Waldenström Macroglobulinemia (WM) is a disorder of lymphoplasmacytoid cells that inhabit lymph nodes and the bone marrow. WM cells are characterized by secretion of monoclonal pentameric IgM. These cells are CD19+, CD20+, CD22+, CD38+, CD138+/- and phenotypically resemble IgM plasmablasts or plasma cells. In addition, 91% of WM cases carry an activating mutation of MyD88 (L265P). Mature resting B cells can be driven to differentiate to IgM secreting plasmablasts and plasma cells with similar phenotypes using the TLR4 ligand lipopolysaccharide (LPS). We have demonstrated that LPS (+ cytokine)-differentiated cells become Bcl-xL dependent during this process, rendering them sensitive to the Bcl-xL/Bcl-2 inhibitor ABT-737. For this reason, we hypothesized that activation of MyD88 in WM cells could drive Bcl-xL dependence in a similar manner conferring ABT-737 sensitivity. We treated three WM cell lines, BCWM.1, MWCL-1 and RPCI-WM1 which all harbor the MyD88 (L265P) mutation with ABT-737. We found varying levels of resistance to ABT-737 with an IC50 > 2 μM for all three lines as compared with the ABT-737 sensitive multiple myeloma cell line MM.1s which has an IC50 of 0.4 μM. The RPCI-WM1 cell line was the most insensitive to ABT-737-induced apoptosis with no apoptosis above baseline up to 1.6 μM of drug.

Since the WM cell lines were not sensitive to direct inhibition of intrinsic survival regulators, we then examined the sensitivity of these cell lines to other activators of the intrinsic apoptosis pathway. Two of the three cell lines were moderately sensitive to bortezomib with IC50 ≈ 5 nM as compared with the sensitive multiple myeloma cell line MM.1s with an IC50 of 2 nM. The RPCI-WM1 cell line was insensitive to bortezomib with no apoptosis above baseline up to 20 nM bortezomib. Similarly, we found that two of the cell lines were moderately sensitive to arsenic trioxide with an IC50 ≈ 6 μM as compared with the multiple myeloma cell line MM1.s (IC50 ≈ 4 μM). The RPCI-WM1 cell line was insensitive to ATO as well with an IC50 > 20 μM.

Given the lack of sensitivity of the three WM cell lines we tested to Bcl-xL/Bcl-2 inhibition with ABT-737 treatment, and that RPCI-WM1 appears insensitive to multiple inducers of intrinsic apoptosis, we examined the expression levels of Bcl-2 family members in these cells. Both BCWM.1 cells and MWCL-1 cells expressed Bim mRNA at very low levels with MWCL-1 expressing no detectable Bim at the protein level. Surprisingly, more moderate levels of Bim were detected in RPCI-WM1 cells. These findings were confirmed at the mRNA level by qRT-PCR. Bcl-xL and Mcl-1 were detectable in all three lines at moderate levels while Bcl-2 which was only expressed at significant levels in MWCL-1 cells and undetectable in BCWM.1 cells. We examined the expression levels of the Bax and Bak in these cells and remarkably there was no detectable Bax and very small amounts of Bak protein in RPCI-WM1 cells. Consistent with a defect in gene expression, Bax mRNA was also low in RPCI-WM1. This was not due to copy number variation, as determined by array-CGH in both the initial patient isolate and the established cell line. Additionally, no loss of Bax, Bak or Bim (Bcl2l11) was observed in SNP array analysis of 46 patients with WM. Interestingly, Bak mRNA levels in RPCI-WM1 were similar to the other WM lines, suggesting a defect in translation or post-translational regulation is responsible for the low protein expression.

These results lead us to conclude that these WM cell lines are not sensitive to Bcl-xL/ Bcl-2 inhibition despite activation of MyD88. We have further shown that there are multiple and distinct differences in Bcl-2 family protein expression that lead to this insensitivity. While low levels of Bim combined with expression of Mcl-1 confer resistance to ABT-737 in MWCL-1 and BCWM.1, the lack of Bax and Bak confers resistance to intrinsic apoptotic stimuli in general in RPCI-WM1. Moreover, the loss of Bax and Bak protein expression occur through distinct mechanisms. These WM cell lines demonstrate that sensitivity to agents that kill through the intrinsic apoptotic pathway may vary within a disease that is characterized by a single activating mutation and suggests that additional heterogeneous events regulate the expression of Bcl-2 family proteins in WM.

Disclosures:

Leleu:CELGENE: Honoraria; JANSSEN: Honoraria. Lonial:Millennium: Consultancy; Celgene: Consultancy; Novartis: Consultancy; BMS: Consultancy; Sanofi: Consultancy; Onyx: Consultancy. Boise:Onyx Pharmaceuticals: Consultancy.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution