In this issue of Blood, Treon and colleagues provide strong evidence that mutations in MYD88 and CXCR4 dictate clinical presentation and survival in Waldenström macroglobulinemia (WM).1 

Determination of the MYD88 and CXCR4 mutation status in WM has clinical implications.

Determination of the MYD88 and CXCR4 mutation status in WM has clinical implications.

Close modal

WM is a rare malignancy of immunoglobulin M–secreting B cells.2  Recent work identified recurrent somatically acquired activating mutations in MYD88 as well as in CXCR4 in WM patient samples.3,4  In more than 90% of WM patient samples, the MYD88 L265P mutation is detectable. This aberration, which is also found in other lymphoma subtypes such as the activated B cell–like subtype of diffuse large B-cell lymphoma (ABC DLBCL) or mucosa-associated lymphoid tissue lymphoma, leads to constitutive activation of the oncogenic nuclear factor-κB signaling pathway.5-7  CXCR4 is a chemokine receptor that promotes survival of WM cells.4  Different mutations affecting the C terminus of CXCR4 can be identified in roughly 30% of WM patients.4  Although these analyses provided important insights into the molecular pathogenesis of WM, it remained unclear if these genetic aberrations impact clinical presentation or survival of affected patients.

In this issue, Treon et al investigate the clinical implications of MYD88 and CXCR4 mutations in WM.1  Virtually all patients harboring mutations in CXCR4 also exhibited mutated MYD88. Patient samples harboring CXCR4 mutations were further distinguished based on the mutation pattern (nonsense vs frameshift mutations). Interestingly, the mutation status of CXCR4 and MYD88 was associated with significant differences in clinical presentation and outcome (see figure).1  Patients with mutated MYD88 and CXCR4 nonsense mutations exhibited bone marrow infiltration significantly more frequently, had higher serum immunoglobulin M levels, and presented with symptomatic disease, including hyperviscosity, syndrome more frequently. In contrast, patients with mutated MYD88 and CXCR4 frameshift or nonsense mutations presented less frequently with adenopathy. Finally, patients that were wild type for both MYD88 and CXCR4 were characterized by adverse survival compared with patients with mutated MYD88 alone or patients that harbored both mutations. Taken together, these results suggest that the MYD88 and CXCR4 mutation status determines clinical presentation and outcome of patients diagnosed with WM (see figure).1 

From a clinical standpoint, the results by Treon et al can potentially be highly relevant for WM treatment. A better understanding which oncogenic pathways are activated and used in WM is a prerequisite for the optimal utilization of novel therapeutic agents. To this end, the determination of the MYD88 and CXCR4 mutation status might help us identify novel subgroups of WM that differ with respect to clinical presentation and prognosis. Additionally, these subgroups might benefit differentially to novel therapeutic strategies. Bruton’s tyrosine kinase, which interacts with MYD88, is a promising target for the treatment of WM, and encouraging results were obtained by using the Bruton’s tyrosine kinase inhibitor ibrutinib in relapsed and refractory WM.8  Similarly, interleukin-1 receptor–associated kinase inhibitors that are being developed for clinical use hold promise for the treatment of WM because MYD88 coordinates the assembly of a signaling complex consisting of different members of the interleukin-1 receptor–associated kinase family.5,6  Finally, the use of CXCR4 inhibitors such as plerixafor might be promising in WM patients with mutated CXCR4. Importantly, large and well-designed clinical trials with accompanying scientific programs are required to investigate if specific subtypes of WM respond preferentially to novel compounds. This approach will pave the way to more specific and less toxic treatment regimens for patients with WM.

Conflict-of-interest disclosure: The author declares no competing financial interests.

1
Treon
 
SP
Cao
 
Y
Xu
 
L
Yang
 
G
Liu
 
X
Hunter
 
ZR
Somatic mutations in MYD88 and CXCR4 are determinants of clinical presentation and overall survival in Waldenström macroglobulinemia.
Blood
2014
, vol. 
123
 
18
(pg. 
2791
-
2796
)
2
Janz
 
S
Waldenström macroglobulinemia: clinical and immunological aspects, natural history, cell of origin, and emerging mouse models.
ISRN Hematol
2013
, vol. 
2013
 pg. 
815325
 
3
Treon
 
SP
Xu
 
L
Yang
 
G
, et al. 
MYD88 L265P somatic mutation in Waldenström’s macroglobulinemia.
N Engl J Med
2012
, vol. 
367
 
9
(pg. 
826
-
833
)
4
Hunter
 
Z
Xu
 
L
Yang
 
G
, et al. 
The genomic landscape of Waldenström’s macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis [published online ahead of print December 23, 2013].
Blood
2013
5
Yang
 
G
Zhou
 
Y
Liu
 
X
, et al. 
A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenström macroglobulinemia.
Blood
2013
, vol. 
122
 
7
(pg. 
1222
-
1232
)
6
Ngo
 
VN
Young
 
RM
Schmitz
 
R
, et al. 
Oncogenically active MYD88 mutations in human lymphoma.
Nature
2011
, vol. 
470
 
7332
(pg. 
115
-
119
)
7
Gachard
 
N
Parrens
 
M
Soubeyran
 
I
, et al. 
IGHV gene features and MYD88 L265P mutation separate the three marginal zone lymphoma entities and Waldenström macroglobulinemia/lymphoplasmacytic lymphomas.
Leukemia
2013
, vol. 
27
 
1
(pg. 
183
-
189
)
8
Treon
 
SP
Tripsas
 
CK
Yang
 
G
, et al. 
 
A prospective multicenter study of the Bruton’s tyrosine kinase inhibitor ibrutinib in patients with relapsed or refractory Waldenström’s macroglobulinemia [abstract]. Blood. 2013. Abstract 251
Sign in via your Institution