as 50% of treated patients,47  and more recently, secondary pancreatic cancers in patients with melanoma and BRAF-V600E mutations treated with single inhibitors.48  Although these can be perhaps tolerated in adults with life-threatening melanomas, their incidence is less acceptable in children with non life-threatening LCH. Second, several trials with BRAF inhibitors have shown that not all neoplasms with BRAF-V600E mutations respond similarly. This is not surprising in view of the results discussed earlier of some variants of MAP2K1 mutations being shown to result in resistance to MEK inhibition. These findings indicate the need to further define the molecular context in which the BRAF mutation exists.6,49  Third, determination of the type of the RAF mutation is very important as not all mutations respond to inhibition, and some may get stimulated by inhibitors directed to the BRAF-V600E mutation potentially causing tumor progression,50  such as has been seen in patients with mutations which result in high levels of RAS. Fourth, the duration of therapy has not been established and prolonged therapy may be needed, possibly even longer for the histiocytic disorders than for melanoma patients, and finally resistance mechanisms may rapidly develop to overcome initial sensitivity to BRAF inhibition.51  Combination drug regimens, therefore, such as the combination of BRAF and other ERK pathway inhibitors or BRAF/MEK inhibitors with chemotherapy, may be more effective in preventing development of resistance and may additionally prove to be less toxic,52  including less tumorigenic.

The finding that BRAF-V600E mutations were common in LCH lesional cells has led to an explosion of interest in the biology of LCH and other histiocytic disorders. However, the increased understanding of LCH biology has not yet lead to any change in risk stratification or front-line treatment strategies. More studies are needed to correlate BRAF, ARAF, or MAP2K1 mutations with clinical risk status while awaiting the results of the ongoing clinical trials utilizing BRAF inhibitors. Experience with melanoma patients suggests that future clinical trials will most likely involve a combination of targeted agents or targeted agents combined with chemotherapy to ensure optimal efficacy, as well as safety, of this promising new approach.

Sheila Weitzman, The Hospital for Sick Children, 555 University Ave, Toronto, ON, M5G 1X8 Canada; Phone: 416-813-7654; Fax: 416-813-5327; e-mail: sheila.weitzman@sickkids.ca.

1
Arico'
 
M
Girschikofsky
 
M
Genereau
 
T
, et al. 
Langerhans cell histiocytosis in adults: report from the International Registry of the Histiocyte Society
Eur J Cancer
2003
, vol. 
39
 
16
(pg. 
2341
-
2348
)
2
Krafchik
 
B
, et al. 
Weitzman
 
S
Egeler
 
M
Histiocytosis of skin in children and adults
Histiocytic disorders of children and adults
2005
Ed 1
Cambridge
Cambridge University Press
(pg. 
130
-
147
)
3
Abla
 
O
Egeler
 
RM
Weitzman
 
S
Langerhans cell histiocytosis: Current concepts and treatments
Cancer Treatm Rev
2010
, vol. 
36
 
34
(pg. 
354
-
359
)
4
Badalian-Very
 
G
Vergilio
 
JA
Degar
 
BA
, et al. 
Recurrent BRAF mutations in Langerhans cell histiocytosis
Blood
2010
, vol. 
116
 
11
(pg. 
1919
-
1923
)
5
Montagut
 
C
Settleman
 
J
Targeting the RAF-MEK-ERK pathway in cancer therapy
Cancer Lett
2009
, vol. 
283
 
2
(pg. 
125
-
134
)
6
Nelson
 
DS
Quispel
 
W
Badalian-very
 
G
, et al. 
Somatic activating ARAF mutations in Langerhans cell histiocytosis
Blood
2014
, vol. 
123
 
20
(pg. 
3152
-
3155
)
7
Brown
 
NA
Furtado
 
LV
Betz
 
BL
, et al. 
High prevalence of somatic MAP2K1 mutations in BRAF V600E negative Langerhans cell histiocytosis
Blood
2014
, vol. 
124
 
10
(pg. 
1655
-
1658
)
8
Sahm
 
F
Capper
 
D
Preusser
 
M
, et al. 
BRAF V600E mutant protein is expressed in cells of variable maturation in Langerhans cell histiocytosis
Blood
2012
, vol. 
120
 
12
(pg. 
e28
-
e34
)
9
Berres
 
ML
Lim
 
KP
Peters
 
T
, et al. 
BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups
J Exp Med
2015
, vol. 
212
 
2
pg. 
281
 
10
Yousem
 
SA
Colby
 
TV
Chen
 
YY
Chen
 
WG
Weiss
 
LM
Pulmonary Langerhans cell histiocytosis: molecular analysis of clonality
Am J Surg Pathol
2001
, vol. 
25
 
5
(pg. 
630
-
636
)
11
Yousem
 
SA
Dacic
 
S
Nikiforov
 
YE
, et al. 
Pulmonary Langerhans cell histiocytosis: profiling of multifocal tumors using next-generation sequencing identifies concordant occurrence of BRAF V600E mutations
Chest
2013
, vol. 
143
 
6
(pg. 
1679
-
1684
)
12
Badalian-Very
 
G
Vergilio
 
JA
Degar
 
BA
Rodriguez-Galindo
 
C
Barrett
 
JR
Recent advances in the understanding of Langerhans cell histiocytosis
Br J Haematol
2011
, vol. 
156
 
2
(pg. 
163
-
172
)
13
Emile
 
JF
Charlotte
 
F
Amoura
 
Z
, et al. 
BRAF mutations in Erdheim-Chester disease
J Clin Oncol
2013
, vol. 
31
 
3
pg. 
398
 
14
Ceci
 
A
Terlizzi
 
MD
Colella
 
R
, et al. 
Langerhans cell histiocytosis in childhood: results from the Italian cooperative AIEOP-CNR-HX 83 study
Med Pediatr Oncol
1993
, vol. 
21
 
4
(pg. 
259
-
264
)
15
Gadner
 
H
Heitger
 
A
Grois
 
N
, et al. 
Treatment strategy for disseminated Langerhans cell histiocytosis
Med Pediatr Oncol
1994
, vol. 
23
 
2
(pg. 
72
-
80
)
16
Morimoto
 
A
Ikushima
 
S
Kinugawa
 
N
, et al. 
Improved outcome in the treatment of pediatric multifocal Langerhans cell histiocytosis. Results from the Japan Langerhans cell histiocytosis study group-96 protocol
Cancer
2006
, vol. 
107
 
3
(pg. 
613
-
619
)
17
Gadner
 
H
Grois
 
N
Arico
 
M
, et al. 
A randomized trial of treatment for multisystem Langerhans cell histiocytosis
J Pediatr
2001
, vol. 
138
 
5
(pg. 
728
-
734
)
18
Gadner
 
H
Grois
 
N
Potschger
 
U
, et al. 
Improved outcome in multisystem Langerhans cell histiocytosis is associated with therapy intensification
Blood
2008
, vol. 
111
 
5
(pg. 
2556
-
2562
)
19
Gadner
 
H
Minkov
 
M
Grois
 
N
, et al. 
Therapy prolongation improves outcome in multisystem Langerhans cell histiocytosis
Blood
2013
, vol. 
121
 
25
(pg. 
5006
-
5014
)
20
Simko
 
S
Abhyankar
 
H
Lupo
 
P
, et al. 
Cytarabine monotherapy for de novo and recurrent Langerhans cell histiocytosis
30th Annual Histiocyte Society Meeting
October 28-30, 2014
Toronto, Canada
 
Scientific Session IV, p. 59
21
Minkov
 
M
Steiner
 
M
Potschger
 
U
, et al. 
Reactivations in multisystem Langerhans cell histiocytosis: data of the international LCH registry
J Pediatr
2008
, vol. 
153
 
5
(pg. 
700
-
705
)
22
Weitzman
 
S
Braier
 
J
Donadieu
 
J
, et al. 
2′-Chlorodeoxyadenosine (2-CDA) as salvage therapy for Langerhans cell histiocytosis (LCH). Results of the LCH-98 protocol of the Histiocyte Society
Pediatr Blood Cancer
2009
, vol. 
53
 
7
(pg. 
1271
-
1276
)
23
Abraham
 
A
Alsultan
 
A
Jeng
 
M
, et al. 
Clofarabine Salvage therapy for refractory high risk Langerhans cell Histiocytosis
Pediatr Blood Cancer
2013
, vol. 
60
 
6
(pg. 
E19
-
E22
)
24
Simko
 
SJ
Tran
 
HD
Jones
 
J
, et al. 
Clofarabine salvage therapy in refractory multifocal histiocytic disorders, including Langerhans cell histiocytosis, juvenile xanthogranuloma and Rosai-Dorfman disease
Pediatr Blood Cancer
2014
, vol. 
61
 
3
(pg. 
479
-
487
)
25
Donadieu
 
J
Bernard
 
F
van Noesel
 
M
, et al. 
Cladribine and cytarabine in refractory multisystem Langerhans cell histiocytosis: results of an international phase 2 study
Blood
2015
, vol. 
126
 
12
(pg. 
1415
-
1423
)
26
Rossi
 
D
Amaral
 
D
Latella
 
A
, et al. 
Reduced doses of cladribine and cytarabine regimen was effective and well tolerated in patients with refractory-risk multisystem Langerhans cell histiocytosis
British J Hematol
 
Prepublished on May 5, 2015, as DOI 10.1111/bjh.13475
27
Caselli
 
D
Arico'
 
M
EBMT paediatric working party. The role of BMT in childhood histiocytosis
Bone Marrow Transplant
2008
, vol. 
41
 
52
(pg. 
S8
-
S13
)
28
Steiner
 
M
Matthes-Martin
 
S
Attarbaschi
 
A
, et al. 
Improved outcome of treatment-resistant high-risk Langerhans cell histiocytosis after allogeneic stem cell transplantation with reduced-intensity conditioning
Bone Marrow Transplant
2005
, vol. 
36
 
3
(pg. 
215
-
225
)
29
Kudo
 
K
Ohga
 
S
Morimoto
 
A
, et al. 
Improved outcome of refractory Langerhans cell histiocytosis in children with hematopoietic stem cell transplantation in Japan
Bone Marrow Transplant
2010
, vol. 
45
 
5
(pg. 
901
-
906
)
30
Veys
 
PA
Nanduri
 
V
Baker
 
KS
, et al. 
Haematopoietic stem cell transplantation for refractory Langerhans cell histiocytosis: outcome by intensity of conditioning
British J Haematol
2015
, vol. 
169
 
5
(pg. 
711
-
718
)
31
Cantu
 
MA
Lupo
 
PJ
Bilgi
 
M
Hicks
 
MJ
Allen
 
CE
McClain
 
KL
Optimal therapy for adults with Langerhans cell histiocytosis bone lesions
PLoS One
2012
, vol. 
7
 
8
pg. 
e43257
 
32
Girschikofsky
 
M
Arico'
 
M
Castillo
 
D
, et al. 
Management of adult patients with Langerhans cell histiocytosis: recommendation from an expert panel on behalf of Euro-Histio-Net
Orphanet J Rare Dis
2013
, vol. 
8
 pg. 
72
 
33
Derenzini
 
E
Fina
 
MP
Stefoni
 
V
, et al. 
MACOP-B regimen in the treatment of adult Langerhans cell histiocytosis: experience on seven patients
Ann Oncol
2010
, vol. 
21
 
6
(pg. 
1173
-
1178
)
34
Montella
 
L
Merola
 
C
Merola
 
G
Petillo
 
L
Palmieri
 
G
Zoledronic acid in treatment of bone lesions by Langerhans cell histiocytosis
J Bone Miner Metab
2009
, vol. 
27
 
1
(pg. 
110
-
113
)
35
Saven
 
A
Burian
 
C
Cladribine activity in adult Langerhans cell histiocytosis
Blood
1999
, vol. 
93
 
12
(pg. 
4125
-
4130
)
36
McClain
 
KL
drug therapy for the treatment of Langerhans cell histiocytosis
Expert Opin Pharmacother
2005
, vol. 
6
 
14
(pg. 
2435
-
2441
)
37
Ingram
 
W
Desai
 
SR
Gibbs
 
JS
Mufti
 
G
Reduced-intensity conditioned allogeneic haematopoietic transplantation in an adult with Langerhans cell histiocytosis and thrombocytopenia with absent radii
Bone Marrow Transplant
2006
, vol. 
37
 
7
(pg. 
713
-
715
)
38
Caponetti
 
GC
Miranda
 
RN
Althof
 
PA
, et al. 
Immunohistochemical and molecular cytogenetic evaluation of potential targets for tyrosine kinase inhibitors in Langerhans cell histiocytosis
Hum Pathol
2012
, vol. 
43
 
12
(pg. 
2223
-
2228
)
39
Janku
 
F
Amin
 
HM
Yang
 
D
Garrido-Laguna
 
I
Trent
 
JC
Kurzrock
 
R
Response of histiocytoses to imatinib mesylate: fire to ashes
J Clin Oncol
2010
, vol. 
28
 
31
(pg. 
e633
-
e636
)
40
Montella
 
L
Insabato
 
L
Palmieri
 
G
Imatinib mesylate for cerebral Langerhans cell histiocytosis
N Engl J Med
2004
, vol. 
351
 
10
(pg. 
1034
-
1035
)
41
Haroche
 
J
Cohen-Aubart
 
F
Emile
 
JF
, et al. 
Reproducible and sustained efficacy of targeted therapy with vemurafenib in patients with BRAFV600E-Mutated Erdheim-Chester disease
J Clin Oncol
2015
, vol. 
33
 
5
(pg. 
411
-
418
)
42
Kieran
 
MW
Cohen
 
KJ
Doz
 
F
, et al. 
Complete radiographic responses in pediatric patients with BRAFV600E–positive tumors including high-grade gliomas: preliminary results of an ongoing phase 1/2a safety and pharmacokinetics (PK) study of dabrafenib
J Clin Oncol
2014
, vol. 
32
 
suppl
pg. 
5s
  
Abstract 10056
43
Héritier
 
S
Jehanne
 
M
Leverger
 
G
, et al. 
Vemurafenib use in an infant for high-risk Langerhans cell histiocytosis
JAMA Oncol
2015
, vol. 
1
 
6
(pg. 
836
-
838
)
44
Donadieu
 
J
Armari-Alla
 
C
Templier
 
I
, et al. 
First use of vemurafenib in children LCH with neurodegenerative LCH
30th Annual Histiocyte Society Meeting
October 28-30, 2014
Toronto, Canada
 
Abstract 3, p. 35
45
Charles
 
J
Beani
 
JC
Fiandrino
 
G
Busser
 
B
Major response to vemurafenib in patient with severe cutaneous Langerhans cell histiocytosis
J Am Acad Dermatol
2014
, vol. 
71
 
3
(pg. 
e97
-
e99
)
46
Bubolz
 
AM
Weissinger
 
SE
Stenzinger
 
A
, et al. 
Potential clinical implications of BRAF mutations in histiocytic proliferations
Oncotarget
2014
, vol. 
5
 
12
(pg. 
4060
-
4070
)
47
Sloot
 
S
Fedorenko
 
IV
Smalley
 
KS
, et al. 
Long-term effects of BRAF inhibitors in melanoma treatment: friend or foe?
Expert Opin Pharmacother
2014
, vol. 
15
 
5
(pg. 
589
-
592
)
48
Grey
 
A
Cooper
 
A
McNeil
 
C
, et al. 
Progression of KRAS mutant pancreatic adenocarcinoma during vemurafenib treatment in a patient with metastatic melanoma
Intern Med J
2014
, vol. 
44
 
6
(pg. 
597
-
600
)
49
Rahman
 
MA
Salajegheh
 
A
Smith
 
RA
Lam
 
AK
BRAF inhibitor therapy for melanoma, thyroid and colorectal cancers: development of resistance and future prospects
Curr Cancer Drug Targets
2014
, vol. 
14
 
2
(pg. 
128
-
143
)
50
Lito
 
P
Rosen
 
N
Solit
 
DB
Tumor adaptation and resistance to RAF i(2)nhibitors
Nat Med
2013
, vol. 
19
 
11
(pg. 
1401
-
1409
)
51
Van Allen
 
EM
Wagle
 
N
Sucker
 
A
, et al. 
The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma
Cancer Discov
2014
, vol. 
4
 
1
(pg. 
94
-
109
)
52
Flaherty
 
KT
Robert
 
C
Hersey
 
P
, et al. 
Improved survival with MEK inhibition in BRAF-mutated melanoma
N Engl J Med
2012
, vol. 
367
 
2
(pg. 
107
-
114
)

Competing Interests

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

Author notes

Off-label drug use: Braf inhibitors as therapy for LCH.