Hairy cell leukemia (HCL) is a distinct clinicopathological entity whose underlying genetic lesion has remained a mystery for over half a century. The BRAF V600E mutation is now recognized as the causal genetic event of HCL because it is somatic, present in the entire tumor clone, detectable in almost all cases at diagnosis (encompassing the whole disease spectrum), and stable at relapse. BRAF V600E leads to the constitutive activation of the RAF-MEK-extracellular signal-regulated kinase (ERK) signaling pathway which represents the key event in the molecular pathogenesis of HCL. KLF2 and CDNK1B (p27) mutations may cooperate with BRAF V600E in promoting leukemic transformation. Sensitive molecular assays for detecting BRAF V600E allow HCL (highly responsive to purine analogs) to be better distinguished from HCL-like disorders, which are treated differently. In vitro preclinical studies on purified HCL cells proved that BRAF and MEK inhibitors can induce marked dephosphorylation of MEK/ERK, silencing of RAF-MEK-ERK pathway transcriptional output, loss of the HCL-specific gene expression profile signature, change of morphology from “hairy” to “smooth,” and eventually apoptosis. The overall response rate of refractory/relapsed HCL patients to the BRAF inhibitor vemurafenib approached 100%, with 35% to 40% complete remissions (CRs). The median relapse free-survival was about 19 months in patients who had achieved CR and 6 months in those who had obtained a partial response. Future therapeutic perspectives include: (1) combining BRAF inhibitors with MEK inhibitors or immunotherapy (anti-CD20 monoclonal antibody) to increase the percentage of CRs and (2) better understanding of the molecular mechanisms underlying resistance of HCL cells to BRAF inhibitors.

Hairy cell leukemia (HCL) is a rare mature B-cell lymphoid malignancy that is listed as a distinct entity in the World Health Organization (WHO) classification of lymphohemopoietic tissues.1  Characteristically, HCL occurs in older patients (median age about 60 years) with male predominance (male:female ratio = 4:1) and usually presents with pancytopenia and monocytopenia associated with hepatosplenomegaly in the absence of lymph node enlargement.1  This clinical presentation reflects the marked infiltration of bone marrow (BM), spleen, and liver (usually with sparing of lymph nodes) by leukemic cells with wide cytoplasm and long, slender cell-surface projections. This hairy morphology can be appreciated only by careful examination of smear preparations and gives the disease its vivid, descriptive name.2  The HCL cells are positive for the B-cell–associated antigens CD19, CD20, and CD22 and typically coexpress the CD103, CD25, and CD11c molecules.1  As compared to other lymphoid malignancies, HCL responds well to therapy with purine analogs (ie, cladribine and pentostatin).3 

In spite of such peculiar clinicopathological features, the genetic lesion underlying HCL has remained a mystery for over half a century since the original description of the disease in 1958 under the term of leukemic reticuloendotheliosis.4  A major obstacle to investigating the genome and functional features of HCL has been the inability to collect enough numbers of leukemic cells for analysis. In fact, HCL patients usually present with pancytopenia and a low number of circulating tumor cells; moreover, the BM aspiration frequently results in dry-tap and leukemic cells cannot usually be collected from markedly enlarged spleen because splenectomy is rarely performed in the purine analogs era. Additional problems in the genomic and functional studies of HCL are the very low proliferative index of leukemic cells and the lack of true HCL cell lines5,6  as well as of faithful animal models of the disease.7 

Cytogenetic/fluorescence in situ hybridization, comparative genomic hybridization, and microarray single-nucleotide polymorphism genotyping did not identify chromosomal translocations or copy-number aberrations consistently associated with HCL at significant frequencies (≥10% of cases).8-12  Both gene expression (GEP)13  and microRNA14  profiles revealed a unique molecular signature that, at least in part, may explain some distinctive functional features of HCL cells, such as the hairy morphology, the associated BM fibrosis, the typical adhesion properties, and the selective homing to particular anatomical sites.15  High-density genome-wide single-nucleotide polymorphism genotyping clearly showed that HCL has a remarkable stable genome,16  as compared with other B-cell neoplasms. However, none of the above approaches was able to unravel any recurrent genetic alteration responsible for the disease.

Using a whole-exome sequencing approach, in 2011 we discovered the BRAF V600E mutation as the causal genetic event of HCL.17  The characteristics of this mutation and its role in the pathogenesis of HCL are explained in more detail in the following sections.

Unique characteristics of the BRAF V600E mutation in HCL

BRAF V600E is a point mutation (substitution of a thymine with adenine at position 1799 on exon 15) that results in the change of amino acid 600 from valine (V) to glutamate (E). The BRAF V600E mutation causes constitutive activation of the RAS-RAF-MEK-ERK signaling pathway, independent of extracellular signals (see next section). Notably, among B-cell neoplasms, HCL is the only pathological condition in which the molecular pathogenesis is mediated by an activating point mutation of a kinase-encoding gene.17 BRAF mutations have been detected in >97% of HCL patients so far investigated.17-36 

Only 1 study26  showed a lower frequency of BRAF mutations (79%) in 53 bona fide HCL patients studied. However, 5 of 11 BRAF cases had a IGHV4-34 rearrangement that is in general absent in classical HCL. Unlike that observed in other BRAF-mutated tumors that may carry mutations other than V600E (eg, V600K and V600R in melanoma), virtually all HCL patients were V600E+.17-36  In 2 V600E HCL patients, novel, potentially functionally relevant mutations in exon 11 (F468C and D449E) were detected.35 

There are a number of findings suggesting that BRAF V600E is the disease-defining genetic event in HCL. First, this mutation, with very rare exceptions, is detectable at diagnosis in virtually all HCL patients,17-36  encompassing the whole clinical spectrum of the disease, including cases presenting with leukocytosis or without splenomegaly. Interestingly, the BRAF V600E mutation could be detected in anatomical sites unusually involved with HCL, such as lymph nodes,37  again pointing to its key role in the pathogenesis of the disease. Second, the BRAF V600E mutation is very stable over time, being consistently detectable at relapse, even after decades from the initial diagnosis. Third, in each individual patient, the mutation appears to be present in the entire tumor cell clone as demonstrated by the percentage of allelic variants at next-generation sequencing and by immunostaining of BM biopsies with an anti-BRAF V600E monoclonal antibody.33  Finally, among B-cell chronic lymphoproliferative disorders, the BRAF V600E mutation is very specific for HCL.17,21,38 

BRAF V600E–mediated constitutive activation of the RAS-RAF-MEK-ERK pathway is the key event in the molecular pathogenesis of HCL

The BRAF protein (encoded by the BRAF proto-oncogene at chromosome 7q24) is a member of the serine–threonine kinase RAF family (comprising RAF-1/CRAF, ARAF, BRAF). BRAF is a key component of the RAS-RAF-MEK-ERK signaling pathway (Figure 1). RAF proteins share many structural and biological properties but they differ in basal kinase activity that is, in turn, depending on the phosphorylation status of these molecules. In particular, BRAF appears to have a basal kinase activity higher than CRAF because serine 445 is constitutively phosphorylated in BRAF whereas the homologous site (serine 338) in CRAF is not and needs to be phosphorylated in order to exploit maximal RAS-induced activation.39,40  This explains why a single mutation at codon 600 (V600E) results in constitutive activation of BRAF but not of CRAF41  and also why BRAF is the most frequently mutated RAF protein in human cancers.42 

Figure 1

The RAS-RAF-MEK-ERK signaling pathway. (A) The RAS-RAF-MEK-ERK signaling pathway is physiologically triggered by the binding of a surface receptor tyrosine kinase (RTK) to its ligand. This activates RAS and, in turn, RAFs (BRAF and, not shown, CRAF). BRAF-CRAF heterodimers phosphorylate the MEK1 and MEK2 kinases (pMEK), which in turn phosphorylate ERK1 and ERK2 (pERK). Then, active ERKs phosphorylate several substrates in the cytoplasm (not shown) as well as in the nucleus, where they initiate a transcriptional response (eg, through the activator protein 1 transcription complex) that includes cyclin-D1 upregulation and that promotes cell survival and proliferation, as well as feedback inhibitory mechanisms (not shown) to counterregulate pathway activity. The latter, if uncontrolled, can result in neoplastic transformation. Indeed, the BRAF V600E mutation renders BRAF constitutively active independent from upstream regulatory signals and from heterodimerization with CRAF. (B) In vivo activation of the BRAF-MEK-ERK pathway in HCL patients is illustrated by the expression of pERK (red) and cyclin-D1 (nuclear, brown) by BM leukemic hairy cells (counterstained with hematoxylin in the top panel and double stained for the surface B-cell marker CD20 (surface, blue) in the bottom panel). Pictures of immunohistochemical stainings were taken with the 40×/0.85 objective (U Plan Apo) of a BX61 microscope equipped with a DP71 digital camera, using cell^B x.y acquisition software (all from Olympus).

Figure 1

The RAS-RAF-MEK-ERK signaling pathway. (A) The RAS-RAF-MEK-ERK signaling pathway is physiologically triggered by the binding of a surface receptor tyrosine kinase (RTK) to its ligand. This activates RAS and, in turn, RAFs (BRAF and, not shown, CRAF). BRAF-CRAF heterodimers phosphorylate the MEK1 and MEK2 kinases (pMEK), which in turn phosphorylate ERK1 and ERK2 (pERK). Then, active ERKs phosphorylate several substrates in the cytoplasm (not shown) as well as in the nucleus, where they initiate a transcriptional response (eg, through the activator protein 1 transcription complex) that includes cyclin-D1 upregulation and that promotes cell survival and proliferation, as well as feedback inhibitory mechanisms (not shown) to counterregulate pathway activity. The latter, if uncontrolled, can result in neoplastic transformation. Indeed, the BRAF V600E mutation renders BRAF constitutively active independent from upstream regulatory signals and from heterodimerization with CRAF. (B) In vivo activation of the BRAF-MEK-ERK pathway in HCL patients is illustrated by the expression of pERK (red) and cyclin-D1 (nuclear, brown) by BM leukemic hairy cells (counterstained with hematoxylin in the top panel and double stained for the surface B-cell marker CD20 (surface, blue) in the bottom panel). Pictures of immunohistochemical stainings were taken with the 40×/0.85 objective (U Plan Apo) of a BX61 microscope equipped with a DP71 digital camera, using cell^B x.y acquisition software (all from Olympus).

Close modal

Under physiological conditions, the RAS-RAF-MEK-ERK signaling pathway can be activated through binding of a variety of receptors (including tyrosine kinases) with their cognate ligands43,44  (Figure 1). These interactions trigger, as first events, the recruitment of the adaptor molecules Grb2 to the inner part of the cell membrane and its binding to the guanine nucleotide-exchange factor SOS. Activated SOS, in turn, displaces guanosine diphosphate from RAS, promoting the formation of guanosine triphosphate–RAS.44  Then, RAS in its activated guanosine triphosphate–bound state recruits BRAF to the cell membrane where it is phosphorylated at threonine 599 and serine 602.44  Activated (phosphorylated) BRAF can form BRAF-CRAF heterodimers45,46  that in turn lead to phosphorylation and activation of MEK1 and MEK2.44  Finally, activated MEK1/2 phosphorylate extracellular signal-regulated kinases 1 and 2 (ERK1 and ERK2). Then, activated (phosphorylated) ERK can translocate from the cytoplasm into the nucleus where it phosphorylates the transcription factors activator protein 1 and nuclear factor of activated T cells with impact on transcription of many target genes that promote cell-cycle progression and survival.47,48 

The V600E phosphomimetic substitution occurs within the BRAF-activation segment and functionally results in a marked enhancement of its kinase activity in a constitutive manner.49  BRAF mutants (especially those carrying V600E) can phosphorylate ERK as monomers and in a RAS-independent manner.49  Thus, similarly to what occurs in other BRAF-mutated tumors (including melanomas and papillary thyroid carcinomas), BRAF V600E in HCL constitutively activate the RAF-MEK-ERK pathway,17,50  leading to enhanced survival. The implication of the RAF-MEK-ERK pathway activation in the molecular pathogenesis of HCL is also supported by immunohistochemical and western blot studies showing that, in the presence of the BRAF V600E mutation, the downstream targets MEK and ERK are phosphorylated and that such a status can be reverted by specific BRAF inhibitors.17,50-52  Notably, constitutive ERK activation in HCL was previously hypothesized to provide a survival signal for the leukemic hairy cells53  in antagonism with the proapoptotic effect of p38-JNK activation (triggered by the interaction of HCL cells with the vitronectin-positive cells in the splenic red pulp).53,54  Moreover, the constitutive activation of the RAF-MEK-ERK pathway likely contributes to some of the immunophenotypic features of HCL, such as expression of cyclin D155-57  and negativity for p27.58-60 

There is still scarce information on the genetic events that may contribute to development of HCL by cooperating with BRAF V600E. Recently, missense mutations of KLF2, a transcription factor important for the homeostasis and differentiation of peripheral B-cell subsets including marginal zone B cells, were reported in 4 of 24 HCL cases (16%), as well as in 19 of 96 splenic marginal zone lymphomas (20%), and 17 of 154 diffuse large B-cell lymphomas (11%).61  Clonal KLF2 mutants, which were mostly monoallelic, also included disruptive variants (eg, nonsense or frame-shifting alleles), and the loss-of-function nature of 1 missense mutation was experimentally documented. Soon after, using whole-exome sequencing, Dietrich et al identified recurrent mutations involving the EZH2, ARID1A, and the cell-cycle inhibitor CDKN1B (p27) genes in the genome of purine analog-refractory HCL patients.62  In particular, clonal deleterious CDKN1B mutations were detected in 13 of 81 cases (16%), strongly suggesting that they may represent driver events in the pathogenesis of HCL. Because CDNK1B is involved in the control of the cell-cycle progression, these findings suggest that alterations in the regulation of cell cycle and senescence may play an important role in the pathogenesis of HCL.

Cell of origin of HCL

GEP studies had previously shown that HCL derives from the transformation of a memory B cell.13  Similarly to normal memory B cells, HCL cells showed a conservation in proliferation, apoptosis, and DNA metabolism programs, but clearly differed from them in the expression of genes controlling cell adhesion and response to chemokines.13 

More recently, it has been reported that hemopoietic stem cells (HSCs) or B-cell lymphoid progenitors from HCL patients carry the BRAF V600E mutation.7  Transplantation of BRAF V600E-mutant HSCs from an HCL patient into immunodeficient mice resulted in their stable engraftment, revealing the functional self-renewal capacity of HCL HSCs. However, none of the transplanted animals developed a full HCL phenotype.7  At present, it remains unclear whether the development of the HCL phenotype requires a permissive epigenetic background (present only in a particular stage of cell differentiation) and/or the acquirement of further genetic lesions, for example, mutations of KLF261  or CDKN1B/p27.62 

Hairy morphology is dependent upon BRAF V600E-mutant–mediated activation of the RAS-RAF-MEK-ERK pathway

Recently, in vitro studies on purified HCL cells using BRAF and MEK inhibitors52,63  have clearly shown that “hairyness” represents an intrinsic property of HCL cells which is closely related to the deregulated BRAF activity, rather than reflecting the derivation from an unidentified subset of normal memory B cells with “hairy” appearance or being the result of microenvironmental stimuli (as also supported by the observation that HCL cells retain their “hairy” morphology even after several days of culture in vitro). Acquiring this unique feature may have a finalistic effect, that is, to increase the cell surface area of HCL cells in order to favor their interaction with other cells or with ligands in the extracellular matrix.

Notably, in vitro exposure of primary HCL cells to BRAF and MEK inhibitors resulted in conversion of their morphology from “hairy” to “smooth”52  (Figure 2). The inhibitors exerted their activity specifically on HCL because the morphology of primary leukemic cells from patients with HCL-like disorders was not changed.52  Analysis of how BRAF and MEK inhibitors affected the unique GEP of HCL clearly highlighted β-actin and LST1 (which were both significantly downregulated) as the most likely candidate molecules in determining the hairy morphology of HCL cells. This is consistent with a previous observation that β-actin, in its polymerized form (F-actin), is predominantly expressed in the cortical cytoskeleton of HCL, suggesting a role in sustaining the filamentous membrane projections.64  Moreover, LST1 is important in forming actin-containing long-membrane protrusions.65,66 

Figure 2

Loss of the hairy morphology upon BRAF inhibition. Leukemic cells purified from patients with (A) HCL or (B) HCL-v were exposed in vitro to vemurafenib (vemuraf.) 1 µM or drug vehicle as control (Ctrl.) for 2 or 3 days, and then costained with phalloidin (labeling in green the cytoskeleton of hairy projections rich in F-actin), Annexin-V (ANXA5, labeling in red the dying cells), and Draq5 (labeling the nucleus), followed by confocal fluorescence microscopy showing 2-dimensional images of representative cells and their corresponding 3-dimensional reconstruction. After blocking BRAF V600E with vemurafenib, HCL (but not HCL-v) cells become smoother and smaller while being still alive (ANXA5 HCL cells showing severely shortened green surface projections). This hair loss is then followed by apoptosis of HCL (but not HCL-v) cells, which become ANXA5+ (red) and lose any phalloidin-positive (green) cytoskeletal structures. These images were taken with a LSM510 laser-scanning confocal microscope (Zeiss) equipped with laser emission lines at 458, 488, 543, and 633 nm, using an objective Plan-Apocromat 63×/1.4 NA with oil immersion and LSM510 Zeiss software.

Figure 2

Loss of the hairy morphology upon BRAF inhibition. Leukemic cells purified from patients with (A) HCL or (B) HCL-v were exposed in vitro to vemurafenib (vemuraf.) 1 µM or drug vehicle as control (Ctrl.) for 2 or 3 days, and then costained with phalloidin (labeling in green the cytoskeleton of hairy projections rich in F-actin), Annexin-V (ANXA5, labeling in red the dying cells), and Draq5 (labeling the nucleus), followed by confocal fluorescence microscopy showing 2-dimensional images of representative cells and their corresponding 3-dimensional reconstruction. After blocking BRAF V600E with vemurafenib, HCL (but not HCL-v) cells become smoother and smaller while being still alive (ANXA5 HCL cells showing severely shortened green surface projections). This hair loss is then followed by apoptosis of HCL (but not HCL-v) cells, which become ANXA5+ (red) and lose any phalloidin-positive (green) cytoskeletal structures. These images were taken with a LSM510 laser-scanning confocal microscope (Zeiss) equipped with laser emission lines at 458, 488, 543, and 633 nm, using an objective Plan-Apocromat 63×/1.4 NA with oil immersion and LSM510 Zeiss software.

Close modal

On the contrary, expression of genes that were previously hypothesized to play a role in controlling HCL morphology,15  including growth-arrest specific 7 (GAS7, required for assembling actin filaments and neurite outgrowth in neuronal cells67 ) or the CD9 molecule (which colocalizes with F-actin and is also involved in neurite growth68 ), were not affected by the exposure of HCL cells to the BRAF and MEK inhibitors.52  Thus, the role of these and other molecules previously associated with the hairy phenotype, such as pp52(LSP1)69,70  or the member of the Rho family of small GTPases (CDC42, RAC1, RHOA),71,72  in determining the morphology of HCL cells remains unclear.

The diagnosis of HCL is mainly based upon clinical/laboratory findings (usually splenomegaly associated with pancytopenia and monocytopenia) and the typical morphology and immunophenotype of leukemic cells (bright Ig+, CD20+, CD22+, CD11+, CD25+, CD103+, CD123+).1,73  Immunohistochemical staining for Annexin A1 (ANXA1)74  and/or the search for the BRAF V600E mutation18,21,25,38  are very helpful in confirming the diagnosis.

Immunostaining for Annexin A1

ANXA1 was selected as a potential HCL diagnostic marker because at GEP it was found to be specifically upregulated in HCL.13  Immunostaining of BM biopsies with a monoclonal antibody directed against a fixative-resistant epitope of the ANXA1 protein (when run in parallel with an anti-CD20 monoclonal antibody) is highly specific for HCL at first diagnosis.74  However, it is not suitable for monitoring residual disease after therapy because the detection of low numbers of ANXA1+ HCL cells is made difficult by the presence of the overwhelming population of myeloid elements, macrophages, and T cells (that also express ANXA1).13,75-77  Under these circumstances, double immunostaining for ANXA1(cytoplasmic)/Pax5 (nuclear) or BRAF V600E mutation studies (see next section) may overcome the problem.

Detection of BRAF V600E by molecular assays and immunohistochemistry

The BRAF V600E mutation can be detected in BM or peripheral blood (PB) samples by Sanger sequencing or other more sensitive polymerase chain reaction (PCR)-based techniques18,21,25,38  (Figure 3). These molecular assays are usually applied to fresh samples but they may also work in archival Romanowsky-stained air-dried PB and BM smears and even in fixed-decalcified/paraffin-embedded BM trephine biopsies.78  An alternative approach for detecting BRAF V600E in routine paraffin sections is based on immunostaining with the monoclonal antibody (VE1) specifically directed against the BRAF V600E mutant.30,79  This antibody has been claimed to represent a reliable surrogate for molecular PCR assays80  but further studies are required to validate these findings

Figure 3

Differential diagnosis between HCL and its mimickers. (A) A needle biopsy of a bulky abdominal mass between the liver and the spleen, featuring mature lymphoid cells that infiltrate pancreatic glandular structures (left panel; hematoxylin and eosin [H&E] staining); the immunohistochemical staining with Annexin-1 (ANXA1; red labeling in the right panel) confirms the HCL origin of this infiltrate in such an unusual anatomic site. (B) A BM biopsy involved in SMZL with its typical intrasinusoidal infiltration pattern, which is highlighted by the B-cell markers CD79a (red; left panel) and PAX5 (brown; right panel). SMZL cells do not express ANXA1 (blue, right panel, with ANXA1-positive myeloid and T cells serving as internal control). Pictures of immunohistochemical stainings shown in panels A and B were taken with the 40×/0.85 objective (U Plan Apo) of a BX61 microscope equipped with a DP71 digital camera, using cell^B x.y acquisition software (all from Olympus). (C) A genetic diagnosis of HCL (vs HCL-v) can be easily made in whole-blood samples through BRAF-mutant allele-specific DNA PCR followed by agarose gel electrophoresis. The BRAF V600E gel band is visible in the blood sample from an HCL patient (known to contain <1% of leukemic cells by flow cytometry) but not in the blood sample from an HCL-v patient (containing >40% leukemic cells), which shows only the BRAF wild-type amplicon. The gel lanes to the left of the HCL and HCL-v samples represent a serial dilution (from 100% to 0%) of BRAF-mutant alleles with wild-type ones, which sets the detection limit of this qualitative assay at 0.05% mutant alleles. Dashed lines separate lanes of the same gels that were repositioned for clarity.

Figure 3

Differential diagnosis between HCL and its mimickers. (A) A needle biopsy of a bulky abdominal mass between the liver and the spleen, featuring mature lymphoid cells that infiltrate pancreatic glandular structures (left panel; hematoxylin and eosin [H&E] staining); the immunohistochemical staining with Annexin-1 (ANXA1; red labeling in the right panel) confirms the HCL origin of this infiltrate in such an unusual anatomic site. (B) A BM biopsy involved in SMZL with its typical intrasinusoidal infiltration pattern, which is highlighted by the B-cell markers CD79a (red; left panel) and PAX5 (brown; right panel). SMZL cells do not express ANXA1 (blue, right panel, with ANXA1-positive myeloid and T cells serving as internal control). Pictures of immunohistochemical stainings shown in panels A and B were taken with the 40×/0.85 objective (U Plan Apo) of a BX61 microscope equipped with a DP71 digital camera, using cell^B x.y acquisition software (all from Olympus). (C) A genetic diagnosis of HCL (vs HCL-v) can be easily made in whole-blood samples through BRAF-mutant allele-specific DNA PCR followed by agarose gel electrophoresis. The BRAF V600E gel band is visible in the blood sample from an HCL patient (known to contain <1% of leukemic cells by flow cytometry) but not in the blood sample from an HCL-v patient (containing >40% leukemic cells), which shows only the BRAF wild-type amplicon. The gel lanes to the left of the HCL and HCL-v samples represent a serial dilution (from 100% to 0%) of BRAF-mutant alleles with wild-type ones, which sets the detection limit of this qualitative assay at 0.05% mutant alleles. Dashed lines separate lanes of the same gels that were repositioned for clarity.

Close modal

Differential diagnosis between HCL and HCL-like disorders

The immunohistochemical and molecular techniques may be particularly useful in confirming the diagnosis of HCL in unusual anatomical sites (Figure 3) and/or in distinguishing classic HCL from HCL-like disorders, such as splenic marginal zone lymphoma (SMZL), HCL variant (HCL-v),81  and splenic diffuse red pulp small B-cell lymphoma.81  This distinction is clinically important because classic HCL is treated differently from HCL-like disorders. The main features of HCL vs HCL-like disorders are briefly described in the following paragraphs and are summarized in Table 1.

Table 1

Differential diagnosis between HCL and HCL-like neoplasms (SMZL, HCL-v, SDRPBCL)

FeaturesHCLSMZLHCL-vSDRPBCL
Morphology     
 Nucleus* Oval, sometimes “coffee bean” shaped Round Round/oval Round/oval, sometimes eccentric 
 Nucleolus* Absent Absent or small Clearly evident Absent or small 
 Cytoplasm* Abundant, pale Moderately abundant, basophil, sometimes with plasmacytoid features Abundant Moderately abundant, basophil, sometimes with plasmacytoid features 
 Cell surface Thin, often long projections distributed all over the surface (circumferential) Projections with a polar (noncircumferential) distribution Similar to HCL Short and thick projections with polar distribution 
 BM infiltration pattern Diffuse and/or interstitial Nodular and/or intrasinusoidal Diffuse, intrasinusoidal, and/or interstitial Intrasinusoidal, interstitial, and/or nodular 
 Splenic infiltration pattern Red pulp, with effacement of white pulp White pulp Red pulp Diffuse (red and white pulp) 
Immunophenotype and genetic lesions     
 Immunophenotypic markers CD20+, CD5, CD10, CD23CD103+, CD25+, CD11c+, CD123+, AnnexinA1+, BRAF V600E+ CD20+, CD5, CD10, CD23CD103−/+, CD25−/+, CD11c+/, CD123, AnnexinA1, BRAF V600E CD20+, CD5, CD10, CD23CD103+, CD25, CD11c+, CD123, AnnexinA1, BRAF V600E CD20+, CD5−/+, CD10, CD23−/+,CD103−/+, CD25, CD11c+, CD123, AnnexinA1, BRAF V600E 
 Genetic lesions BRAF V600E mutation in >97% of cases; KLF2 and CDKN1B mutations each in 16% of cases Del(7q) in ∼30% of cases, NOTCH2 mutations in 10%-25% of cases, NF-κB pathway mutations in ∼35% of cases, KLF2 mutations in 20%-40% of cases MAP2K1 mutations in 50% of cases; 17p (TP53) deletion in ∼30% of cases Not well characterized 
FeaturesHCLSMZLHCL-vSDRPBCL
Morphology     
 Nucleus* Oval, sometimes “coffee bean” shaped Round Round/oval Round/oval, sometimes eccentric 
 Nucleolus* Absent Absent or small Clearly evident Absent or small 
 Cytoplasm* Abundant, pale Moderately abundant, basophil, sometimes with plasmacytoid features Abundant Moderately abundant, basophil, sometimes with plasmacytoid features 
 Cell surface Thin, often long projections distributed all over the surface (circumferential) Projections with a polar (noncircumferential) distribution Similar to HCL Short and thick projections with polar distribution 
 BM infiltration pattern Diffuse and/or interstitial Nodular and/or intrasinusoidal Diffuse, intrasinusoidal, and/or interstitial Intrasinusoidal, interstitial, and/or nodular 
 Splenic infiltration pattern Red pulp, with effacement of white pulp White pulp Red pulp Diffuse (red and white pulp) 
Immunophenotype and genetic lesions     
 Immunophenotypic markers CD20+, CD5, CD10, CD23CD103+, CD25+, CD11c+, CD123+, AnnexinA1+, BRAF V600E+ CD20+, CD5, CD10, CD23CD103−/+, CD25−/+, CD11c+/, CD123, AnnexinA1, BRAF V600E CD20+, CD5, CD10, CD23CD103+, CD25, CD11c+, CD123, AnnexinA1, BRAF V600E CD20+, CD5−/+, CD10, CD23−/+,CD103−/+, CD25, CD11c+, CD123, AnnexinA1, BRAF V600E 
 Genetic lesions BRAF V600E mutation in >97% of cases; KLF2 and CDKN1B mutations each in 16% of cases Del(7q) in ∼30% of cases, NOTCH2 mutations in 10%-25% of cases, NF-κB pathway mutations in ∼35% of cases, KLF2 mutations in 20%-40% of cases MAP2K1 mutations in 50% of cases; 17p (TP53) deletion in ∼30% of cases Not well characterized 
*

In smears stained according to May-Grünwald Giemsa.

Unlike HCL, SMZL usually shows an intrasinusoidal infiltration pattern in the BM (clearly highlighted by immunohistochemistry for CD20) (Figure 3). Moreover, the SMZL cells circulating in the PB exhibit shorter hairy projections that are usually “polarized” to 1 area of the cell surface (unlike the circumferential hairy projections of HCL). Finally, SMZL cells are negative for the typical markers of HCL (especially CD103, CD25, and Annexin A1) (Figure 3), do not harbor BRAF V600E,21,38  and frequently carry Notch2 mutations.82,83 

A more difficult task is the differential diagnosis between HCL and HCL-v. The most important criteria for the diagnosis of HCL-v are: (1) the higher frequency of leukocytosis (observed in only 10%-15% of HCL cases); (2) the absence of monocytopenia (a rather constant feature in HCL); (3) the morphological features of both HCL (hairy surface) and prolymphocytic leukemia (single prominent nucleolus); (4) the negativity for CD25, ANXA1, BRAF V600E, and CD123; and (5) the presence of MAP2K1 mutations84  in 50% of cases.

The splenic diffuse red pulp small B-cell lymphoma (SDRPBCL) shows several overlapping features with HCL-v because both tumors are negative for CD25, ANXA1, and BRAF V600E.81,85  The red pulp is infiltrated by tumor cells with plasmacytoid appearance. Genomic studies have not been performed in SDRPBCL.

Although a rare disease, during the past 50 years, HCL has served as a paradigm for the development of new forms of therapy. For >2 decades, the only available therapeutic intervention in HCL was splenectomy. The first significant improvement in the care of this disease occurred in the early 80s, with the introduction of interferon.86  This drug usually induced a partial response, sometimes durable, in 40% to 80% of patients.87,88  Currently, the gold-standard therapy for HCL is based on the purine analogs cladribine and pentostatin89,90  (introduced in the 90s), both of which induce durable complete responses (CRs) in 80% to 85% of patients.3,91  Combining cladribine with the anti-CD20 monoclonal antibody (rituximab) may produce even better results.92 

In spite of these great advances, disease relapses occur in ∼40% to 50% of patients and these events are associated with a progressively worse response to purine analogs.93,94  Moreover, over time, therapy with purine analogs can lead to cumulative myelotoxic effects and profound immune suppression. Anti-CD22 immunotoxin (moxetumomab pasudotox) showed high clinical activity in this setting of patients.95  More recently, the availability of BRAF and MEK inhibitors, originally developed for the treatment of BRAF-mutated metastatic melanoma,96,97  offer a new therapeutic opportunity for HCL patients with refractory or relapsed disease.

Preclinical studies with BRAF inhibitors

Preclinical studies in HCL have been difficult to perform due to the absence of true human HCL cell lines5,6  and animal models of the disease.7  Therefore, the activity of inhibitors of BRAF (vemurafenib and dabrafenib) and MEK (trametinib) has been investigated in vitro on primary leukemic cells purified from HCL patients,52  a rather difficult task because of the BM fibrosis and the low percentage of circulating tumor cells that are typical of HCL. Notably, BRAF or MEK inhibitors caused marked dephosphorylation of MEK/ERK in HCL cells, silencing of the RAS-RAF-MEK-ERK pathway transcriptional output, loss of the HCL-specific GEP signature, change of the morphology from “hairy” to “smooth,” and eventually apoptosis.52  Usually, apoptosis followed changes in hairy morphology (Figure 2) and was associated with the upregulation of some proapoptotic genes, such as BIM/BCL2L11 and CDKN1C/p57-KIP2.52  These findings are in keeping with the concept that HCL growth is predominantly sustained by inhibition of apoptosis52  and that the proliferative index of HCL (<5%) is 1 of the lowest among B-cell neoplasms.58  Interestingly, BRAF inhibitors induced these effects specifically in HCL but not in primary leukemic cells from patients with HCL-like disorders. Collectively, these results strongly supported and informed the clinical use of BRAF and MEK inhibitors in HCL.

Therapy of relapsed/refractory HCL with BRAF inhibitors

The clinical activity of BRAF inhibitors was initially described in anecdotal cases of refractory/relapsed HCL.98-105  Recently, we reported the results of 2 phase-2 clinical trials (1 carried out in Italy and the other in the United States) in 54 patients with refractory/relapsed HCL treated with the BRAF inhibitor vemurafenib.63  In both studies, the drug was administered at the dose of 960 mg twice daily (as previously used in metastatic melanoma) for a median of 16 weeks in the Italian study and 18 weeks in the US study.63  A total of 49 patients were available for response. The overall response rates were 96% (CR = 35%; Figure 4) and 100% (CR = 42%) in the Italian trial (n = 25 patients) and the US study (n = 24 patients), respectively. Response to the drug was rapid, usually occurring within a median of 2 to 3 months.63 

Figure 4

Example of a complete remission obtained with vemurafenib in relapsed/refractory HCL. The BM biopsies of a patient with multiple HCL relapses is shown (A) before and (B) after 8 weeks of treatment with vemurafenib. H&E (top panels) and CD20 (red; bottom panels) stainings show a massive leukemic infiltration which is largely cleared by vemurafenib (<10% residual HCL cells posttherapy). Pictures of immunohistochemical stainings were taken with the 40×/0.85 objective (U Plan Apo) of a BX61 microscope equipped with a DP71 digital camera, using cell^B x.y acquisition software (all from Olympus).

Figure 4

Example of a complete remission obtained with vemurafenib in relapsed/refractory HCL. The BM biopsies of a patient with multiple HCL relapses is shown (A) before and (B) after 8 weeks of treatment with vemurafenib. H&E (top panels) and CD20 (red; bottom panels) stainings show a massive leukemic infiltration which is largely cleared by vemurafenib (<10% residual HCL cells posttherapy). Pictures of immunohistochemical stainings were taken with the 40×/0.85 objective (U Plan Apo) of a BX61 microscope equipped with a DP71 digital camera, using cell^B x.y acquisition software (all from Olympus).

Close modal

After a median follow-up of 23 months, the median relapse-free survival in the Italian trial was 19 months in patients who had achieved a CR and 6 months in those who had obtained a partial response63  (Figure 5); the median treatment-free survival in these cases was 25 and 18 months, respectively. In the US study, the progression-free survival and the overall survival at 1 year were 73% and 91%, respectively.63 

Figure 5

Relapse-free and treatment-free survival after vemurafenib in relapsed/refractory HCL. Kaplan-Meier curves displaying the survival free (A) from relapse and (B) from a subsequent antileukemic treatment in the HCL-PG01 phase-2 Italian trial at a median follow-up of 23 months after the last vemurafenib dose. Relapse was defined as the reappearance of 1 or more cytopenias (neutrophils, <1.5 × 109/L; platelets, <100 × 109/L; or hemoglobin, <11 g/dL). Time to relapse (A) was longer (by log-rank test) in patients who achieved a complete remission vs a partial remission. This difference was less evident regarding time to next treatment (B), as various patients relapsing after a partial remission developed mild cytopenias (eg, ∼1 N × 109/L or ∼80 platelets × 109/L) which remained relatively stable for some time and did not immediately trigger a subsequent treatment. Reprinted from Tiacci et al63  with permission.

Figure 5

Relapse-free and treatment-free survival after vemurafenib in relapsed/refractory HCL. Kaplan-Meier curves displaying the survival free (A) from relapse and (B) from a subsequent antileukemic treatment in the HCL-PG01 phase-2 Italian trial at a median follow-up of 23 months after the last vemurafenib dose. Relapse was defined as the reappearance of 1 or more cytopenias (neutrophils, <1.5 × 109/L; platelets, <100 × 109/L; or hemoglobin, <11 g/dL). Time to relapse (A) was longer (by log-rank test) in patients who achieved a complete remission vs a partial remission. This difference was less evident regarding time to next treatment (B), as various patients relapsing after a partial remission developed mild cytopenias (eg, ∼1 N × 109/L or ∼80 platelets × 109/L) which remained relatively stable for some time and did not immediately trigger a subsequent treatment. Reprinted from Tiacci et al63  with permission.

Close modal

In both trials, the toxicity of vemurafenib was similar to that previously reported in patients with metastatic melanoma.106-108  Drug-related adverse events mostly affected the skin (especially rash, photosensitivity, palmar/plantar fibrosis, warts) and the joints (arthralgia, arthritis). Dose reduction was required in 50% to 58% of patients in the 2 trials.63  Patients with arthralgias may benefit from a low dose of steroids.

Secondary tumors were observed in a total of 7 patients. Among the 3 patients in the Italian trial, 2 developed a basal cell carcinoma and 1 a superficial melanoma; 3 and 1 patients from the US study developed squamous cell carcinoma and a basal cell carcinoma, respectively.63  All tumors were managed by simple excision. Similar to what was observed in patients with metastatic melanoma, these tumors are likely related to a paradoxic effect of the BRAF inhibitor toward the skin keratinocytes or melanocytes when they carry a preexisting RAS mutation (especially affecting H-RAS).109 

Notably, vemurafenib showed no significant myelotoxicity. This suggest its potential usefulness not only in refractory/relapsed HCL patients but also as a frontline drug in patients who have to discontinue purine analogs because of severe side effects or in patients who, at the time of initial diagnosis, present with a severe opportunistic infection that precludes the use of myelotoxic agents.110 

Mechanisms of resistance to BRAF inhibitors in HCL

Similar to what was observed in metastatic melanoma,111-113  the dramatic responses achieved with vemurafenib in refractory/relapsed HCL were frequently followed by relapse, likely due to development of mechanisms of resistance to the BRAF inhibitor. At least in melanoma, the resistance mechanisms so-far identified in vitro and also confirmed in patients include: (1) the activation of tyrosine kinase receptors (eg, MET114 ) via interaction with microenvironmental factors (eg, HGF114 ) leading to the reactivation of the RAS-RAF-MEK-ERK pathway through CRAF, and also to the activation of alternative signaling pathways, such as PI3K-AKT; (2) the emergence of N-RAS115  mutations that induce the reactivation of the RAS-RAF-MEK-ERK signaling pathway, mainly through CRAF116 ; (3) the emergence of activating mutations of MEK117 ; (4) the overexpression of COT (encoded by the MAP3K8 gene),118  a protein kinase that can activate ERK through a MEK-dependent (but BRAF-independent) mechanism; (5) the copy-number amplification of the BRAF mutant119 ; and (6) the alternative splicing of the BRAF mutant leading to the generation of a smaller molecule able to form dimers that can evade the action of BRAF inhibitors.120 

At present, there is only scarce knowledge of the mechanisms of resistance to BRAF inhibitors operating in HCL. Like in melanoma, acquired mutations of BRAF are not implicated in the mechanism of resistance. In at least 1 patient in the phase-2 HCL trial, resistance to vemurafenib was due to the emergence of K-RAS mutations.63  Interestingly, at immunohistochemistry in the BM biopsy, 6 of 13 patients in the Italian trial showed residual leukemic cells that still expressed phospho-ERK despite prolonged exposure (16-20 weeks) to vemurafenib. These findings strongly suggest that, in at least some HCL patients, the growth of leukemic cells is still closely dependent on the RAS-RAF-MEK-ERK signaling pathway that is likely reactivated through mechanisms that bypass the activity of the BRAF inhibitors. Functional studies and gene sequencing of leukemic cells before and after treatment with BRAF inhibitors are needed to further elucidate the mechanisms of resistance to these agents in HCL.

Due to the lack of true HCL cell lines5,6  and the paucity of primary HCL cells that can usually be collected from HCL patients, a faithful BRAF V600E–based mouse model of the disease could be extremely useful both for functional studies and for testing the activity of new drugs. Moreover, because BRAF inhibitors do not appear to fully eradicate the disease, further studies on the nature of preleukemic HSCs7  may have pathogenetic implications.

In anecdotal cases, lower doses of vemurafenib (eg, 240 mg twice daily) proved to be effective in HCL patients with refractory or relapsed disease.98-100,121,122  In the phase-2 HCL trials,63  vemurafenib was administered at the dose of 960 mg twice daily (as previously used in metastatic melanoma) and mostly reduced to 720 mg (in the Italian trial) or 480 mg (in the US trial), sometimes followed by reescalation. Prospective clinical studies are necessary to investigate the optimal dose and treatment duration of BRAF inhibitors in HCL.

The duration of the response to BRAF inhibitors in refractory/relapsed HCL patients appears to correlate with the depth of the response (being usually longer in patients achieving a CR).63  It has been reported that duration of response to BRAF inhibitors in naive HCL patients may be shorter than that achievable with purine analogs.122  However, these results are based on anecdotal cases treated with a low dose of vemurafenib.

Future therapeutic strategies should be directed to overcome the mechanism of resistance with the aim of improving the rate and duration of CR and possibly reducing the incidence of secondary skin tumors. Based on previous experience in melanoma patients, clinical trials using dabrafenib (another BRAF inhibitor) alone or BRAF plus MEK inhibitors for dual targeting of the RAS-RAF-MEK-ERK signaling pathway97  are also ongoing in refractory/relapsed HCL. Combining a BRAF inhibitor with an anti-CD20 monoclonal antibody represents another interesting therapeutic strategy. The rationale for this approach is to use immunotherapy to eradicate the residual BRAF inhibitor-resistant HCL cells.

The authors thank L. De Carolis, G. Schiavoni, V. Pettirossi, A. Santi, and P. Sportoletti for their contribution to their research program on hairy cell leukemia.

This work was supported by grants from the Associazione Italiana per la Ricerca sul Cancro (MCO-10007 [B.F.] and IG-14447) (E.T.), the European Research Council (FP7/2007-2013 “Hairy Cell Leukemia,” 617471) (E.T.), the Hairy Cell Leukemia Foundation (B.F. and E.T.), Ministero dell’Istruzione, Università e Ricerca (PRIN 20104HBZ8E [B.F.] and Futuro in Ricerca 2010-RBFR10WT2K [E.T.]), Ministero della Salute (Giovani Ricercatori GR-2010-2303444) (E.T.), the Associazione Umbra contro le Leucemie e i Linfomi (B.F.), and Roche Pharmaceuticals (B.F.). E.T. is a Scholar in Clinical Research of the Leukemia & Lymphoma Society (contract no. 2030-14).

Contribution: B.F. wrote the text and prepared the table; E.T. prepared the figures and helped in writing the text and preparing the table; and M.P.M. commented on, edited, and approved the paper.

Conflict-of-interest disclosure: B.F. and E.T. filed a patent on the use of the BRAF V600E mutation as an HCL biomarker. B.F. received research funding from Roche. M.P.M. declares no competing financial interests.

Correspondence: Brunangelo Falini, Institute of Hematology and Center for Hemato-Oncology Research (C.R.E.O.), University and Hospital of Perugia, Perugia, Italy; e-mail: brunangelo.falini@unipg.it.

1
Foucar
 
K
Falini
 
B
Catovsky
 
D
Stein
 
H
 
Hairy cell leukaemia. In: Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon, France: International Agency for Research on Cancer (IARC); 2008:188-190
2
Schrek
 
R
Donnelly
 
WJ
“Hairy” cells in blood in lymphoreticular neoplastic disease and “flagellated” cells of normal lymph nodes.
Blood
1966
, vol. 
27
 
2
(pg. 
199
-
211
)
3
Grever
 
MR
How I treat hairy cell leukemia.
Blood
2010
, vol. 
115
 
1
(pg. 
21
-
28
)
4
Bouroncle
 
BA
Wiseman
 
BK
Doan
 
CA
Leukemic reticuloendotheliosis.
Blood
1958
, vol. 
13
 
7
(pg. 
609
-
630
)
5
Tiacci
 
E
Pucciarini
 
A
Bigerna
 
B
, et al. 
Absence of BRAF-V600E in the human cell lines BONNA-12, ESKOL, HAIR-M, and HC-1 questions their origin from hairy cell leukemia.
Blood
2012
, vol. 
119
 
22
(pg. 
5332
-
5333
)
6
Weston-Bell
 
NJ
Hendriks
 
D
Sugiyarto
 
G
, et al. 
Hairy cell leukemia cell lines expressing annexin A1 and displaying B-cell receptor signals characteristic of primary tumor cells lack the signature BRAF mutation to reveal unrepresentative origins.
Leukemia
2013
, vol. 
27
 
1
(pg. 
241
-
245
)
7
Chung
 
SS
Kim
 
E
Park
 
JH
, et al. 
Hematopoietic stem cell origin of BRAFV600E mutations in hairy cell leukemia.
Sci Transl Med
2014
, vol. 
6
 
238
pg. 
238ra71
 
8
Sambani
 
C
Trafalis
 
DT
Mitsoulis-Mentzikoff
 
C
, et al. 
Clonal chromosome rearrangements in hairy cell leukemia: personal experience and review of literature.
Cancer Genet Cytogenet
2001
, vol. 
129
 
2
(pg. 
138
-
144
)
9
Andersen
 
CL
Gruszka-Westwood
 
A
Østergaard
 
M
, et al. 
A narrow deletion of 7q is common to HCL, and SMZL, but not CLL.
Eur J Haematol
2004
, vol. 
72
 
6
(pg. 
390
-
402
)
10
Nordgren
 
A
Corcoran
 
M
Sääf
 
A
, et al. 
Characterisation of hairy cell leukaemia by tiling resolution array-based comparative genome hybridisation: a series of 13 cases and review of the literature.
Eur J Haematol
2010
, vol. 
84
 
1
(pg. 
17
-
25
)
11
Hockley
 
SL
Morgan
 
GJ
Leone
 
PE
, et al. 
High-resolution genomic profiling in hairy cell leukemia-variant compared with typical hairy cell leukemia.
Leukemia
2011
, vol. 
25
 
7
(pg. 
1189
-
1192
)
12
Rinaldi
 
A
Kwee
 
I
Young
 
KH
, et al. 
Genome-wide high resolution DNA profiling of hairy cell leukaemia.
Br J Haematol
2013
, vol. 
162
 
4
(pg. 
566
-
569
)
13
Basso
 
K
Liso
 
A
Tiacci
 
E
, et al. 
Gene expression profiling of hairy cell leukemia reveals a phenotype related to memory B cells with altered expression of chemokine and adhesion receptors.
J Exp Med
2004
, vol. 
199
 
1
(pg. 
59
-
68
)
14
Kitagawa
 
Y
Brahmachary
 
M
Tiacci
 
E
Dalla-Favera
 
R
Falini
 
B
Basso
 
K
A microRNA signature specific for hairy cell leukemia and associated with modulation of the MAPK-JNK pathways.
Leukemia
2012
, vol. 
26
 
12
(pg. 
2564
-
2567
)
15
Tiacci
 
E
Liso
 
A
Piris
 
M
Falini
 
B
Evolving concepts in the pathogenesis of hairy-cell leukaemia.
Nat Rev Cancer
2006
, vol. 
6
 
6
(pg. 
437
-
448
)
16
Forconi
 
F
Poretti
 
G
Kwee
 
I
, et al. 
High density genome-wide DNA profiling reveals a remarkably stable profile in hairy cell leukaemia.
Br J Haematol
2008
, vol. 
141
 
5
(pg. 
622
-
630
)
17
Tiacci
 
E
Trifonov
 
V
Schiavoni
 
G
, et al. 
BRAF mutations in hairy-cell leukemia.
N Engl J Med
2011
, vol. 
364
 
24
(pg. 
2305
-
2315
)
18
Boyd
 
EM
Bench
 
AJ
van ’t Veer
 
MB
, et al. 
High resolution melting analysis for detection of BRAF exon 15 mutations in hairy cell leukaemia and other lymphoid malignancies.
Br J Haematol
2011
, vol. 
155
 
5
(pg. 
609
-
612
)
19
Zhang
 
X
Reis
 
M
Khoriaty
 
R
, et al. 
Sequence analysis of 515 kinase genes in chronic lymphocytic leukemia.
Leukemia
2011
, vol. 
25
 
12
(pg. 
1908
-
1910
)
20
Chapman
 
MA
Lawrence
 
MS
Keats
 
JJ
, et al. 
Initial genome sequencing and analysis of multiple myeloma.
Nature
2011
, vol. 
471
 
7339
(pg. 
467
-
472
)
21
Quesada
 
V
Conde
 
L
Villamor
 
N
, et al. 
Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia.
Nat Genet
2011
, vol. 
44
 
1
(pg. 
47
-
52
)
22
Arcaini
 
L
Zibellini
 
S
Boveri
 
E
, et al. 
The BRAF V600E mutation in hairy cell leukemia and other mature B-cell neoplasms.
Blood
2012
, vol. 
119
 
1
(pg. 
188
-
191
)
23
Blombery
 
PA
Wong
 
SQ
Hewitt
 
CA
, et al. 
Detection of BRAF mutations in patients with hairy cell leukemia and related lymphoproliferative disorders.
Haematologica
2012
, vol. 
97
 
5
(pg. 
780
-
783
)
24
Lennerz
 
JK
Klaus
 
BM
Marienfeld
 
RB
Möller
 
P
Pyrosequencing of BRAF V600E in routine samples of hairy cell leukaemia identifies CD5+ variant hairy cell leukaemia that lacks V600E.
Br J Haematol
2012
, vol. 
157
 
2
(pg. 
267
-
269
)
25
Schnittger
 
S
Bacher
 
U
Haferlach
 
T
, et al. 
Development and validation of a real-time quantification assay to detect and monitor BRAFV600E mutations in hairy cell leukemia.
Blood
2012
, vol. 
119
 
13
(pg. 
3151
-
3154
)
26
Xi
 
L
Arons
 
E
Navarro
 
W
, et al. 
Both variant and IGHV4-34-expressing hairy cell leukemia lack the BRAF V600E mutation.
Blood
2012
, vol. 
119
 
14
(pg. 
3330
-
3332
)
27
Langabeer
 
SE
O’Brien
 
D
Liptrot
 
S
, et al. 
Correlation of the BRAF V600E mutation in hairy cell leukaemia with morphology, cytochemistry and immunophenotype.
Int J Lab Hematol
2012
, vol. 
34
 
4
(pg. 
417
-
421
)
28
Ewalt
 
M
Nandula
 
S
Phillips
 
A
, et al. 
Real-time PCR-based analysis of BRAF V600E mutation in low and intermediate grade lymphomas confirms frequent occurrence in hairy cell leukaemia.
Hematol Oncol
2012
, vol. 
30
 
4
(pg. 
190
-
193
)
29
Langabeer
 
SE
Quinn
 
F
O’Brien
 
D
, et al. 
Incidence of the BRAF V600E mutation in chronic lymphocytic leukaemia and prolymphocytic leukaemia.
Leuk Res
2012
, vol. 
36
 
4
(pg. 
483
-
484
)
30
Andrulis
 
M
Penzel
 
R
Weichert
 
W
von Deimling
 
A
Capper
 
D
Application of a BRAF V600E mutation-specific antibody for the diagnosis of hairy cell leukemia.
Am J Surg Pathol
2012
, vol. 
36
 
12
(pg. 
1796
-
1800
)
31
Jebaraj
 
BM
Kienle
 
D
Bühler
 
A
, et al. 
BRAF mutations in chronic lymphocytic leukemia.
Leuk Lymphoma
2013
, vol. 
54
 
6
(pg. 
1177
-
1182
)
32
Raess
 
PW
Mintzer
 
D
Husson
 
M
, et al. 
BRAF V600E is also seen in unclassifiable splenic B-cell lymphoma/leukemia, a potential mimic of hairy cell leukemia.
Blood
2013
, vol. 
122
 
17
(pg. 
3084
-
3085
)
33
Akarca
 
AU
Shende
 
VH
Ramsay
 
AD
, et al. 
BRAF V600E mutation-specific antibody, a sensitive diagnostic marker revealing minimal residual disease in hairy cell leukaemia.
Br J Haematol
2013
, vol. 
162
 
6
(pg. 
848
-
851
)
34
Rider
 
T
Powell
 
R
Gover
 
R
, et al. 
Molecular detection of BRAF-V600E is superior to flow cytometry for disease evaluation in hairy cell leukaemia.
Hematol Oncol
2014
, vol. 
32
 
3
(pg. 
158
-
161
)
35
Tschernitz
 
S
Flossbach
 
L
Bonengel
 
M
Roth
 
S
Rosenwald
 
A
Geissinger
 
E
Alternative BRAF mutations in BRAF V600E-negative hairy cell leukaemias.
Br J Haematol
2014
, vol. 
165
 
4
(pg. 
529
-
533
)
36
Brown
 
NA
Weigelin
 
HC
Bailey
 
N
, et al. 
Requisite analytic and diagnostic performance characteristics for the clinical detection of BRAF V600E in hairy cell leukemia: a comparison of 2 allele-specific PCR assays.
Appl Immunohistochem Mol Morphol
2015
, vol. 
23
 
8
(pg. 
590
-
600
)
37
Okada
 
K
Kunitomi
 
A
Sakai
 
K
, et al. 
Hairy cell leukemia with systemic lymphadenopathy: detection of BRAF mutations in both lymph node and peripheral blood specimens.
Intern Med
2015
, vol. 
54
 
11
(pg. 
1397
-
1402
)
38
Tiacci
 
E
Schiavoni
 
G
Forconi
 
F
, et al. 
Simple genetic diagnosis of hairy cell leukemia by sensitive detection of the BRAF-V600E mutation.
Blood
2012
, vol. 
119
 
1
(pg. 
192
-
195
)
39
Marais
 
R
Light
 
Y
Paterson
 
HF
Mason
 
CS
Marshall
 
CJ
Differential regulation of Raf-1, A-Raf, and B-Raf by oncogenic ras and tyrosine kinases.
J Biol Chem
1997
, vol. 
272
 
7
(pg. 
4378
-
4383
)
40
Chong
 
H
Lee
 
J
Guan
 
KL
Positive and negative regulation of Raf kinase activity and function by phosphorylation.
EMBO J
2001
, vol. 
20
 
14
(pg. 
3716
-
3727
)
41
Emuss
 
V
Garnett
 
M
Mason
 
C
Marais
 
R
Mutations of C-RAF are rare in human cancer because C-RAF has a low basal kinase activity compared with B-RAF.
Cancer Res
2005
, vol. 
65
 
21
(pg. 
9719
-
9726
)
42
Davies
 
H
Bignell
 
GR
Cox
 
C
, et al. 
Mutations of the BRAF gene in human cancer.
Nature
2002
, vol. 
417
 
6892
(pg. 
949
-
954
)
43
Ravandi
 
F
Talpaz
 
M
Estrov
 
Z
Modulation of cellular signaling pathways: prospects for targeted therapy in hematological malignancies.
Clin Cancer Res
2003
, vol. 
9
 
2
(pg. 
535
-
550
)
44
Vakiani
 
E
Solit
 
DB
KRAS and BRAF: drug targets and predictive biomarkers.
J Pathol
2011
, vol. 
223
 
2
(pg. 
219
-
229
)
45
Weber
 
CK
Slupsky
 
JR
Kalmes
 
HA
Rapp
 
UR
Active Ras induces heterodimerization of cRaf and BRaf.
Cancer Res
2001
, vol. 
61
 
9
(pg. 
3595
-
3598
)
46
Rushworth
 
LK
Hindley
 
AD
O’Neill
 
E
Kolch
 
W
Regulation and role of Raf-1/B-Raf heterodimerization.
Mol Cell Biol
2006
, vol. 
26
 
6
(pg. 
2262
-
2272
)
47
Zhang
 
W
Liu
 
HT
MAPK signal pathways in the regulation of cell proliferation in mammalian cells.
Cell Res
2002
, vol. 
12
 
1
(pg. 
9
-
18
)
48
Flockhart
 
RJ
Armstrong
 
JL
Reynolds
 
NJ
Lovat
 
PE
NFAT signalling is a novel target of oncogenic BRAF in metastatic melanoma.
Br J Cancer
2009
, vol. 
101
 
8
(pg. 
1448
-
1455
)
49
Wan
 
PT
Garnett
 
MJ
Roe
 
SM
, et al. 
Cancer Genome Project
Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF.
Cell
2004
, vol. 
116
 
6
(pg. 
855
-
867
)
50
Tiacci
 
E
Schiavoni
 
G
Martelli
 
MP
, et al. 
Constant activation of the RAF-MEK-ERK pathway as a diagnostic and therapeutic target in hairy cell leukemia.
Haematologica
2013
, vol. 
98
 
4
(pg. 
635
-
639
)
51
Warden
 
DW
Ondrejka
 
S
Lin
 
J
Durkin
 
L
Bodo
 
J
Hsi
 
ED
Phospho-ERK(THR202/Tyr214) is overexpressed in hairy cell leukemia and is a useful diagnostic marker in bone marrow trephine sections.
Am J Surg Pathol
2013
, vol. 
37
 
2
(pg. 
305
-
308
)
52
Pettirossi
 
V
Santi
 
A
Imperi
 
E
, et al. 
BRAF inhibitors reverse the unique molecular signature and phenotype of hairy cell leukemia and exert potent antileukemic activity.
Blood
2015
, vol. 
125
 
8
(pg. 
1207
-
1216
)
53
Kamiguti
 
AS
Harris
 
RJ
Slupsky
 
JR
Baker
 
PK
Cawley
 
JC
Zuzel
 
M
Regulation of hairy-cell survival through constitutive activation of mitogen-activated protein kinase pathways.
Oncogene
2003
, vol. 
22
 
15
(pg. 
2272
-
2284
)
54
Xia
 
Z
Dickens
 
M
Raingeaud
 
J
Davis
 
RJ
Greenberg
 
ME
Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis.
Science
1995
, vol. 
270
 
5240
(pg. 
1326
-
1331
)
55
Roovers
 
K
Davey
 
G
Zhu
 
X
Bottazzi
 
ME
Assoian
 
RK
Alpha5beta1 integrin controls cyclin D1 expression by sustaining mitogen-activated protein kinase activity in growth factor-treated cells.
Mol Biol Cell
1999
, vol. 
10
 
10
(pg. 
3197
-
3204
)
56
Miranda
 
RN
Briggs
 
RC
Kinney
 
MC
Veno
 
PA
Hammer
 
RD
Cousar
 
JB
Immunohistochemical detection of cyclin D1 using optimized conditions is highly specific for mantle cell lymphoma and hairy cell leukemia.
Mod Pathol
2000
, vol. 
13
 
12
(pg. 
1308
-
1314
)
57
Sherman
 
MJ
Hanson
 
CA
Hoyer
 
JD
An assessment of the usefulness of immunohistochemical stains in the diagnosis of hairy cell leukemia.
Am J Clin Pathol
2011
, vol. 
136
 
3
(pg. 
390
-
399
)
58
Chilosi
 
M
Chiarle
 
R
Lestani
 
M
, et al. 
Low expression of p27 and low proliferation index do not correlate in hairy cell leukaemia.
Br J Haematol
2000
, vol. 
111
 
1
(pg. 
263
-
271
)
59
Bhatt
 
KV
Spofford
 
LS
Aram
 
G
McMullen
 
M
Pumiglia
 
K
Aplin
 
AE
Adhesion control of cyclin D1 and p27Kip1 levels is deregulated in melanoma cells through BRAF-MEK-ERK signaling.
Oncogene
2005
, vol. 
24
 
21
(pg. 
3459
-
3471
)
60
Bhatt
 
KV
Hu
 
R
Spofford
 
LS
Aplin
 
AE
Mutant B-RAF signaling and cyclin D1 regulate Cks1/S-phase kinase-associated protein 2-mediated degradation of p27Kip1 in human melanoma cells.
Oncogene
2007
, vol. 
26
 
7
(pg. 
1056
-
1066
)
61
Piva
 
R
Deaglio
 
S
Famà
 
R
, et al. 
The Krüppel-like factor 2 transcription factor gene is recurrently mutated in splenic marginal zone lymphoma.
Leukemia
2015
, vol. 
29
 
2
(pg. 
503
-
507
)
62
Dietrich
 
S
Hüllein
 
J
Lee
 
SC
, et al. 
Recurrent CDKN1B (p27) mutations in hairy cell leukemia.
Blood
2015
, vol. 
126
 
8
(pg. 
1005
-
1008
)
63
Tiacci
 
E
Park
 
JH
De Carolis
 
L
, et al. 
Targeting mutant BRAF in relapsed or refractory hairy-cell leukemia.
N Engl J Med
2015
, vol. 
373
 
18
(pg. 
1733
-
1747
)
64
Caligaris-Cappio
 
F
Bergui
 
L
Tesio
 
L
Corbascio
 
G
Tousco
 
F
Marchisio
 
PC
Cytoskeleton organization is aberrantly rearranged in the cells of B chronic lymphocytic leukemia and hairy cell leukemia.
Blood
1986
, vol. 
67
 
1
(pg. 
233
-
239
)
65
Raghunathan
 
A
Sivakamasundari
 
R
Wolenski
 
J
Poddar
 
R
Weissman
 
SM
Functional analysis of B144/LST1: a gene in the tumor necrosis factor cluster that induces formation of long filopodia in eukaryotic cells.
Exp Cell Res
2001
, vol. 
268
 
2
(pg. 
230
-
244
)
66
Schiller
 
C
Diakopoulos
 
KN
Rohwedder
 
I
, et al. 
LST1 promotes the assembly of a molecular machinery responsible for tunneling nanotube formation.
J Cell Sci
2013
, vol. 
126
 
Pt 3
(pg. 
767
-
777
)
67
Ju
 
YT
Chang
 
AC
She
 
BR
, et al. 
gas7: a gene expressed preferentially in growth-arrested fibroblasts and terminally differentiated Purkinje neurons affects neurite formation.
Proc Natl Acad Sci USA
1998
, vol. 
95
 
19
(pg. 
11423
-
11428
)
68
Schmidt
 
C
Künemund
 
V
Wintergerst
 
ES
Schmitz
 
B
Schachner
 
M
CD9 of mouse brain is implicated in neurite outgrowth and cell migration in vitro and is associated with the alpha 6/beta 1 integrin and the neural adhesion molecule L1.
J Neurosci Res
1996
, vol. 
43
 
1
(pg. 
12
-
31
)
69
Jongstra-Bilen
 
J
Janmey
 
PA
Hartwig
 
JH
Galea
 
S
Jongstra
 
J
The lymphocyte-specific protein LSP1 binds to F-actin and to the cytoskeleton through its COOH-terminal basic domain.
J Cell Biol
1992
, vol. 
118
 
6
(pg. 
1443
-
1453
)
70
Miyoshi
 
EK
Stewart
 
PL
Kincade
 
PW
Lee
 
MB
Thompson
 
AA
Wall
 
R
Aberrant expression and localization of the cytoskeleton-binding pp52 (LSP1) protein in hairy cell leukemia.
Leuk Res
2001
, vol. 
25
 
1
(pg. 
57
-
67
)
71
Zhang
 
X
Machii
 
T
Matsumura
 
I
, et al. 
Constitutively activated Rho guanosine triphosphatases regulate the growth and morphology of hairy cell leukemia cells.
Int J Hematol
2003
, vol. 
77
 
3
(pg. 
263
-
273
)
72
Chaigne-Delalande
 
B
Deuve
 
L
Reuzeau
 
E
, et al. 
RhoGTPases and p53 are involved in the morphological appearance and interferon-alpha response of hairy cells.
Am J Pathol
2006
, vol. 
168
 
2
(pg. 
562
-
573
)
73
Morgan
 
EA
Yu
 
H
Pinkus
 
JL
Pinkus
 
GS
Immunohistochemical detection of hairy cell leukemia in paraffin sections using a highly effective CD103 rabbit monoclonal antibody.
Am J Clin Pathol
2013
, vol. 
139
 
2
(pg. 
220
-
230
)
74
Falini
 
B
Tiacci
 
E
Liso
 
A
, et al. 
Simple diagnostic assay for hairy cell leukaemia by immunocytochemical detection of annexin A1 (ANXA1).
Lancet
2004
, vol. 
363
 
9424
(pg. 
1869
-
1870
)
75
Sadik
 
W
Coupland
 
S
Thachil
 
J
Hairy cell leukaemia, negative for conventional cell markers, diagnosed using antibodies to annexin A1 and T-bet.
Br J Haematol
2010
, vol. 
151
 
3
pg. 
207
 
76
Wotherspoon
 
A
Attygalle
 
A
Mendes
 
LS
Bone marrow and splenic histology in hairy cell leukaemia.
Best Pract Res Clin Haematol
2015
, vol. 
28
 
4
(pg. 
200
-
207
)
77
Tóth-Lipták
 
J
Piukovics
 
K
Borbényi
 
Z
Demeter
 
J
Bagdi
 
E
Krenács
 
L
A comprehensive immunophenotypic marker analysis of hairy cell leukemia in paraffin-embedded bone marrow trephine biopsies--a tissue microarray study.
Pathol Oncol Res
2015
, vol. 
21
 
1
(pg. 
203
-
211
)
78
Thomas
 
C
Amanuel
 
B
Finlayson
 
J
Grieu-Iacopetta
 
F
Spagnolo
 
DV
Erber
 
WN
BRAF mutation detection in hairy cell leukaemia from archival haematolymphoid specimens.
Pathology
2015
, vol. 
47
 
4
(pg. 
349
-
354
)
79
Uppal
 
G
Ly
 
V
Wang
 
ZX
, et al. 
The utility of BRAF V600E mutation-specific antibody VE1 for the diagnosis of hairy cell leukemia.
Am J Clin Pathol
2015
, vol. 
143
 
1
(pg. 
120
-
125
)
80
Brown
 
NA
Betz
 
BL
Weigelin
 
HC
Elenitoba-Johnson
 
KS
Lim
 
MS
Bailey
 
NG
Evaluation of allele-specific PCR and immunohistochemistry for the detection of BRAF V600E mutations in hairy cell leukemia.
Am J Clin Pathol
2015
, vol. 
143
 
1
(pg. 
89
-
99
)
81
Piris
 
M
Foucar
 
K
Mollejo
 
M
Campo
 
E
Falini
 
B
 
Splenic B-cell lymphoma/leukaemia, unclassificable. In: Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon, France: International Agency for Research on Cancer (IARC); 2008:191-193
82
Rossi
 
D
Trifonov
 
V
Fangazio
 
M
, et al. 
The coding genome of splenic marginal zone lymphoma: activation of NOTCH2 and other pathways regulating marginal zone development.
J Exp Med
2012
, vol. 
209
 
9
(pg. 
1537
-
1551
)
83
Kiel
 
MJ
Velusamy
 
T
Betz
 
BL
, et al. 
Whole-genome sequencing identifies recurrent somatic NOTCH2 mutations in splenic marginal zone lymphoma.
J Exp Med
2012
, vol. 
209
 
9
(pg. 
1553
-
1565
)
84
Waterfall
 
JJ
Arons
 
E
Walker
 
RL
, et al. 
High prevalence of MAP2K1 mutations in variant and IGHV4-34-expressing hairy-cell leukemias.
Nat Genet
2014
, vol. 
46
 
1
(pg. 
8
-
10
)
85
Traverse-Glehen
 
A
Baseggio
 
L
Bauchu
 
EC
, et al. 
Splenic red pulp lymphoma with numerous basophilic villous lymphocytes: a distinct clinicopathologic and molecular entity?
Blood
2008
, vol. 
111
 
4
(pg. 
2253
-
2260
)
86
Quesada
 
JR
Reuben
 
J
Manning
 
JT
Hersh
 
EM
Gutterman
 
JU
Alpha interferon for induction of remission in hairy-cell leukemia.
N Engl J Med
1984
, vol. 
310
 
1
(pg. 
15
-
18
)
87
Federico
 
M
Frassoldati
 
A
Lamparelli
 
T
, et al. 
Long-term results of alpha interferon as initial therapy and splenectomy as consolidation therapy in patients with hairy cell leukemia. Final report from the Italian Cooperative Group for HCL.
Ann Oncol
1994
, vol. 
5
 
8
(pg. 
725
-
731
)
88
Grever
 
M
Kopecky
 
K
Foucar
 
MK
, et al. 
Randomized comparison of pentostatin versus interferon alfa-2a in previously untreated patients with hairy cell leukemia: an intergroup study.
J Clin Oncol
1995
, vol. 
13
 
4
(pg. 
974
-
982
)
89
Spiers
 
AS
Moore
 
D
Cassileth
 
PA
, et al. 
Remissions in hairy-cell leukemia with pentostatin (2′-deoxycoformycin).
N Engl J Med
1987
, vol. 
316
 
14
(pg. 
825
-
830
)
90
Piro
 
LD
Carrera
 
CJ
Carson
 
DA
Beutler
 
E
Lasting remissions in hairy-cell leukemia induced by a single infusion of 2-chlorodeoxyadenosine.
N Engl J Med
1990
, vol. 
322
 
16
(pg. 
1117
-
1121
)
91
Maevis
 
V
Mey
 
U
Schmidt-Wolf
 
G
Schmidt-Wolf
 
IG
Hairy cell leukemia: short review, today’s recommendations and outlook.
Blood Cancer J
2014
, vol. 
4
 pg. 
e184
 
92
Ravandi
 
F
O’Brien
 
S
Jorgensen
 
J
, et al. 
Phase 2 study of cladribine followed by rituximab in patients with hairy cell leukemia.
Blood
2011
, vol. 
118
 
14
(pg. 
3818
-
3823
)
93
Else
 
M
Dearden
 
CE
Matutes
 
E
, et al. 
Long-term follow-up of 233 patients with hairy cell leukaemia, treated initially with pentostatin or cladribine, at a median of 16 years from diagnosis.
Br J Haematol
2009
, vol. 
145
 
6
(pg. 
733
-
740
)
94
Rosenberg
 
JD
Burian
 
C
Waalen
 
J
Saven
 
A
Clinical characteristics and long-term outcome of young hairy cell leukemia patients treated with cladribine: a single-institution series.
Blood
2014
, vol. 
123
 
2
(pg. 
177
-
183
)
95
Kreitman
 
RJ
Pastan
 
I
Immunoconjugates in the management of hairy cell leukemia.
Best Pract Res Clin Haematol
2015
, vol. 
28
 
4
(pg. 
236
-
245
)
96
Shelledy
 
L
Roman
 
D
Vemurafenib: first-in-class BRAF-mutated inhibitor for the treatment of unresectable or metastatic melanoma.
J Adv Pract Oncol
2015
, vol. 
6
 
4
(pg. 
361
-
365
)
97
Eroglu
 
Z
Ribas
 
A
Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and place in therapy.
Ther Adv Med Oncol
2016
, vol. 
8
 
1
(pg. 
48
-
56
)
98
Dietrich
 
S
Glimm
 
H
Andrulis
 
M
von Kalle
 
C
Ho
 
AD
Zenz
 
T
BRAF inhibition in refractory hairy-cell leukemia.
N Engl J Med
2012
, vol. 
366
 
21
(pg. 
2038
-
2040
)
99
Follows
 
GA
Sims
 
H
Bloxham
 
DM
, et al. 
Rapid response of biallelic BRAF V600E mutated hairy cell leukaemia to low dose vemurafenib.
Br J Haematol
2013
, vol. 
161
 
1
(pg. 
150
-
153
)
100
Peyrade
 
F
Re
 
D
Ginet
 
C
, et al. 
Low-dose vemurafenib induces complete remission in a case of hairy-cell leukemia with a V600E mutation.
Haematologica
2013
, vol. 
98
 
2
(pg. 
e20
-
e22
)
101
Munoz
 
J
Schlette
 
E
Kurzrock
 
R
Rapid response to vemurafenib in a heavily pretreated patient with hairy cell leukemia and a BRAF mutation.
J Clin Oncol
2013
, vol. 
31
 
20
(pg. 
e351
-
e352
)
102
Samuel
 
J
Macip
 
S
Dyer
 
MJ
Efficacy of vemurafenib in hairy-cell leukemia.
N Engl J Med
2014
, vol. 
370
 
3
(pg. 
286
-
288
)
103
Bailleux
 
C
Robert
 
G
Ginet
 
C
, et al. 
Successful re-treatment of a relapsed V600E mutated HCL patient with low-dose vemurafenib.
Oncoscience
2014
, vol. 
2
 
1
(pg. 
44
-
49
)
104
Vergote
 
V
Dierickx
 
D
Janssens
 
A
, et al. 
Rapid and complete hematological response of refractory hairy cell leukemia to the BRAF inhibitor dabrafenib.
Ann Hematol
2014
, vol. 
93
 
12
(pg. 
2087
-
2089
)
105
Blachly
 
JS
Lozanski
 
G
Lucas
 
DM
Grever
 
MR
Kendra
 
K
Andritsos
 
LA
Cotreatment of hairy cell leukemia and melanoma with the BRAF inhibitor dabrafenib.
J Natl Compr Canc Netw
2015
, vol. 
13
 
1
(pg. 
9
-
13
)
106
Flaherty
 
KT
Puzanov
 
I
Kim
 
KB
, et al. 
Inhibition of mutated, activated BRAF in metastatic melanoma.
N Engl J Med
2010
, vol. 
363
 
9
(pg. 
809
-
819
)
107
Chapman
 
PB
Hauschild
 
A
Robert
 
C
, et al. 
BRIM-3 Study Group
Improved survival with vemurafenib in melanoma with BRAF V600E mutation.
N Engl J Med
2011
, vol. 
364
 
26
(pg. 
2507
-
2516
)
108
Sosman
 
JA
Kim
 
KB
Schuchter
 
L
, et al. 
Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib.
N Engl J Med
2012
, vol. 
366
 
8
(pg. 
707
-
714
)
109
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
, vol. 
366
 
3
(pg. 
207
-
215
)
110
Maurer
 
H
Haas
 
P
Wengenmayer
 
T
Lübbert
 
M
Duyster
 
J
Zeiser
 
R
Successful vemurafenib salvage treatment in a patient with primary refractory hairy cell leukemia and pulmonary aspergillosis.
Ann Hematol
2014
, vol. 
93
 
8
(pg. 
1439
-
1440
)
111
Wagle
 
N
Emery
 
C
Berger
 
MF
, et al. 
Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling.
J Clin Oncol
2011
, vol. 
29
 
22
(pg. 
3085
-
3096
)
112
Shi
 
H
Hugo
 
W
Kong
 
X
, et al. 
Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy.
Cancer Discov
2014
, vol. 
4
 
1
(pg. 
80
-
93
)
113
Rizos
 
H
Menzies
 
AM
Pupo
 
GM
, et al. 
BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact.
Clin Cancer Res
2014
, vol. 
20
 
7
(pg. 
1965
-
1977
)
114
Straussman
 
R
Morikawa
 
T
Shee
 
K
, et al. 
Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion.
Nature
2012
, vol. 
487
 
7408
(pg. 
500
-
504
)
115
Nazarian
 
R
Shi
 
H
Wang
 
Q
, et al. 
Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation.
Nature
2010
, vol. 
468
 
7326
(pg. 
973
-
977
)
116
Trunzer
 
K
Pavlick
 
AC
Schuchter
 
L
, et al. 
Pharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanoma.
J Clin Oncol
2013
, vol. 
31
 
14
(pg. 
1767
-
1774
)
117
Marusiak
 
AA
Edwards
 
ZC
Hugo
 
W
, et al. 
Mixed lineage kinases activate MEK independently of RAF to mediate resistance to RAF inhibitors.
Nat Commun
2014
, vol. 
5
 pg. 
3901
 
118
Johannessen
 
CM
Boehm
 
JS
Kim
 
SY
, et al. 
COT drives resistance to RAF inhibition through MAP kinase pathway reactivation.
Nature
2010
, vol. 
468
 
7326
(pg. 
968
-
972
)
119
Shi
 
H
Moriceau
 
G
Kong
 
X
, et al. 
Melanoma whole-exome sequencing identifies (V600E)B-RAF amplification-mediated acquired B-RAF inhibitor resistance.
Nat Commun
2012
, vol. 
3
 pg. 
724
 
120
Poulikakos
 
PI
Persaud
 
Y
Janakiraman
 
M
, et al. 
RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E).
Nature
2011
, vol. 
480
 
7377
(pg. 
387
-
390
)
121
Sári
 
E
Nagy
 
ZG
Baghy
 
K
, et al. 
Treatment of refractory hairy cell leukemia with a BRAF-inhibitor: lessons to be learnt.
Pathol Oncol Res
2014
, vol. 
20
 
4
(pg. 
973
-
980
)
122
Dietrich
 
S
Pircher
 
A
Endris
 
V
, et al. 
BRAF inhibition in hairy cell leukemia with low-dose vemurafenib.
Blood
2016
, vol. 
127
 
23
(pg. 
2847
-
2855
)
Sign in via your Institution