Mast cell leukemia (MCL) is a very rare form of aggressive systemic mastocytosis accounting for < 1% of all mastocytosis. It may appear de novo or secondary to previous mastocytosis and shares more clinicopathologic aspects with systemic mastocytosis than with acute myeloid leukemia. Symptoms of mast cell activation—involvement of the liver, spleen, peritoneum, bones, and marrow—are frequent. Diagnosis is based on the presence of ≥ 20% atypical mast cells in the marrow or ≥ 10% in the blood; however, an aleukemic variant is frequently encountered in which the number of circulating mast cells is < 10%. The common phenotypic features of pathologic mast cells encountered in most forms of mastocytosis are unreliable in MCL. Unexpectedly, non-KIT D816V mutations are frequent and therefore, complete gene sequencing is necessary. Therapy usually fails and the median survival time is < 6 months. The role of combination therapies and bone marrow transplantation needs further investigation.

Mastocytosis is a heterogeneous group of disorders characterized by the abnormal growth and accumulation of mast cells (MCs) in one or more organ systems.1,2  Diagnostic criteria and classification were recently updated by the WHO.1  This classification defines 7 disease variants (Table 1): cutaneous mastocytosis, indolent systemic mastocytosis (ISM), systemic mastocytosis (SM) with an associated clonal hematologic non–MC-lineage disease (SM-AHNMD), aggressive SM (ASM), MC leukemia (MCL), MC sarcoma, and extracutaneous mastocytoma. SM is defined by major and minor criteria. The diagnosis is established if at least one major and one minor or at least 3 minor criteria are present (Table 2). SM is subclassified according to MC burden, involvement of non-MC lineages, and aggressiveness of the disease (C findings).1,3  The clinical course is variable, ranging from asymptomatic with normal life expectancy to highly aggressive with poor prognosis.4-6 

Table 1

WHO classification of mastocytosis

VariantSubvariants
Cutaneous mastocytosis Urticaria pigmentosa 
 Maculopapular 
 Diffuse 
 Solitary mastocytoma of skin 
Indolent systemic mastocytosis Smoldering SM 
 Isolated bone marrow mastocytosis 
Systemic mastocytosis with an associated clonal hematologic non–MC-lineage disease SM-AML 
 SM-MDS 
 SM-MPD 
 SM-CEL or SM-HES 
 SM-CMML 
 SM-NHL 
Aggressive systemic mastocytosis  
MC leukemia  
MC sarcoma  
Extracutaneous mastocytom  
VariantSubvariants
Cutaneous mastocytosis Urticaria pigmentosa 
 Maculopapular 
 Diffuse 
 Solitary mastocytoma of skin 
Indolent systemic mastocytosis Smoldering SM 
 Isolated bone marrow mastocytosis 
Systemic mastocytosis with an associated clonal hematologic non–MC-lineage disease SM-AML 
 SM-MDS 
 SM-MPD 
 SM-CEL or SM-HES 
 SM-CMML 
 SM-NHL 
Aggressive systemic mastocytosis  
MC leukemia  
MC sarcoma  
Extracutaneous mastocytom  

Adapted from Swerdlow et al.1 

MC indicates mast cell; SM, systemic mastocytosis; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasia; CEL, chronic eosinophilic leukemia; HES, hypereosinophilic syndrome; CMML, chronic myelomonocytic leukemia; and NHL, non-Hodgkin lymphoma.

Table 2

WHO diagnostic criteria (2008) for systemic mastocytosis

Criteria
Major 
    Multifocal, dense infiltrates of MC (≥ 15 MC in aggregates) in BM and/or other extracutaneous organ(s) 
Minor 
    (1) > 25% of MC in the infiltrate of biopsy sections are spindle-shaped or have atypical morphology or, of all MC in the BM aspirate smears, > 25% are immature or atypical 
    (2) Activating point mutation at codon 816 of c-KIT in BM, blood, or another extracutaneous organ 
    (3) MC in BM, blood, or extracutaneous organs express CD2 and/or CD25 in addition to normal MC markers 
    (4) Total serum tryptase persistently exceeds 20 ng/mL* 
Criteria
Major 
    Multifocal, dense infiltrates of MC (≥ 15 MC in aggregates) in BM and/or other extracutaneous organ(s) 
Minor 
    (1) > 25% of MC in the infiltrate of biopsy sections are spindle-shaped or have atypical morphology or, of all MC in the BM aspirate smears, > 25% are immature or atypical 
    (2) Activating point mutation at codon 816 of c-KIT in BM, blood, or another extracutaneous organ 
    (3) MC in BM, blood, or extracutaneous organs express CD2 and/or CD25 in addition to normal MC markers 
    (4) Total serum tryptase persistently exceeds 20 ng/mL* 

Diagnosis of systemic mastocytosis requires the presence of the major criterion and 1 minor criterion or at least 3 minor criteria.

MC indicates mast cell; and BM, bone marrow.

*

Serum tryptase is not valid as a criterion if there is an associated clonal myeloid disorder.

MCL is a very rare form of ASM, involving < 0.5% of all mastocytosis patients in the French Reference Center for Mastocytosis (CEREMAST). It is characterized by the leukemic spread of MCs, with frequent and multiple organ involvement such as the liver, peritoneum, spleen, bone, and marrow. MCL shares more clinical and biologic aspects with ASM than with acute myeloid leukemia (AML).7  MCL diagnosis requires the presence of SM criteria with additional features including leukemic infiltration of bone marrow (BM) and/or blood by at least 20% high-grade MC1  as well as the infiltration of extracutaneous organs by neoplastic MC. The threshold > 20% of bone marrow mast cells (BMMCs) should be confirmed by cytologic analysis of BM smears thus avoiding false-positive diagnosis if the count is done on BM biopsies. “An aleukemic” form of MCL is frequent. This diagnosis is made if the number of circulating MCs is < 10%.1  Thus, a cytologic analysis of aspiration smear preparations, from both blood and marrow, is required to make a MCL diagnosis.7 

MCL can occur de novo or secondary to SM. Lim et al, in a large cohort of 342 patients with adult SM,6  reported an overall risk of transformation of 6%, in the majority of cases to AML (86%), or to MCL (13%). Transformation occurred most often from SM-AHNMD or ASM and was correlated to advanced age, history of weight loss, anemia, thrombocytopenia, hypoalbuminemia, and an excess of BM blasts.

A comprehensive immunophenotyping study has shown that an immature BMMC phenotype (CD25+/FceR+lo/FSClo/SSClo/CD45lo) in the absence of coexisting normal MCs in the bone marrow, is correlated with multilineage hematopoietic involvement by the KIT D816V mutation which might be associated with higher risk of progression to a more aggressive disease.8  Nevertheless, the prognostic significance of these observations for predicting clinical outcome in patients with SM requires further investigation. Therapeutic options are limited and the disease is most often fatal within a few months.6,7,9 

We performed a literature search using the PubMed database for all proven MCL cases, according to the WHO criteria,1  and propose here a review based on all published MCL cases since 1950 found, with 4 additional personal unpublished cases. Therefore, this review includes 51 MCL cases; in 41 cases, we had extensive clinical and biologic data which allowed us to analyze both de novo MCL (n = 30) as well as secondary MCL (n = 11) including a few cases with a previous history of pediatric mastocytosis (n = 4).

MCL was described as early as 1906 by Joachim.10  It is a very rare subcategory of SM accounting for approximately 1% of American adult SM,6  and < 0.5% of all French cases of mastocytosis reported in the CEREMAST database.

The median patient age at diagnosis was 52 years (range, 5-76 years) with a female predominance (sex ratio, F/M, of 1.5). Ethnic origin was available for 21 cases and consisted of 18 white and 3 Asian patients. No familial cases have been reported so far. Eleven patients (27%) had a history of mastocytosis and were considered as secondary MCL; whereas 30 patients (73%) were considered as having de novo MCL.

In this cohort, de novo MCL patients were older than patients with secondary MCL with a median age at diagnosis of, respectively, 51 years and 35 years. This apparent younger age in the secondary MCL group is because of the 4 patients with pediatric mastocytosis which had evolved into MCL. After exclusion of these 4 pediatric cases, the median age of both groups at MCL diagnosis was 51.5 years. Women presented more frequently with de novo MCL (60%). Ethnic origin was not different between the MCL subgroups, both mostly composed of whites (85%). After a short follow-up, 66% of the patients had died with a median survival time of 6 months (1-98 months).

MC activation symptoms (MCAS) were frequently observed (Table 3). They included flushes (60%), fever (52%), malaise (36%), diarrhea (28%), and tachycardia (20%). Pollakiuria (7%), neuropsychiatric symptoms (6%), and severe prefracturar osteoporosis (6%) were rare. Many patients suffered from asthenia (78%), severe weight loss of > 10% of total body weight (38%), and anorexia (20%). Symptoms of MC activation appeared more frequent among patients without KIT D816V mutation (85%) than in the group with this mutation (61%).

Table 3

Clinical and biologic characteristics of MCL patients: literature review

MCL
MCL with pediatric mastocytosis, n = 4
All, N = 51De novo, n = 30Secondary, n = 11
Median age at diagnosis, y (range) 52 (5-76) 51.5 (18-76) 35 (5-75) 24 (5-53) 
Female/Male 30/20 18/11 5/6 3/1 
Ethnicity, n (%)     
    White 18/21 (86) 13/15 (87) 5/6 (83) 2 (50) 
    Asiatic 3/21 (14) 2/15 (13) 1/6 (16) 1 (25) 
Familial mastocytosis, n (%) 0 (0) 0 (0) 0 (0) 0 (0) 
Median time from symptoms to diagnosis, mo (range) 5 (0-108) 3 (0-24) 6 (0-108) 4.5 (0-108) 
History of mastocytosis, n (%) 11/40 (27)  11 (100) 4 (100) 
Aleukemic MCL, n (%) 29/47 (62) 15 (50) 7 (70) 4 (100) 
Organ involvement, n (%)     
    Skin, UP 15/50 (30) 4 (14) 5 (45) 4 (100) 
    Hepatomegaly 32/47 (68) 21 (78) 6 (60) 2 (50) 
    Splenomegaly 38/49 (65) 23 (82) 9 (82) 4 (100) 
    Lymph nodes 16/43 (37) 11 (44) 3 (33) 1/2 (50) 
    Ascites 9/50 (18) 5 (17) 1 (9) 0 (0) 
Gastroduodenal ulcer, n (%) 11/38 (29) 10 (38.4) 0 (0) 0 (0) 
AHNMD, n (%) 4/40 (10) 3 (11) 
MCAS, n (%) 39/50 (78) 24 (83) 8 (73) 3 (75) 
KITmutation 28/51 (55) 15 (50) 5 (45) 2 (50) 
    D816V KIT mutation 13/28 (46) 6 (40) 1 (20) 
    WT KIT 2/28 (7) 1 (6) 1 (20) 
    502-503 KIT mutation 3/28 (11) 2 (13) 1 (20) 
    522/560/654 KIT mutation 3/28 (11) 1 (6) 2 (40) 
Abnormal cytogenetics, n (%)* 4/24 (17) 3 (27)   
Phenotype, n (%)     
    CD2+ 15/29 (52) 6 (46) 
    CD25+ 18/24 (75) 6 (50) 1/3 
    CD2/CD25+ 3/24 1/3 
    CD2/CD25 9/24 2/3 
    CD2+/CD25+ 13/24 
    CD2+/CD25 1/24 
Laboratory data, median (range)     
    Tryptase, ng/L 433 (21-2357) 433 (21-742) 250 (173-2357) 173.5 (173-200) 
    Hemoglobin, g/dL 9.9 (5.4-14) 9 (5.4-13.7) 11 (8.1-13.3) 12.5 (8.1-13.3) 
    Platelets, G/L 110 (5-318) 82 (5-202) 111 (30-150) 136 (101-318) 
    PMN, G/L 3.65 (0.99-15.3) 6.0 (0.99-14)  8.5 (1.7-15.3) 
    BM mastocytes % 50 (20-100) 60 (20-100) 60 (25-90) 60 (25-75) 
    Blood mastocytes % 5.5 (0-72) 7 (0-72) 0 (0-12) 2 (one tested) 
Outcome     
    Death, n (%) 33/48 (69) 24 (83) 5 (62) 2 (50%) 
    Median survival time, mo (range) 6 (0.5-98) 4 (0.5-24) 5 (1-18)  
MCL
MCL with pediatric mastocytosis, n = 4
All, N = 51De novo, n = 30Secondary, n = 11
Median age at diagnosis, y (range) 52 (5-76) 51.5 (18-76) 35 (5-75) 24 (5-53) 
Female/Male 30/20 18/11 5/6 3/1 
Ethnicity, n (%)     
    White 18/21 (86) 13/15 (87) 5/6 (83) 2 (50) 
    Asiatic 3/21 (14) 2/15 (13) 1/6 (16) 1 (25) 
Familial mastocytosis, n (%) 0 (0) 0 (0) 0 (0) 0 (0) 
Median time from symptoms to diagnosis, mo (range) 5 (0-108) 3 (0-24) 6 (0-108) 4.5 (0-108) 
History of mastocytosis, n (%) 11/40 (27)  11 (100) 4 (100) 
Aleukemic MCL, n (%) 29/47 (62) 15 (50) 7 (70) 4 (100) 
Organ involvement, n (%)     
    Skin, UP 15/50 (30) 4 (14) 5 (45) 4 (100) 
    Hepatomegaly 32/47 (68) 21 (78) 6 (60) 2 (50) 
    Splenomegaly 38/49 (65) 23 (82) 9 (82) 4 (100) 
    Lymph nodes 16/43 (37) 11 (44) 3 (33) 1/2 (50) 
    Ascites 9/50 (18) 5 (17) 1 (9) 0 (0) 
Gastroduodenal ulcer, n (%) 11/38 (29) 10 (38.4) 0 (0) 0 (0) 
AHNMD, n (%) 4/40 (10) 3 (11) 
MCAS, n (%) 39/50 (78) 24 (83) 8 (73) 3 (75) 
KITmutation 28/51 (55) 15 (50) 5 (45) 2 (50) 
    D816V KIT mutation 13/28 (46) 6 (40) 1 (20) 
    WT KIT 2/28 (7) 1 (6) 1 (20) 
    502-503 KIT mutation 3/28 (11) 2 (13) 1 (20) 
    522/560/654 KIT mutation 3/28 (11) 1 (6) 2 (40) 
Abnormal cytogenetics, n (%)* 4/24 (17) 3 (27)   
Phenotype, n (%)     
    CD2+ 15/29 (52) 6 (46) 
    CD25+ 18/24 (75) 6 (50) 1/3 
    CD2/CD25+ 3/24 1/3 
    CD2/CD25 9/24 2/3 
    CD2+/CD25+ 13/24 
    CD2+/CD25 1/24 
Laboratory data, median (range)     
    Tryptase, ng/L 433 (21-2357) 433 (21-742) 250 (173-2357) 173.5 (173-200) 
    Hemoglobin, g/dL 9.9 (5.4-14) 9 (5.4-13.7) 11 (8.1-13.3) 12.5 (8.1-13.3) 
    Platelets, G/L 110 (5-318) 82 (5-202) 111 (30-150) 136 (101-318) 
    PMN, G/L 3.65 (0.99-15.3) 6.0 (0.99-14)  8.5 (1.7-15.3) 
    BM mastocytes % 50 (20-100) 60 (20-100) 60 (25-90) 60 (25-75) 
    Blood mastocytes % 5.5 (0-72) 7 (0-72) 0 (0-12) 2 (one tested) 
Outcome     
    Death, n (%) 33/48 (69) 24 (83) 5 (62) 2 (50%) 
    Median survival time, mo (range) 6 (0.5-98) 4 (0.5-24) 5 (1-18)  

MCL indicates mast cell leukemia; UP, urticaria pigmentosa; AHNMD, associated clonal hematologic non–MC-lineage disease; MCAS, mast cell activation syndrome including flushes, fever, malaise, diarrhea, and tachycardia; WT, wild type; PMN, polymorphonuclear; and BM, bone marrow.

*

Abnormal cytogenetics, 5q deletion (2 patients with AHNMD), t(10;16) (q22;q13q22), and t(8;21)(q22;q22) (2 patients).

No information on the other 2 patients at 4 months.

On physical examination, hepatomegaly and splenomegaly were the most frequent clinical signs and were present, respectively, in 68% and 65% of the patients with a median spleen enlargement of 21 cm (ranging from 13 to 25 cm). Lymph node enlargement was found in 37% and skin involvement was only present in 30% of all cases. Gastrointestinal manifestations frequently included gastroduodenal ulcers (29%)11-20  which were often complicated by gastrointestinal hemorrhage (64%). Gastroduodenal ulcers seem to be more frequently associated with the KIT D816V (12.5%) mutation than with other KIT mutations (0%). Ascites and portal hypertension were present in 18% and 16% of cases, respectively (Table 3).

De novo or secondary MCL

A comparison between de novo (n = 30) and secondary (n = 11) MCL subgroups revealed more frequent cutaneous manifestations (urticaria pigmentosa) in secondary MCL. In secondary MCL, this is certainly because of the previous mastocytosis. Other clinical features—including hepatosplenomegaly, lymph node enlargement, ascites, and symptoms of MC activation—were comparable between the 2 groups. Interestingly, no gastroduodenal ulcers were observed in patients with secondary MCL whereas they were found in 38% of de novo MCL cases. As gastroduodenal ulcers are linked to the release of histamine by pathologic MC,21,22  we could hypothesize that in de novo MCL, pathologic MC are highly aggressive, proliferate rapidly, infiltrate all organs (including the digestive tract), and degranulate easily, releasing large amounts of histamine, leading to gastroduodenal ulcer formation.

Mast cell leukemia and clonal hematologic non–MC-lineage disease

MCL is rarely associated with clonal hematologic non-MC disease. Only 4 cases (8%) were found in the literature. AHNMD were mainly myelodysplastic syndromes (n = 3) and a chronic myelomonocytic leukemia (n = 1). Clinical symptoms were not different from other MCL manifestations, and the aleukemic form of the disease was predominant. AHNMD was only found in the KIT D816V mutation subgroup (n = 3/7). Cytogenetic studies revealed a 5q deletion in 2 cases which was more likely related to the MDS clone rather than to MC clones. The prognosis was also poor and not different from MCL without AHNMD (Tables 34).

Table 4

Patients' characteristics according to KIT sequencing: literature review

No D816V mutation, n = 15§D816V mutations, n = 13
Media age at diagnosis, y (range) 46 (18-66) 58 (52-76) 
Female/Male 10/4 8/5 
Ethnicity, n (%)   
    White 6 (85) 3 (75) 
    Asiatic 1 (15) 1 (25) 
Familial mastocytosis 0 (0) 0 (0) 
Median time from symptoms to diagnosis, mo (range) 5 (0-108) 5 (0-10) 
History of mastocytosis, n (%) 4 (33) 1 (15) 
Aleukemic MCL, n (%) 10 (83) 8 (61) 
Organ involvement, n (%)   
    Skin (UP) 4 (28) 3 (23) 
    Hepatomegaly 8 (57) 8 (66) 
    Splenomegaly 10 (71) 10 (77) 
    Lymph nodes 3 (25) 3 (23) 
    Ascites 2 (15) 5 (38) 
Gastroduodenal ulcer, n (%) 0 (0) 1 (12.5) 
AHNMD, n (%) 0 (0) 3 (43) 
MCAS, n (%) 12 (85) 8 (61) 
KITmutation (%)*   
    D816V KIT mutation  13 (100) 
    WT KIT 2 (25)  
    Exon 9 mutation 3 (37.5)  
    Exon 10 mutation 1 (12.5)  
    Exon 11 mutation 1 (12,5)  
    Exon 13 mutation 1 (12.5)  
    Not available 7 (46)  
Abnormal cytogenetics, n (%) 1 (12.5) 1 (8) 
(t(8;21)(q22;q22))  
Phenotype, n (%)   
    CD2+ 4 (33) 8 (80) 
    CD25+ 4 (36) 11 (92) 
    CD2/CD25+ 1 (8) 1 (8) 
    CD2/CD25 5 (42) 1 (8) 
    CD2+/CD25+ 3 (25) 8 (66) 
    CD2+/CD25 1 (8) 0 (0) 
Biological data, median (range)   
    Tryptase, ng/L 400 (173-742) 200 (111-2357) 
    Hemoglobin, g/dL 10 (5.4-12.5) 10 (7-13.7) 
    Platelets, ×109/L 117.5 (45-318) 74 (5-210) 
    PMN, ×109/L 1.5 (1.1-4) 10.5 (3.5-14) 
    BM mastocytes, % (range) 64 (23-100) 60 (30-90) 
    Blood mastocytes, % (range) 3.5 (1-31) 3.5 (1-20) 
Outcome   
    Death, n (%) 9 (69) 6 (46) 
    Median survival time, mo (range) 5 (0.5-29) 5.5 (2-23) 
No D816V mutation, n = 15§D816V mutations, n = 13
Media age at diagnosis, y (range) 46 (18-66) 58 (52-76) 
Female/Male 10/4 8/5 
Ethnicity, n (%)   
    White 6 (85) 3 (75) 
    Asiatic 1 (15) 1 (25) 
Familial mastocytosis 0 (0) 0 (0) 
Median time from symptoms to diagnosis, mo (range) 5 (0-108) 5 (0-10) 
History of mastocytosis, n (%) 4 (33) 1 (15) 
Aleukemic MCL, n (%) 10 (83) 8 (61) 
Organ involvement, n (%)   
    Skin (UP) 4 (28) 3 (23) 
    Hepatomegaly 8 (57) 8 (66) 
    Splenomegaly 10 (71) 10 (77) 
    Lymph nodes 3 (25) 3 (23) 
    Ascites 2 (15) 5 (38) 
Gastroduodenal ulcer, n (%) 0 (0) 1 (12.5) 
AHNMD, n (%) 0 (0) 3 (43) 
MCAS, n (%) 12 (85) 8 (61) 
KITmutation (%)*   
    D816V KIT mutation  13 (100) 
    WT KIT 2 (25)  
    Exon 9 mutation 3 (37.5)  
    Exon 10 mutation 1 (12.5)  
    Exon 11 mutation 1 (12,5)  
    Exon 13 mutation 1 (12.5)  
    Not available 7 (46)  
Abnormal cytogenetics, n (%) 1 (12.5) 1 (8) 
(t(8;21)(q22;q22))  
Phenotype, n (%)   
    CD2+ 4 (33) 8 (80) 
    CD25+ 4 (36) 11 (92) 
    CD2/CD25+ 1 (8) 1 (8) 
    CD2/CD25 5 (42) 1 (8) 
    CD2+/CD25+ 3 (25) 8 (66) 
    CD2+/CD25 1 (8) 0 (0) 
Biological data, median (range)   
    Tryptase, ng/L 400 (173-742) 200 (111-2357) 
    Hemoglobin, g/dL 10 (5.4-12.5) 10 (7-13.7) 
    Platelets, ×109/L 117.5 (45-318) 74 (5-210) 
    PMN, ×109/L 1.5 (1.1-4) 10.5 (3.5-14) 
    BM mastocytes, % (range) 64 (23-100) 60 (30-90) 
    Blood mastocytes, % (range) 3.5 (1-31) 3.5 (1-20) 
Outcome   
    Death, n (%) 9 (69) 6 (46) 
    Median survival time, mo (range) 5 (0.5-29) 5.5 (2-23) 

MCL indicates mast cell leukemia; UP, urticaria pigmentosa; AHNMD, associated clonal hematologic non–MC-lineage disease; MCAS, mast cell activation syndrome; PMN, polymorphonuclear; and BM, bone marrow.

*

Exon 9 mutation at codons 501-502-503; exon 10 mutation at codon 522, exon 11 mutation at codon 560, exon 13 mutation at codon 654; not available by KIT sequencing.

Abnormal cytogenetics, 5q deletion (2 patients with AHNMD), t(10;16) (q22;q13q22) and t(8;21)(q22;q22) (2 patients).

One missing.

§

Two patients with previous mast cell sarcoma and 2 patients with previous pediatric mastocytosis.

Table 5

Literature review of MCL cases

AuthorsYear of publicationNo. of patientsAge at diagnosis, yWHO MCLDe novo MCLAleukemic MCLc-KIT mutationsSurvival time, mo
Hissard et al83  1951 47 Yes No Yes NA NA 
Waters et al37  1956 Yes No Yes NA 
Efrati et al15  1957 52 Yes Yes No NA 
Friedman et al16  1958 NA Yes Yes No NA 
Brinkmann et al84  1959 51 Yes Yes NA NA NA 
Szweda et al19  1962 50 Yes Yes Yes NA 
Mutter et al85  1963 59 Yes Yes Yes NA 
Daniel et al14  1975 39 Yes Yes No NA 
Clancy et al86  1976 46 Yes Yes NA NA 
Coser at al12  1980 54 Yes Yes No NA 
Kimura et al87  1979 39 Yes Yes No NA NA 
Horny et al88  1986 75 Yes No No NA 
Dalton et al13  1986 57 Yes Yes No NA 
Travis et al20  1986 52 Yes Yes No NA 
Baghestanian et al11  1996 34 Yes Yes No NA 
Le Cam et al18  1997 44 Yes Yes Yes NA 
Escribano et al89  1997 67 Yes Yes Yes NA 16 
Kyriakou et al17  1998 72 Yes Yes Yes NA 
Pauls et al90  1999 48 Yes  No Negative 
Lin et al79  2002 25 Yes No No NA NA 
Horny et al91  2002 48 Yes Yes Yes Negative NA 
Pardanani et al65  2003 NA Yes Yes NA Negative 0, 5 
Chen et al82  2003 18 Yes Yes Yes Negative  
Akin et al92  2004 25 Yes No Yes Phe522Cys  
Noack et al9  2004 75 Yes Yes Yes D816V  
Gotlib et al73  2005 48 Yes Yes No D816V 
Penack et al46  2005 65 Yes Yes No D816V  
Valentini et al7  2007 10 56* Yes No Yes  24§ 
Krauth et al93  2007 35 Yes No Yes WT  
Brcic et al77  2007 Yes No No NA  
Aichberger et al72  2008 71 Yes Yes No D816V  
Yoshida et al94  2009 52 Yes Yes No D816V  
Arredondo et al24  2010 61 Yes No Yes Negative  
Mital et al30  2011 53 Yes Yes Yes dup 502-503 18 
Spector et al33  2011 42 Yes No Yes V654A  
Chantorn et al36  2012 23 Yes No Yes NA  
Georgin-Lavialle et al29  2012 66 Yes Yes Yes del(501;502)  
Georgin-Lavialle et al32  2012 42 Yes No NA val 560 gly  
Joris et al95  2012 52 Yes Yes Yes WT 
Hermine et al Unpublished 29 Yes Yes Yes dup 501-502 
Damaj et al Unpublished 76, 58 Yes Yes Yes D816V (2) 7, 11 
AuthorsYear of publicationNo. of patientsAge at diagnosis, yWHO MCLDe novo MCLAleukemic MCLc-KIT mutationsSurvival time, mo
Hissard et al83  1951 47 Yes No Yes NA NA 
Waters et al37  1956 Yes No Yes NA 
Efrati et al15  1957 52 Yes Yes No NA 
Friedman et al16  1958 NA Yes Yes No NA 
Brinkmann et al84  1959 51 Yes Yes NA NA NA 
Szweda et al19  1962 50 Yes Yes Yes NA 
Mutter et al85  1963 59 Yes Yes Yes NA 
Daniel et al14  1975 39 Yes Yes No NA 
Clancy et al86  1976 46 Yes Yes NA NA 
Coser at al12  1980 54 Yes Yes No NA 
Kimura et al87  1979 39 Yes Yes No NA NA 
Horny et al88  1986 75 Yes No No NA 
Dalton et al13  1986 57 Yes Yes No NA 
Travis et al20  1986 52 Yes Yes No NA 
Baghestanian et al11  1996 34 Yes Yes No NA 
Le Cam et al18  1997 44 Yes Yes Yes NA 
Escribano et al89  1997 67 Yes Yes Yes NA 16 
Kyriakou et al17  1998 72 Yes Yes Yes NA 
Pauls et al90  1999 48 Yes  No Negative 
Lin et al79  2002 25 Yes No No NA NA 
Horny et al91  2002 48 Yes Yes Yes Negative NA 
Pardanani et al65  2003 NA Yes Yes NA Negative 0, 5 
Chen et al82  2003 18 Yes Yes Yes Negative  
Akin et al92  2004 25 Yes No Yes Phe522Cys  
Noack et al9  2004 75 Yes Yes Yes D816V  
Gotlib et al73  2005 48 Yes Yes No D816V 
Penack et al46  2005 65 Yes Yes No D816V  
Valentini et al7  2007 10 56* Yes No Yes  24§ 
Krauth et al93  2007 35 Yes No Yes WT  
Brcic et al77  2007 Yes No No NA  
Aichberger et al72  2008 71 Yes Yes No D816V  
Yoshida et al94  2009 52 Yes Yes No D816V  
Arredondo et al24  2010 61 Yes No Yes Negative  
Mital et al30  2011 53 Yes Yes Yes dup 502-503 18 
Spector et al33  2011 42 Yes No Yes V654A  
Chantorn et al36  2012 23 Yes No Yes NA  
Georgin-Lavialle et al29  2012 66 Yes Yes Yes del(501;502)  
Georgin-Lavialle et al32  2012 42 Yes No NA val 560 gly  
Joris et al95  2012 52 Yes Yes Yes WT 
Hermine et al Unpublished 29 Yes Yes Yes dup 501-502 
Damaj et al Unpublished 76, 58 Yes Yes Yes D816V (2) 7, 11 

MCL indicates mast cell leukemia; NA, not available; and WT, wild type.

*

Median age.

Seven of 10 cases were aleukemic MCL.

Six patients were D816V c-KIT positive and 2 patients were D816V c-KIT negative.

§

Median survival time at last reported follow-up.

Blood and marrow characteristics

The percentage of circulating MC ranged from 0% to 72% of all leukocytes. Aleukemic MCL was the most frequent form found, making up 62% of all cases; 70% of secondary MCL and 55% of de novo MCL. Anemia was always present at diagnosis with a median hemoglobin value of 9.9 g/dL and a lower hemoglobin level in de novo (9 g/dL) than in secondary (11 g/dL) MCL patients. Thrombocytopenia was also frequent. The median platelet count was 110 G/L in the whole group of patients with lower median platelet counts in the de novo than in the secondary MCL patients (82 vs 111 G/L).

Levels of polymorphonuclear cells were within normal values with a median count of 3.65 G/L. The median bone marrow mast cell infiltration was 50%, ranging from 20% to 100% with no difference between de novo and secondary MCL subgroups. Serum tryptase levels were always increased with a median level of 433 ng/L (Table 3).

Morphology

Normal MCs and their progenitors display 4 morphologic stages of differentiation in BM smears stained with May-Grünwald-Giemsa: (1) ungranulated but tryptase-positive blast cells, (2) metachromatic blast, (3) promastocyte, and (4) typically round mononuclear mature MC with metachromatic granules in the cytoplasm.

Malignant MC from MCL patients may exhibit the features classically encountered in mastocytosis with mature MC displaying morphologic abnormalities such as elongated cytoplasmic extensions (spindle-shaped MC), an eccentric oval nucleus, hypo-granulated cytoplasm, and possible focal granule accumulations (Figure 1).

Figure 1

Cytologic abnormalities of mast cells in MCL. Mast cells from MCL range from mature mast cells (A-D) to more immature cells with (E-I) promastocytes and (J-K) even metachromatic or (K) ungranulated blasts. (A-B) Circulating round mast cells; (C) spindle-shaped mast cell; (D) packed and polar granule aggregates; (E-G) degranulated/hypogranulated promastocytes; (H-I) coalescent granules; (I) lacunar cytoplasmic areas; (J-K) undifferentiated immature cells with metachromatic granules and prominent nucleoli.

Figure 1

Cytologic abnormalities of mast cells in MCL. Mast cells from MCL range from mature mast cells (A-D) to more immature cells with (E-I) promastocytes and (J-K) even metachromatic or (K) ungranulated blasts. (A-B) Circulating round mast cells; (C) spindle-shaped mast cell; (D) packed and polar granule aggregates; (E-G) degranulated/hypogranulated promastocytes; (H-I) coalescent granules; (I) lacunar cytoplasmic areas; (J-K) undifferentiated immature cells with metachromatic granules and prominent nucleoli.

Close modal

MCL may also typically exhibit immature forms of MC, seldom seen in other types of mastocytosis, reminiscent of the whole morphologic differentiation chronology. Promastocytes, metachromatic blasts, and even ungranulated blasts are seen with specific abnormalities such as polarized or coalescent granules, cytoplasmic lacunar areas, prominent nucleoli, fine nuclear chromatin, and bi- or multilobed nuclei (Figure 1).1,3 

MCL differs from all other forms of SM in that diagnosis can rely on cytology alone.1,7,23  In addition to qualitative features and to make the diagnosis, on cytologic analysis, MC should represent > 20% of nucleated cells in marrow aspirates or > 10% in peripheral blood. The diagnosis of an aleukemic variant of MCL is met if the count of MC on peripheral blood is < 10%.

Beyond these criteria for diagnosis, the detection of circulating mature MC is abnormal and must prompt investigation for MCL.

Immunophenotype

In mastocytosis, abnormal antigen expression on MC is identified either by flow cytometry or immunohistochemistry. Expression of CD2 and/or CD25 is the most common feature of abnormal MC and is recognized as a minor criteria for diagnosis of mastocytosis.1 

MC from MCL can express both CD25 and CD2, although the expression of CD25 and particularly CD2 was not found in, respectively, 25% and 48% of cases, and one-third of MCL cases reported in the literature had a double-negative CD2/CD25 immunophenotype (Table 3).24-26  In line with the classic association of the KIT D816V mutation with the expression of CD2/CD25 on MC in SM, the positive coexpression of CD2 and CD25 was very frequent among MCL patients who were KIT D816V positive (66%) compared with those who were KIT D816V negative (25%). Thus, the common feature of pathologic MC encountered in most forms of mastocytosis is unreliable in MCL.

Recently, it has been proposed that aggressive forms of mastocytosis, including MCL, exhibit an immature phenotype, in contrast to the more indolent forms which are characterized by an activated immunophenotype.8,27  It follows that MC from MCL may express immaturity markers such as CD123, CD34, HLA-DR, and display reduced expression of CD117 and FceRI. The immature phenotype is correlated with multilineage KIT mutations in bone marrow and a poor prognosis.8,27  From this observation, arises the concept that the occurrence of a KIT mutation in an early progenitor may result in multilineage involvement, early blockade of maturation of MC, and aggressive disease. In contrast, mutations that occur in a later-stage progenitor remain MC-restricted and lead to late maturation blockade with a mature and activated MC immunophenotype.27,28  At present, the significance of the phenotypic heterogeneity according to different histologic, clinical, and genetic features remains unresolved.

Cytogenetic and molecular characteristics

Cytogenetic studies were available for 23 of 51 reported cases. A normal cytogenetic profile was found in the majority of cases (83%). Two patients (9%) had a 5q deletion, and were diagnosed as having MCL associated with myelodysplastic syndrome,7  with the deletion of 5q probably being related to the myelodysplastic clone rather than to MCL itself. Two other patients with de novo MCL had t(10;16)(q22;q13q22) and t(8;21)(q22;q22). According to these findings, there is no recurrent cytogenetic abnormality in MCL.

c-KIT mutations

The KIT D816V mutation was detected in 13 of 28 patients studied (Table 4). In 8 patients without KIT D816V, the entire KIT gene was sequenced. In 2 cases, KIT was wild type (WT), whereas in the other 6 cases, mutations were detected in exon 9 (n = 3),29,30  exon 10 (n = 1),31  exon 11 (n = 1),32  and exon 13 (n = 1).33  It is noteworthy that these mutations are also found in stromal gastrointestinal tumors, which are aggressive. Most of these non-D816V mutations have only recently been described, 1 in 2004 and 6 since 2011. This may explain why in previous literature reviews, made when the KIT gene had not been entirely sequenced, the KIT D816V mutation was described as the most frequently encountered mutation. We now know that in MCL this is not so.

In the non-KIT D816V mutation subgroup, 4 patients had a history of mastocytosis including 2 MC sarcomas and 2 with pediatric cutaneous mastocytosis, that is, 33% of the patients studied, in contrast to only 1 of 7 patients in the KIT D816V mutation group (15%).

Death occurred more frequently in the subgroup without D816V mutation (69%) than for those with the D816V mutation (46%), although the median survival time did not differ.

TET2

TET2 (Ten-Eleven-Translocation 2) is 1 of 3 homologous human proteins (TET1, TET2, and TET3) that may play a role in the epigenetic regulation of transcription. TET2 mutations have been reported in 29% of systemic mastocytosis, with 15% ISM, 40% ASM, and 35% being SM-AHNMD.34  Recent data from our group reporting on 74 patients with cutaneous and SM, including 4 with MCL, showed the TET2 mutation as a potential marker to diagnose and predict severe forms of the disease.35 

Interestingly, 4 patients developed MCL after pediatric mastocytosis (Table 6).30,31,36,37  In 1 case, diffuse cutaneous mastocytosis evolved into MCL, leading to death in childhood37  (KIT gene sequencing was not available at that time). In 2 other cases,30,31  maculopapular cutaneous mastocytosis occurred in childhood and developed into MCL 25 and 53 years later. Mutations in exon 9 and 10 were found, with no or low expression of CD2 and CD25. Both patients were treated with imatinib, obtained partial response, and were alive at 5 and 18 months, respectively. The last interesting case36  is that of mastocytoma diagnosed at 5 years of age. After complete surgical excision for aesthetic reasons at age 19, the patient developed aggressive systemic mastocytosis and MCL. Allogeneic stem cell transplantation was ineffective and was followed by rapid disease progression and death. This last case demonstrates that mastocytoma can evolve into MCL. The description of frequent non-D816V KIT mutations among children with cutaneous mastocytosis38  may indicate that mutations in exon 9 and 10 of KIT can lead to severe forms of mastocytosis. Taken together, these 4 cases highlight the risk of evolution of pediatric mastocytosis to MCL.

Table 6

Main features of MCL cases with a previous history of pediatric mastocytosis

AuthorYearSexAge, mo*TypeClinical evolutionMC phenotypeKIT sequencingCytological featuresTreatment
Waters37  1957 DCM 5 y: MCL transformation ND ND Mast cells appeared as large polymorphic cells with acidophilic cytoplasm and large rounded nucleus, few metachromatic granules were discerned in the cytoplasm. None 
     Death within 3 mo due to diffuse hemorrhage.     
Akin31  2004 MPCM 22 y: deterioration of general status CD2neg Phe 522 Cys Round, highly granulated and centrally located nucleus IFNα: no response 
     25 y: ASM CD25neg (exon 10)  Imatinib: PR 
     And 6 mo later: MCL    FU: 5 mo 
Mital30  2011 12 MPCM 47 y: unexplained splenomegaly CD2low 502-503 Dup Large mast cells 2CDA: no response 
     53 y: aleukemic MCL CD25low (exon 9)  Imatinib: PR 
     56 y: MCL evolution    FU: 18 mo 
Chantorn36  2012 Mastocytoma 10 y: MPCM CD2 ND Rare spindle shaped mast cells on bone marrow histology poly chemotherapy, 
     19 y: mastocytoma exeresis CD25+  Atypically spindle shaped morphology on BM smear allo-SCT, Steroids 
     23 y: ASM; MCL and death 5 mo later    Tacrolimus: 
         No response 
AuthorYearSexAge, mo*TypeClinical evolutionMC phenotypeKIT sequencingCytological featuresTreatment
Waters37  1957 DCM 5 y: MCL transformation ND ND Mast cells appeared as large polymorphic cells with acidophilic cytoplasm and large rounded nucleus, few metachromatic granules were discerned in the cytoplasm. None 
     Death within 3 mo due to diffuse hemorrhage.     
Akin31  2004 MPCM 22 y: deterioration of general status CD2neg Phe 522 Cys Round, highly granulated and centrally located nucleus IFNα: no response 
     25 y: ASM CD25neg (exon 10)  Imatinib: PR 
     And 6 mo later: MCL    FU: 5 mo 
Mital30  2011 12 MPCM 47 y: unexplained splenomegaly CD2low 502-503 Dup Large mast cells 2CDA: no response 
     53 y: aleukemic MCL CD25low (exon 9)  Imatinib: PR 
     56 y: MCL evolution    FU: 18 mo 
Chantorn36  2012 Mastocytoma 10 y: MPCM CD2 ND Rare spindle shaped mast cells on bone marrow histology poly chemotherapy, 
     19 y: mastocytoma exeresis CD25+  Atypically spindle shaped morphology on BM smear allo-SCT, Steroids 
     23 y: ASM; MCL and death 5 mo later    Tacrolimus: 
         No response 

MCL indicates mast cell leukemia; M, male; DCM, diffuse cutaneous leukemia; ND, not done; F, female; MPCM, maculopapular cutaneous mastocytosis; ASM, aggressive systemic mastocytosis; IFNa, interferon alpha; PR, partial response; FU, follow-up; BM, bone marrow; and allo-SCT, allogeneic stem cell transplantation.

*

Age at diagnosis of pediatric mastocytosis.

Type of pediatric mastocytosis at diagnosis.

In general, treatments of SM can be divided into 2 broad categories: (1) those intended to control mast cell mediator-related symptoms and reduce disabilities associated with the disease and (2) those intended to limit MC burden in aggressive forms and increase survival by the use of cytoreductive therapy.39  To date, there is no approved standard therapy. For MCL, few options are available for treatment and because of the rarity of the disease very few clinical trials address the question.

Steroids

Corticosteroids are often tried, at least at the onset of the disease. High-dose corticosteroids may induce a reduction in MC burden and improve clinical symptoms including flushing, ascitis, pain, cardiac failure as well as cytopenia. However, their effect is usually transient and precautions should be taken. Proton pump inhibitor therapy should be systematically started concurrently with the introduction of corticosteroids because of the increased risk of gastrointestinal bleeding, particularly in this context.

Interferon-α

In contrast to ASM and ISM,40-45  reports on the use of interferon in MCL are scarce. In one report of 6 patients with MCL who were treated with interferon, only 2 patients obtained a partial response and this was of short duration.46 

2-Chloro-deoxy-adenosine

The rationale behind the use of 2-Chloro-deoxy-adenosine (2-CdA) for the treatment of SM is based on the fact that MC are derived from hematopoietic stem cells and may share a common progenitor with monocytes,47  combined with the knowledge that the drug is particularly toxic to monocytes, both in vitro and in vivo.48 

While the potential value of 2-CdA in the treatment of ASM has been confirmed in several studies,49,50  in MCL, 2-CdA has no24,30  or little7  activity, although in rare cases a prolonged partial response has been observed.9 

Ten patients with a diagnosis of MCL were treated with 2-CdA. Responses were obtained in 3 of them (30%) who were still alive at 17, 24, and 48 months (with no subsequent therapy for 2 patients).7,24,33,51 

In our experience, when 68 patients with the diagnosis of SM were treated with single-agent Cladribine at a dose of 0.14 mg/kg/d for 5 or 7 consecutive days, no significant effects were observed in the subgroup of 3 patients with MCL. Combination therapies with high-dose cytarabine (O.H., oral communication at second annual meeting of reference centers, September 16, 2012), mTOR inhibitors,52  or tyrosine kinase inhibitor (TKI)53  need to be evaluated in patients with MCL.

Targeted therapies or tyrosine kinase inhibitors

The mutation in c-KIT codon 816 found in adult human mastocytosis causes constitutive activation of the KIT kinase.54  Different classes of KIT activating mutations respond differentially to KIT inhibitors depending on the site and the type of mutation.55-58  The presence and/or the type of KIT mutation have clinical prognostic importance.59  Thus, data detailing mutations are useful in predicting drug resistance and supporting individualization of therapy based on the response of specific mutant proteins to specific drugs.

Imatinib.

The rationale behind the use of imatinib in SM stems from its in vitro inhibitory activity of both WT and certain (eg, V560G; F522C) but not all D816V classes of constitutively active KIT receptors.31,60-65  In October 2006, these data led the US Food and Drug Administration to approve the use of imatinib for adult patients with ASM lacking the D816V KIT mutation, or with unknown KIT mutational status, at a dose of 400 mg daily.

Nine patients with a diagnosis of MCL and treated by imatinib have so far been reported. Four (2 adults and 2 children) of them obtained a partial response (44%). The KIT mutations were Phe522Cys and Dup 502-503 in 2 responding patients and another responder had WT KIT. The fourth responding patient had the D816V KIT-positive mutation and was alive after 48 months on imatinib alone.7,30,31,33,65 

Masitinib.

Masitinib (AB1010) is a tyrosine kinase inhibitor which, in vitro, potently and selectively inhibits the mutated form in the juxtamembrane region of the KIT receptor and the KIT WT receptor. It also inhibits the PDGF receptor and the mutated fibroblast growth factor receptor (FGFR3). At the cellular level, masitinib is a more selective inhibitor of KIT WT-dependent cell proliferation (IC50 of 0.2μM), than imatinib mesilate (IC50 of 0.6μM). Masitinib is in general well-tolerated at the doses of 7.5-12 mg/kg/d used for oncology indications, and up to 6 mg/kg/d in nononcology indications.66  The most frequent adverse events—nausea/vomiting, edema, rash, and diarrhea—are manageable, mild to moderate, and mostly resolve under treatment; nevertheless, 1.3% of patients experienced severe neutropenia that might be because of hypersensitivity of neutrophils to KIT inhibition.

Data on MCL patients is scarce. Two MCL patients not carrying the D816V KIT mutation were treated with masitinib.29  One of them obtained partial response, relapsed 7 months after initiation of masatinib, and died from disease progression at 11 months. This drug needs to be further evaluated in this setting.

Dasatinib.

Dasatinib is an oral bioavailable SRC/ABL inhibitor that has activity against multiple imatinib-resistant BCR-ABL isoforms in vitro and in vivo. It demonstrates significant inhibitory activity, in the nanomolar range, against both wild-type KIT and the KIT D816V mutation67  as well as against autophosphorylation of the juxtamembrane domain of mutant KIT and KIT-dependent activation of downstream pathways68  in in vitro and cell-based kinase assays. Growth inhibition of a human mastocytosis cell line carrying KIT D816V has also been achieved with this drug.67  However, its activity has only been modest in clinical settings.51,69-71 

Three patients with MCL have been treated with dasatinib without any efficacy and patients died from disease progression at 1, 3, and 9 months after diagnosis.29,33,72 

PKC 412.

PKC412 (Midostaurin; Novartis), a small molecule inhibitor of multiple type III TK receptors involved in hematopoiesis and leukemia, inhibits all major isoforms of protein kinase C as well as the TK associated with the vascular endothelial growth factor receptor. It can also exert inhibitory activity on other mutated tyrosine kinases implicated in a variety of diseases, including KIT (systemic mast cell disease, gastrointestinal stromal tumors, PDGFR, or FGFR1 myeloproliferative disorders). Midostaurin has shown strong inhibitory activity on neoplastic human MC carrying the KIT D816V mutation in preclinical and clinical settings.53,73,74  However, data reporting on the efficacy of midostaurin in MCL is scarce.

Our experience with PKC-412 is based on 21 patients treated for advanced SM. Four of them had been diagnosed with MCL (n = 2) or MCL with AHNMD (n = 2). Partial responses were obtained in 2 patients (Damaj G.D., and O.H. and O.C., unpublished data). Preliminary results from the phase 2 study of KIT inhibitor midostaurin showed an overall response rate in, advanced SM, of 60% with the majority being major responses (52.5%). Of the 7 patients with MCL (3 patients with AHNMD), 4 (57%) patients achieved major responses, including 3 ongoing incomplete remissions (19+ months in 2 patients and 32+ months in 1 patient). The overall survival in MCL patients was 22.6 months.75  These results are encouraging and combination strategies with chemotherapy or other targeted therapies could be of interest (Figure 2).

Chemotherapy

Various cytostatic drugs have been shown to induce apoptosis and inhibit proliferation of the human MCL cell line HMC-1.76  However, whether patients with MCL may benefit from combination polychemotherapy remains unknown. AML-type induction chemotherapy has been used in 6 of 51 reported MCL patients. However, the response rate was not available for all patients, the median survival time was 7 months, and all patients died between 2 and 29 months from progression or multiorgan failure.77-79  These results suggest a lack of efficacy of AML-type induction polychemotherapy in MCL patients.

Bone marrow transplantation

Allogeneic stem cell transplantation (allo-SCT) could play a potential curative role in MCL. Some reports have shown a positive role of the graft compared with the mast cell effect in decreasing MC burden after the transplant. However, allo-SCT has limited ability to completely eradicate the disease.80,81 

Rapid engraftment of donor MCs has been documented after allo-SCT. Thus, caution should be applied when evaluating the neoplastic MC burden after allo-SCT, and MC should be analyzed carefully to differentiate reactive donor MC from the potential persistence of the neoplastic MC clone.82 

Seven cases of patients who received allo-SCT have been reported. Sustained remission was not achieved in any of them. One patient, however, obtained a response but died 23 months after transplantation in complete remission.7,18,36,81,82 

Recently, Ustun et al (oral communication, European Competence Network on Mastocytosis meeting, September 2012), reported on a retrospective series of 23 patients with SM who received allo-SCT either after myeloablative conditioning (n = 14) or a fludarabine-based reduced-intensity regimen. The best responses were obtained in SM-AHNMD (AML/MDS) and the worst were in MCL patients. They confirmed the results of a previous report in which only rarely was CR obtained after transplantation. Therefore, in the absence of sufficient data in MCL patients, allo-SCT remains to be proven and further evaluation of this strategy in these patients is needed, alone or in combination with drugs to obtain greater reduction in disease burden, which may translate to better long-term control of the disease (Figure 2).

Figure 2

Proposed algorithm for MCL treatment. *Fever, bone pain, flushing, mast cell activation symptoms, cardiac failure, pleural effusion, ascites, cytopenia. **If available through a compassionate patient-named program. #Examples of drugs for clinical studies: cytarabine + 2-chlorodeoxyadenosine (2-CdA); CLAG (clofarabine, aracytine, granulocyte stimulating agents); FLAG (fludarabine, aracytine, granulocyte stimulating agents); tyrosine kinase inhibitors (midostaurin, masitinib, ponatinib, sorafenib), or Aurora kinase inhibitors in combination with other drugs; mammalian target of rapamicin (m-TOR) inhibitors; deoxy nucleotide methyl-transferase inhibitors (azacytidine, decitabine).

Figure 2

Proposed algorithm for MCL treatment. *Fever, bone pain, flushing, mast cell activation symptoms, cardiac failure, pleural effusion, ascites, cytopenia. **If available through a compassionate patient-named program. #Examples of drugs for clinical studies: cytarabine + 2-chlorodeoxyadenosine (2-CdA); CLAG (clofarabine, aracytine, granulocyte stimulating agents); FLAG (fludarabine, aracytine, granulocyte stimulating agents); tyrosine kinase inhibitors (midostaurin, masitinib, ponatinib, sorafenib), or Aurora kinase inhibitors in combination with other drugs; mammalian target of rapamicin (m-TOR) inhibitors; deoxy nucleotide methyl-transferase inhibitors (azacytidine, decitabine).

Close modal

MCL is a rare form of systemic mastocytosis with poor prognosis and very few therapeutic options. It can appear de novo or after previous mastocytosis. It shares more clinicobiologic aspects with ASM than with AML. Hepatosplenomegaly, anemia, and thrombocytopenia are usually present and aleukemic forms are frequent. The serum tryptase level is usually high. MCs are usually CD2 and/or CD25 positive. Wild-type or exon 9 to 13 KIT mutations are not rare and complete gene sequencing is needed. Therapy usually fails and the median survival time is < 6 months. New combination therapies including KIT inhibitors, chemotherapy, 2-CdA, and bone marrow transplantation should in the near future improve the prognosis of this still devastating disease.

The authors thank Alison Foote (Grenoble Clinical Research Center) for critically editing the manuscript with particular attention to language usage.

Contribution: S.G.-L., L.L., and G.D. reviewed and analyzed the literature data; S.G.-L., O.H., and G.D. conceived and designed the study; P.D. M.-O.C., O.H., and G.D. provided unpublished patients' data; and all authors wrote the manuscript and gave final approval of the manuscript.

Conflict-of-interest disclosure: O.H. and P.D. are co-founders of, consultants to, and stockholders in AB Science (Paris, France). The remaining authors declare no competing financial interests.

Correspondence: Gandhi Damaj, MD, Department of Hematology, CHU d'Amiens, Hôpital Sud, Avenue Laennec, 80054, Amiens, France; e-mail: damaj.gandhi@chu-amiens.fr; or Prof Olivier Hermine, Service d'Hématologie Adultes, CNRS UMR 8147 et Centre de Référence sur les Mastocytoses, Hôpital Necker-Enfants Malades, Université Paris Descartes, AP-HP, 149 Rue des Sèvres, 75743 Paris Cedex 15, France; e-mail: ohermine@gmail.com.

1
Swerdlow
 
SH
CE
 
Harris
 
NL
, et al. 
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
Lyon, France
International Agency for Research on Cancer
2
Valent
 
P
Akin
 
C
Escribano
 
L
, et al. 
Standards and standardization in mastocytosis: consensus statements on diagnostics, treatment recommendations and response criteria.
Eur J Clin Invest
2007
, vol. 
37
 
6
(pg. 
435
-
453
)
3
Valent
 
P
Akin
 
C
Sperr
 
WR
, et al. 
Diagnosis and treatment of systemic mastocytosis: state of the art.
Br J Haematol
2003
, vol. 
122
 
5
(pg. 
695
-
717
)
4
Tefferi
 
A
Pardanani
 
A
Clinical, genetic, and therapeutic insights into systemic mast cell disease.
Curr Opin Hematol
2004
, vol. 
11
 
1
(pg. 
58
-
64
)
5
Tefferi
 
A
Pardanani
 
A
Systemic mastocytosis: current concepts and treatment advances.
Curr Hematol Rep
2004
, vol. 
3
 
3
(pg. 
197
-
202
)
6
Lim
 
KH
Tefferi
 
A
Lasho
 
TL
, et al. 
Systemic mastocytosis in 342 consecutive adults: survival studies and prognostic factors.
Blood
2009
, vol. 
113
 
23
(pg. 
5727
-
5736
)
7
Valentini
 
CG
Rondoni
 
M
Pogliani
 
EM
, et al. 
Mast cell leukemia: a report of ten cases.
Ann Hematol
2008
, vol. 
87
 
6
(pg. 
505
-
508
)
8
Teodosio
 
C
Garcia-Montero
 
AC
Jara-Acevedo
 
M
, et al. 
An immature immunophenotype of bone marrow mast cells predicts for multilineage D816V KIT mutation in systemic mastocytosis.
Leukemia
2012
, vol. 
26
 
5
(pg. 
951
-
958
)
9
Noack
 
F
Sotlar
 
K
Notter
 
M
Thiel
 
E
Valent
 
P
Horny
 
HP
Aleukemic mast cell leukemia with abnormal immunophenotype and c-kit mutation D816V.
Leuk Lymphoma
2004
, vol. 
45
 
11
(pg. 
2295
-
2302
)
10
Joachim
 
G
Uber Mastzellenleukamien.
Deutsches Archives fur Klinische Medizin
1906
, vol. 
87
 pg. 
437
 
11
Baghestanian
 
M
Bankl
 
H
Sillaber
 
C
, et al. 
A case of malignant mastocytosis with circulating mast cell precursors: biologic and phenotypic characterization of the malignant clone.
Leukemia
1996
, vol. 
10
 
1
(pg. 
159
-
166
)
12
Coser
 
P
Quaglino
 
D
De Pasquale
 
A
Colombetti
 
V
Prinoth
 
O
Cytobiological and clinical aspects of tissue mast cell leukaemia.
Br J Haematol
1980
, vol. 
45
 
1
(pg. 
5
-
12
)
13
Dalton
 
R
Chan
 
L
Batten
 
E
Eridani
 
S
Mast cell leukaemia: evidence for bone marrow origin of the pathological clone.
Br J Haematol
1986
, vol. 
64
 
2
(pg. 
397
-
406
)
14
Daniel
 
MT
Flandrin
 
G
Bernard
 
J
[Acute mast-cell leukemia. Cytochemical and ultrastructural study, about a particular case (author's transl)].
Nouv Rev Fr Hematol
1975
, vol. 
15
 
3
(pg. 
319
-
332
)
15
Efrati
 
P
Klajman
 
A
Spitz
 
H
Mast cell leukemia? Malignant mastocytosis with leukemia-like manifestations.
Blood
1957
, vol. 
12
 
10
(pg. 
869
-
882
)
16
Friedman
 
BI
Will
 
JJ
Freiman
 
DG
Braunstein
 
H
Tissue mast cell leukemia.
Blood
1958
, vol. 
13
 
1
(pg. 
70
-
78
)
17
Kyriakou
 
D
Kouroumalis
 
E
Konsolas
 
J
, et al. 
Systemic mastocytosis: a rare cause of noncirrhotic portal hypertension simulating autoimmune cholangitis–report of four cases.
Am J Gastroenterol
1998
, vol. 
93
 
1
(pg. 
106
-
108
)
18
Le Cam
 
MT
Wolkenstein
 
P
Cosnes
 
A
, et al. 
[Acute mast cell leukemia disclosed by vasomotor flushing].
Ann Dermatol Venereol
1997
, vol. 
124
 
9
(pg. 
621
-
622
)
19
Szweda
 
JAAJ
Fine
 
G
Nixon
 
RK
Rupe
 
CE
Systemic mast cell disease: a review and report of three cases.
Am J Med
1962
, vol. 
32
 
2
(pg. 
227
-
239
)
20
Travis
 
WD
Li
 
CY
Hoagland
 
HC
Travis
 
LB
Banks
 
PM
Mast cell leukemia: report of a case and review of the literature.
Mayo Clin Proc
1986
, vol. 
61
 
12
(pg. 
957
-
966
)
21
Cherner
 
JA
Jensen
 
RT
Dubois
 
A
O'Dorisio
 
TM
Gardner
 
JD
Metcalfe
 
DD
Gastrointestinal dysfunction in systemic mastocytosis. A prospective study.
Gastroenterology
1988
, vol. 
95
 
3
(pg. 
657
-
667
)
22
Friedman
 
BS
Steinberg
 
SC
Meggs
 
WJ
Kaliner
 
MA
Frieri
 
M
Metcalfe
 
DD
Analysis of plasma histamine levels in patients with mast cell disorders.
Am J Med
1989
, vol. 
87
 
6
(pg. 
649
-
654
)
23
Horny
 
HP
Sotlar
 
K
Valent
 
P
Hartmann
 
K
Mastocytosis: a disease of the hematopoietic stem cell.
Dtsch Arztebl Int
2008
, vol. 
105
 
40
(pg. 
686
-
692
)
24
Arredondo
 
AR
Gotlib
 
J
Shier
 
L
, et al. 
Myelomastocytic leukemia versus mast cell leukemia versus systemic mastocytosis associated with acute myeloid leukemia: a diagnostic challenge.
Am J Hematol
2010
, vol. 
85
 
8
(pg. 
600
-
606
)
25
Horny
 
HP
Sotlar
 
K
Valent
 
P
Mastocytosis: state of the art.
Pathobiology
2007
, vol. 
74
 
2
(pg. 
121
-
132
)
26
Valent
 
P
Mast cell leukemia–a rare form of myeloid leukemia.
Wien Klin Wochenschr
2002
, vol. 
114
 
5-6
(pg. 
173
-
174
)
27
Teodosio
 
C
Garcia-Montero
 
AC
Jara-Acevedo
 
M
, et al. 
Mast cells from different molecular and prognostic subtypes of systemic mastocytosis display distinct immunophenotypes.
J Allergy Clin Immunol
2010
, vol. 
125
 
3
(pg. 
719
-
726
)
28
Garcia-Montero
 
AC
Jara-Acevedo
 
M
Teodosio
 
C
, et al. 
KIT mutation in mast cells and other bone marrow hematopoietic cell lineages in systemic mast cell disorders: a prospective study of the Spanish Network on Mastocytosis (REMA) in a series of 113 patients.
Blood
2006
, vol. 
108
 
7
(pg. 
2366
-
2372
)
29
Georgin-Lavialle
 
S
Lhermitte
 
L
Suarez
 
F
, et al. 
Mast cell leukemia: identification of a new c-Kit mutation, dup(501-502), and response to masitinib, a c-Kit tyrosine kinase inhibitor.
Eur J Haematol
2012
, vol. 
89
 
1
(pg. 
47
-
552
)
30
Mital
 
A
Piskorz
 
A
Lewandowski
 
K
Wasag
 
B
Limon
 
J
Hellmann
 
A
A case of mast cell leukaemia with exon 9 KIT mutation and good response to imatinib.
Eur J Haematol
2011
, vol. 
86
 
6
(pg. 
531
-
535
)
31
Akin
 
C
Fumo
 
G
Yavuz
 
AS
Lipsky
 
PE
Neckers
 
L
Metcalfe
 
DD
A novel form of mastocytosis associated with a transmembrane c-kit mutation and response to imatinib.
Blood
2004
, vol. 
103
 
8
(pg. 
3222
-
3225
)
32
Georgin-Lavialle
 
S
Aguilar
 
C
Guieze
 
R
, et al. 
Mast cell sarcoma, a rare and aggressive entity: report of 2 cases and review of the literature [published online ahead of print November 5, 2012].
J Clin Oncol
 
33
Spector
 
MS
Iossifov
 
I
Kritharis
 
A
, et al. 
Mast-cell leukemia exome sequencing reveals a mutation in the IgE mast-cell receptor beta chain and KIT V654A.
Leukemia
2012
, vol. 
26
 
6
(pg. 
1422
-
1425
)
34
Tefferi
 
A
Levine
 
RL
Lim
 
KH
, et al. 
Frequent TET2 mutations in systemic mastocytosis: clinical, KITD816V and FIP1L1-PDGFRA correlates.
Leukemia
2009
, vol. 
23
 
5
(pg. 
900
-
904
)
35
Soucie
 
E
HK
 
Hanssens
 
K
Mercher
 
T
, et al. 
In aggressive forms of mastocytosis, TET2 loss cooperates with c-KITD816V to transform mast cells.
Blood
2012
, vol. 
120
 
24
(pg. 
4846
-
4849
)
36
Chantorn
 
R
Shwayder
 
T
Death from mast cell leukemia: a young patient with longstanding cutaneous mastocytosis evolving into fatal mast cell leukemia.
Pediatr Dermatol
2012
, vol. 
29
 
5
(pg. 
605
-
609
)
37
Waters
 
WJ
Lacson
 
PS
Mast cell leukemia presenting as urticaria pigmentosa; report of a case.
Pediatrics
1957
, vol. 
19
 
6
(pg. 
1033
-
1042
)
38
Bodemer
 
C
Hermine
 
O
Palmerini
 
F
, et al. 
Pediatric mastocytosis is a clonal disease associated with D(816)V and other activating c-KIT mutations.
J Invest Dermatol
2010
, vol. 
130
 
3
(pg. 
804
-
815
)
39
Valent
 
P
Sperr
 
WR
Akin
 
C
How I treat patients with advanced systemic mastocytosis.
Blood
2010
, vol. 
116
 
26
(pg. 
5812
-
5817
)
40
Kluin-Nelemans
 
HC
Jansen
 
JH
Breukelman
 
H
, et al. 
Response to interferon alfa-2b in a patient with systemic mastocytosis.
N Engl J Med
1992
, vol. 
326
 
9
(pg. 
619
-
623
)
41
Butterfield
 
JH
Response of severe systemic mastocytosis to interferon alpha.
Br J Dermatol
1998
, vol. 
138
 
3
(pg. 
489
-
495
)
42
Worobec
 
AS
Kirshenbaum
 
AS
Schwartz
 
LB
Metcalfe
 
DD
Treatment of three patients with systemic mastocytosis with interferon alpha-2b.
Leuk Lymphoma
1996
, vol. 
22
 
5-6
(pg. 
501
-
508
)
43
Casassus
 
P
Caillat-Vigneron
 
N
Martin
 
A
, et al. 
Treatment of adult systemic mastocytosis with interferon-alpha: results of a multicentre phase II trial on 20 patients.
Br J Haematol
2002
, vol. 
119
 
4
(pg. 
1090
-
1097
)
44
Hauswirth
 
AW
Simonitsch-Klupp
 
I
Uffmann
 
M
, et al. 
Response to therapy with interferon alpha-2b and prednisolone in aggressive systemic mastocytosis: report of five cases and review of the literature.
Leuk Res
2004
, vol. 
28
 
3
(pg. 
249
-
257
)
45
Pardini
 
S
Bosincu
 
L
Bonfigli
 
S
Dore
 
F
Longinotti
 
M
Anaphylactic-like syndrome in systemic mastocytosis treated with alpha-2-interferon.
Acta Haematol
1991
, vol. 
85
 
4
pg. 
220
 
46
Penack
 
O
Sotlar
 
K
Noack
 
F
Horny
 
HP
Thiel
 
E
Notter
 
M
Cladribine therapy in a patient with an aleukemic subvariant of mast cell leukemia.
Ann Hematol
2005
, vol. 
84
 
10
(pg. 
692
-
693
)
47
Kirshenbaum
 
AS
Goff
 
JP
Semere
 
T
Foster
 
B
Scott
 
LM
Metcalfe
 
DD
Demonstration that human mast cells arise from a progenitor cell population that is CD34(+), c-kit(+), and expresses aminopeptidase N (CD13).
Blood
1999
, vol. 
94
 
7
(pg. 
2333
-
2342
)
48
Carrera
 
CJ
Terai
 
C
Lotz
 
M
, et al. 
Potent toxicity of 2-chlorodeoxyadenosine toward human monocytes in vitro and in vivo. A novel approach to immunosuppressive therapy.
J Clin Invest
1990
, vol. 
86
 
5
(pg. 
1480
-
1488
)
49
Pardanani
 
A
Hoffbrand
 
AV
Butterfield
 
JH
Tefferi
 
A
Treatment of systemic mast cell disease with 2-chlorodeoxyadenosine.
Leuk Res
2004
, vol. 
28
 
2
(pg. 
127
-
131
)
50
Kluin-Nelemans
 
HC
Oldhoff
 
JM
Van Doormaal
 
JJ
, et al. 
Cladribine therapy for systemic mastocytosis.
Blood
2003
, vol. 
102
 
13
(pg. 
4270
-
4276
)
51
Aichberger
 
KJ
Gleixner
 
KV
Mirkina
 
I
, et al. 
Identification of proapoptotic Bim as a tumor suppressor in neoplastic mast cells: role of KIT D816V and effects of various targeted drugs.
Blood
2009
, vol. 
114
 
26
(pg. 
5342
-
5351
)
52
Gabillot-Carre
 
M
Lepelletier
 
Y
Humbert
 
M
, et al. 
Rapamycin inhibits growth and survival of D816V-mutated c-kit mast cells.
Blood
2006
, vol. 
108
 
3
(pg. 
1065
-
1072
)
53
Gleixner
 
KV
Mayerhofer
 
M
Aichberger
 
KJ
, et al. 
PKC412 inhibits in vitro growth of neoplastic human mast cells expressing the D816V-mutated variant of KIT: comparison with AMN107, imatinib, and cladribine (2CdA) and evaluation of cooperative drug effects.
Blood
2006
, vol. 
107
 
2
(pg. 
752
-
759
)
54
Furitsu
 
T
Tsujimura
 
T
Tono
 
T
, et al. 
Identification of mutations in the coding sequence of the proto-oncogene c-kit in a human mast cell leukemia cell line causing ligand-independent activation of c-kit product.
J Clin Invest
1993
, vol. 
92
 
4
(pg. 
1736
-
1744
)
55
Longley
 
BJ
Metcalfe
 
DD
Tharp
 
M
, et al. 
Activating and dominant inactivating c-KIT catalytic domain mutations in distinct clinical forms of human mastocytosis.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 
4
(pg. 
1609
-
1614
)
56
Longley
 
BJ
Tyrrell
 
L
Lu
 
SZ
, et al. 
Somatic c-KIT activating mutation in urticaria pigmentosa and aggressive mastocytosis: establishment of clonality in a human mast cell neoplasm.
Nat Genet
1996
, vol. 
12
 
3
(pg. 
312
-
314
)
57
Ma
 
Y
Zeng
 
S
Metcalfe
 
DD
, et al. 
The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations.
Blood
2002
, vol. 
99
 
5
(pg. 
1741
-
1744
)
58
Nakagomi
 
N
Hirota
 
S
Juxtamembrane-type c-kit gene mutation found in aggressive systemic mastocytosis induces imatinib-resistant constitutive KIT activation.
Lab Invest
2007
, vol. 
87
 
4
(pg. 
365
-
371
)
59
Longley
 
BJ
Reguera
 
MJ
Ma
 
Y
Classes of c-KIT activating mutations: proposed mechanisms of action and implications for disease classification and therapy.
Leuk Res
2001
, vol. 
25
 
7
(pg. 
571
-
576
)
60
Zermati
 
Y
De Sepulveda
 
P
Feger
 
F
, et al. 
Effect of tyrosine kinase inhibitor STI571 on the kinase activity of wild-type and various mutated c-kit receptors found in mast cell neoplasms.
Oncogene
2003
, vol. 
22
 
5
(pg. 
660
-
664
)
61
Frost
 
MJ
Ferrao
 
PT
Hughes
 
TP
Ashman
 
LK
Juxtamembrane mutant V560GKit is more sensitive to Imatinib (STI571) compared with wild-type c-kit whereas the kinase domain mutant D816VKit is resistant.
Mol Cancer Ther
2002
, vol. 
1
 
12
(pg. 
1115
-
1124
)
62
Lahortiga
 
I
Akin
 
C
Cools
 
J
, et al. 
Activity of imatinib in systemic mastocytosis with chronic basophilic leukemia and a PRKG2-PDGFRB fusion.
Haematologica
2008
, vol. 
93
 
1
(pg. 
49
-
56
)
63
Droogendijk
 
HJ
Kluin-Nelemans
 
HJ
van Doormaal
 
JJ
Oranje
 
AP
van de Loosdrecht
 
AA
van Daele
 
PL
Imatinib mesylate in the treatment of systemic mastocytosis: a phase II trial.
Cancer
2006
, vol. 
107
 
2
(pg. 
345
-
351
)
64
Piccaluga
 
PP
Rondoni
 
M
Paolini
 
S
Rosti
 
G
Martinelli
 
G
Baccarani
 
M
Imatinib mesylate in the treatment of hematologic malignancies.
Expert Opin Biol Ther
2007
, vol. 
7
 
10
(pg. 
1597
-
1611
)
65
Pardanani
 
A
Elliott
 
M
Reeder
 
T
, et al. 
Imatinib for systemic mast-cell disease.
Lancet
2003
, vol. 
362
 
9383
(pg. 
535
-
536
)
66
Paul
 
C
Sans
 
B
Suarez
 
F
, et al. 
Masitinib for the treatment of systemic and cutaneous mastocytosis with handicap: a phase 2a study.
Am J Hematol
2010
, vol. 
85
 
12
(pg. 
921
-
925
)
67
Shah
 
NP
Lee
 
FY
Luo
 
R
Jiang
 
Y
Donker
 
M
Akin
 
C
Dasatinib (BMS-354825) inhibits KITD816V, an imatinib-resistant activating mutation that triggers neoplastic growth in most patients with systemic mastocytosis.
Blood
2006
, vol. 
108
 
1
(pg. 
286
-
291
)
68
Schittenhelm
 
MM
Shiraga
 
S
Schroeder
 
A
, et al. 
Dasatinib (BMS-354825), a dual SRC/ABL kinase inhibitor, inhibits the kinase activity of wild-type, juxtamembrane, and activation loop mutant KIT isoforms associated with human malignancies.
Cancer Res
2006
, vol. 
66
 
1
(pg. 
473
-
481
)
69
Verstovsek
 
S
Tefferi
 
A
Cortes
 
J
, et al. 
Phase II study of dasatinib in Philadelphia chromosome-negative acute and chronic myeloid diseases, including systemic mastocytosis.
Clin Cancer Res
2008
, vol. 
14
 
12
(pg. 
3906
-
3915
)
70
Purtill
 
D
Cooney
 
J
Sinniah
 
R
, et al. 
Dasatinib therapy for systemic mastocytosis: four cases.
Eur J Haematol
2008
, vol. 
80
 
5
(pg. 
456
-
458
)
71
Kneidinger
 
M
Schmidt
 
U
Rix
 
U
, et al. 
The effects of dasatinib on IgE receptor-dependent activation and histamine release in human basophils.
Blood
2008
, vol. 
111
 
6
(pg. 
3097
-
3107
)
72
Aichberger
 
KJ
Sperr
 
WR
Gleixner
 
KV
Kretschmer
 
A
Valent
 
P
Treatment responses to cladribine and dasatinib in rapidly progressing aggressive mastocytosis.
Eur J Clin Invest
2008
, vol. 
38
 
11
(pg. 
869
-
873
)
73
Gotlib
 
J
Berube
 
C
Growney
 
JD
, et al. 
Activity of the tyrosine kinase inhibitor PKC412 in a patient with mast cell leukemia with the D816V KIT mutation.
Blood
2005
, vol. 
106
 
8
(pg. 
2865
-
2870
)
74
Growney
 
JD
Clark
 
JJ
Adelsperger
 
J
, et al. 
Activation mutations of human c-KIT resistant to imatinib mesylate are sensitive to the tyrosine kinase inhibitor PKC412.
Blood
2005
, vol. 
106
 
2
(pg. 
721
-
724
)
75
Gotlib
 
J
Kluin-Nelemans
 
HC
George
 
TI
, et al. 
KIT inhibitor midostaurin in patients with advanced systemic mastocytosis: results of a planned interim analysis of the global CPKC412D2201 trial.
Blood
2012
, vol. 
120
 
21
 
Abstract 799
76
Samorapoompichit
 
P
Steiner
 
M
Lucas
 
T
, et al. 
Induction of apoptosis in the human mast cell leukemia cell line HMC-1 by various antineoplastic drugs.
Leuk Lymphoma
2003
, vol. 
44
 
3
(pg. 
509
-
515
)
77
Brcic
 
L
Vuletic
 
LB
Stepan
 
J
, et al. 
Mast-cell sarcoma of the tibia.
J Clin Pathol
2007
, vol. 
60
 
4
(pg. 
424
-
425
)
78
Yoshida
 
C
Takeuchi
 
M
Tsuchiyama
 
J
Sadahira
 
Y
Successful treatment of KIT D816V-positive, imatinib-resistant systemic mastocytosis with interferon-alpha.
Intern Med
2009
, vol. 
48
 
22
(pg. 
1973
-
1978
)
79
Lin
 
JT
Lachmann
 
E
Nagler
 
W
Low back pain and myalgias in acute and relapsed mast cell leukemia: a case report.
Arch Phys Med Rehabil
2002
, vol. 
83
 
6
(pg. 
860
-
863
)
80
Spyridonidis
 
A
Thomas
 
AK
Bertz
 
H
, et al. 
Evidence for a graft-versus-mast-cell effect after allogeneic bone marrow transplantation.
Bone Marrow Transplant
2004
, vol. 
34
 
6
(pg. 
515
-
519
)
81
Nakamura
 
R
Chakrabarti
 
S
Akin
 
C
, et al. 
A pilot study of nonmyeloablative allogeneic hematopoietic stem cell transplant for advanced systemic mastocytosis.
Bone Marrow Transplant
2006
, vol. 
37
 
4
(pg. 
353
-
358
)
82
Chen
 
TY
Chen
 
JS
Huang
 
WT
Su
 
WC
Tsao
 
CJ
Rapid engraftment of mast cells of donor origin in a case of acute myeloid leukemia with mast cell leukemia after allogeneic stem cell transplantation.
Bone Marrow Transplant
2003
, vol. 
32
 
1
(pg. 
111
-
114
)
83
Hissard
 
R
Moncourier
 
L
Jacquet
 
J
Examination of a new hematodermia; mastocytosis.
Presse Med
1951
, vol. 
59
 
82
(pg. 
1765
-
1767
)
84
Brinkmann
 
E
Mast cell reticulosis (tissular basophiloma) with histamine flushes and development into tissular basophilic leukemia.
Schweiz Med Wochenschr
1959
, vol. 
89
 (pg. 
1046
-
1048
)
85
Mutter
 
RD
Tannenbaum
 
M
Ultmann
 
JE
Systemic mast cell disease.
Ann Intern Med
1963
, vol. 
59
 (pg. 
887
-
906
)
86
Clancy
 
RL
Gauldie
 
J
Vallieres
 
M
Bienenstock
 
J
Day
 
RP
Pineo
 
GF
An approach to immunotherapy using antibody to IgE in mast cell leukemia.
Cancer
1976
, vol. 
37
 
2
(pg. 
693
-
696
)
87
Kimura
 
A
Tanaka
 
R
Taketomi
 
Y
, et al. 
[A case of mastocytoma with leukemic manifestation of its immature form in the terminal stage (author's transl)].
Rinsho Ketsueki
1979
, vol. 
20
 
9
(pg. 
1137
-
1146
)
88
Horny
 
HP
Parwaresch
 
MR
Kaiserling
 
E
, et al. 
Mast cell sarcoma of the larynx.
J Clin Pathol
1986
, vol. 
39
 
6
(pg. 
596
-
602
)
89
Escribano
 
L
Orfao
 
A
Villarrubia
 
J
, et al. 
Sequential immunophenotypic analysis of mast cells in a case of systemic mast cell disease evolving to a mast cell leukemia.
Cytometry
1997
, vol. 
30
 
2
(pg. 
98
-
102
)
90
Pauls
 
JD
Brems
 
J
Pockros
 
PJ
, et al. 
Mastocytosis: diverse presentations and outcomes.
Arch Intern Med
1999
, vol. 
159
 
4
(pg. 
401
-
405
)
91
Horny
 
HP
Krokowski
 
M
Feller
 
AC
Hintze
 
G
Sotlar
 
K
Valent
 
P
[Aleukemic mast cell leukemia (formerly: “malignant mastocytosis”): an extremely rare form of leukemia. A case report and simultaneously a contribution to revised classification of mastocytosis].
Wien Klin Wochenschr
2002
, vol. 
114
 
5-6
(pg. 
222
-
228
)
92
Akin
 
C
Metcalfe
 
DD
Systemic mastocytosis.
Annu Rev Med
2004
, vol. 
55
 (pg. 
419
-
432
)
93
Krauth
 
MT
Fodinger
 
M
Rebuzzi
 
L
Greul
 
R
Chott
 
A
Valent
 
P
Aggressive systemic mastocytosis with sarcoma-like growth in the skeleton, leukemic progression, and partial loss of mast cell differentiation antigens.
Haematologica
2007
, vol. 
92
 
12
(pg. 
e126
-
e129
)
94
Yoshida
 
M
Nishikawa
 
Y
Yamamoto
 
Y
, et al. 
Mast cell leukemia with rapidly progressing portal hypertension.
Pathol Int
2009
, vol. 
59
 
11
(pg. 
817
-
822
)
95
Joris
 
M
Georgin-Lavialle
 
S
Chandesris
 
MO
, et al. 
Mast cell leukaemia: c-KIT mutations are not always positive.
Case Report Hematol
2012
pg. 
517546
 
Sign in via your Institution