scholarly journals VpreB Surrogate Light Chain Expression in B-Lineage ALL: A Report from the Children's Oncology Group

Author(s):  
Stuart Sheldon Winter ◽  
Amanda McCaustland ◽  
Chunxu Qu ◽  
No'eau Simeona ◽  
Nyla A. Heerema ◽  
...  

Immunotherapies directed against B-cell surface markers have been a common developmental strategy to treat B-cell malignancies. The IgH surrogate light chain (SLC), comprised of the VpreB1 (CD179a) and Lamda5 (CD179b) subunits is expressed on pro- and pre-B cells where it governs preBCR-mediated autonomous survival signaling. We hypothesized that the pre-BCR might merit the development of targeted immunotherapies to decouple "autonomous" signaling in B-lineage acute lymphoblastic leukemia (B-ALL). We used the COG minimal residual disease (MRD) flow panel to assess pre-BCR expression in 36 primary patient samples accrued to COG standard and high-risk B-ALL studies through AALL03B1. We also assessed CD179a expression in 16 cases with Day 29 end-induction samples, pre-selected to have ≥1% MRD. All analyses were performed on a 6-color Becton-Dickinson flow cytometer in a CLIA/CAP-certified laboratory. Among 36 cases tested, thirty-two were at the pre-B and four were at the pro-B stages of developmental arrest. One or both mAbs showed that CD179a was present in ≥20% of the B-lymphoblast population. All cases expressed CD179a in the end-induction B-lymphoblast population. The CD179a component of the SLC is commonly expressed in B-ALL, regardless of genotype, stage of developmental arrest or NCI risk-status.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e19006-e19006
Author(s):  
Stuart S. Winter ◽  
Amanda McCaustland ◽  
No'eau Simeona ◽  
Andrew J. Carroll ◽  
Nyla A. Heerema ◽  
...  

e19006 Background: The surface expression of mature B-cell markers have led to the development of immunotherapies against B-lineage lymphoblastic leukemia/lymphoma (B-ALL/B-LLy). Relapsing clones that have altered surface antigen expression are common means of treatment failure with immunotherapies. The elimination of the pan-B cell repertoire by current B-cell immunotherapies contributes to immune-compromise. A promising target is the pre-BCR surrogate light chain, comprised of the VpreB1 (CD179a) and Lamda5 (CD179b) subunits. Surrogate light chain is expressed on pro- and pre-B cells where it governs preBCR-mediated autonomous survival during B-cell maturation. Gene expression analyses have shown that CD179a is expressed in a sub-set of 10 to 15% of B-ALL cases. Because immunotherapies targeted to restricted stages of B-cell development may overcome the limitations of pan B-cell ablation, we tested the hypothesis that CD179a is more commonly expressed on B-lymphoblasts than previously thought. Methods: Utilizing an annotated set of 36 standard (AALL0331) and high-risk (AALL0232) B-ALL cases accrued to Children’s Oncology Group AALL03B1, we adapted the COG minimal residual disease (MRD) flow panel to include two additional PE- and FITC-conjugated mAbs against CD179a (Biolegend and i2Pharma). We assessed CD179a expression in 16 cases for which we had Day 28 end-induction samples, pre-selected to have ≥1% MRD, as determined by the COG Reference laboratories. Cases with ≥20% CD179a surface expression were determined to be positive for statistical comparisons. All analyses were performed on a 6-color Becton-Dickinson flow cytometer in a CLIA/CAP certified laboratory. Results: Thirty-four cases were arrested at the CD10-positive pre-B stage, and two cases at the CD10-negative pro-B stage. One or both mAbs showed that CD179a was present in ≥20% of the B-lymphoblast population, ranging from 20.2% to 90.6% for all 36 diagnostic samples. All cases expressed CD179a in the end-induction B-lymphoblast population. Compared to gene-expression based predictions, we found a significant difference between expected versus observed flow-based CD179a positivity (two-sided Fisher’s exact test, P< 0.001). We found that CD179a expression was observed in cases having E2A-PBX3, KMT2A, BCR-ABL1 and other re-arrangements that typify mixed phenotype acute leukemias (MPALs). Conclusions: Our results show that CD179a is commonly expressed in B-ALL, regardless of stage, NCI risk features, or molecular aberrations. Because the productively assembled preBCR mediates autonomous survival signaling in pro- and pre-B cells, it may also contribute to the mechanistic basis of MRD in B-ALL. Immunotherapies directed against the CD179a component of the preBCR may spare the immune-compromise that occurs with pan B-cell ablation, and prevent the emergence of therapy-resistant disease in B-ALL/B-LLy.


Blood ◽  
2001 ◽  
Vol 97 (7) ◽  
pp. 2115-2120 ◽  
Author(s):  
Jiann-Shiuh Chen ◽  
Elaine Coustan-Smith ◽  
Toshio Suzuki ◽  
Geoffrey A. Neale ◽  
Keichiro Mihara ◽  
...  

Abstract To identify new markers of minimal residual disease (MRD) in B-lineage acute lymphoblastic leukemia (ALL), gene expression of leukemic cells obtained from 4 patients with newly diagnosed ALL was compared with that of normal CD19+CD10+ B-cell progenitors obtained from 2 healthy donors. By cDNA array analysis, 334 of 4132 genes studied were expressed 1.5- to 5.8-fold higher in leukemic cells relative to both normal samples; 238 of these genes were also overexpressed in the leukemic cell line RS4;11. Nine genes were selected among the 274 overexpressed in at least 2 leukemic samples, and expression of the encoded proteins was measured by flow cytometry. Two proteins (caldesmon and myeloid nuclear differentiation antigen) were only weakly expressed in leukemic cells despite strong hybridization signals in the array. By contrast, 7 proteins (CD58, creatine kinase B, ninjurin1, Ref1, calpastatin, HDJ-2, and annexin VI) were expressed in B-lineage ALL cells at higher levels than in normal CD19+CD10+ B-cell progenitors (P &lt; .05 in all comparisons). CD58 was chosen for further analysis because of its abundant and prevalent overexpression. An anti-CD58 antibody identified residual leukemic cells (0.01% to 1.13%; median, 0.03%) in 9 of 104 bone marrow samples from children with ALL in clinical remission. MRD estimates by CD58 staining correlated well with those of polymerase chain reaction amplification of immunoglobulin genes. These results indicate that studies of gene expression with cDNA arrays can aid the discovery of leukemia markers.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 570-570
Author(s):  
Kadriye Nehir Cosgun ◽  
Rudi W Hendriks ◽  
Ross A Dickins ◽  
Nora Heisterkamp ◽  
Markus Muschen

Abstract Background: IGLL1 and VPREB1 encode for the l5 and VpreB components, respectively, of the surrogate light chain (SLC) of the pre-B cell receptor (pre-BCR). During early B-cell development, immunoglobulin (Ig) heavy chains pairs with SLCs to form the pre-BCR, a central signaling unit that drives proliferation and survival. Accordingly, germline mutations of IGLL1 (Minegishi J Exp Med 1998) and VPREB1 (Conley Immunol Rev 2005) are associated with profound B-cell defects and agammaglobulinemia in humans. However, somatic deletions of VPREB1 gene are frequent lesions in B-ALL and occur in >10% of B-ALL cases (Mangum et al., Leukemia 2014; Geng et al., Cancer Cell 2015), the significance of which is not clear. Since VPREB1 deletions are typically present at the time of diagnosis and are rarely acquired as secondary lesions at the time of relapse (Kuster Blood 2011), we hypothesized that loss of VPREB1 represents an early and essential event in leukemogenesis. Experimental Approach and Results: For genetic gain and loss of function analysis of VPREB1 and IGLL1, we established leukemia models based on pre-B cells from Igll1-/- mice (Kitamura Cell 1992), and Vpreb1-Igll1 double-transgenic mice (Van Loo Immunity 2007). Loss of Igll1 completely abrogated SLC expression on the surface of pre-B cells. In contrast, VpreB1-Igll1 double-transgenic pre-B cells expressed constitutively higher surface levels of SLC as part of their pre-BCR as evidenced by flow cytometry. Compared to wildtype controls, Igll1-/- pre-B cells lacking the ability to express a functional SLCs were more readily transformed by BCR-ABL1 oncogene. However, pre-B cells of VpreB1-Igll1 transgenic mice, were not permissive to BCR-ABL1 mediated transformation. In agreement with these results, VpreB1-Igll1 double-transgenic pre-B cells were resistant transformation by BCR-ABL1 in vivo. BCR-ABL1-transgenic mice with enforced expression Vpreb1-Igll1 remained disease-free for more than nine months, whereas the vast majority of BCR-ABL1-transgenic mice downregulated pre-BCR surface expression and developed lethal B-ALL within 90 days of birth (n=34, P<0.0001). Compared to wildtype pre-B cells, double-transgenic expression of VpreB-Igll1 interfered with oncogenic BCR-ABL1 tyrosine kinase signaling and suppressed phosphorylation of Btk, Syk and Src kinases resulting in cell cycle arrest and reduced colony formation ability. To test whether SLC tumor suppressive function depends on pre-BCR activity, we studied BCR-ABL1-mediated transformation of VpreB1-Igll1 double-transgenic pre-B cells on a Rag1-deficient background. Rag1-/- pro-B cells lack the ability to rearrange Ig V, D and J-gene segments and cannot express a functional Ig mHC, the central structural element of the pre-BCR. Surprisingly, Rag1-/-VpreB1-Igll1 double-transgenic pro-B cells were also resistant to BCR-ABL1-mediated transformation. These findings provide genetic evidence that Vpreb1 and Igll1 exert tumor suppressive effect on B cells regardless of functional pre-BCR expression. Conclusion: The VpreB and Igll1 surrogate light chain components of the pre-BCR act as a tumor suppressors in pre-B ALL cells. Interestingly, the tumor suppressor function of both VpreB and Igll1 is independent from their SLC-role in pre-BCR signaling: Vpreb1- and Iggl1-mediated tumor suppression was effective, regardless of pre-BCR function. These findings provide genetic evidence that VpreB and Igll1 have pre-BCR- and SLC-independent functions that prevent malignant transformation and limit proliferation of normal pre-B cells. Our findings support the hypothesis that VPREB1 deletion represents an early event during clonal evolution towards pre-B ALL, facilitating subsequent steps of leukemic transformation. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 16 (4) ◽  
pp. 18-24 ◽  
Author(s):  
O. A. Beznos ◽  
L. Yu. Grivtsova ◽  
A. V. Popa ◽  
M. A. Shervashidze ◽  
I. N. Serebryakova ◽  
...  

Background. Flow cytometry (FC) algorithms of detection of minimal residual disease (MRD) are well standardized, and approximate to molecular biologic methods. However, besides informative leukemia-associated aberrant immunophenotype, which are selected taking into account a tumor phenotype at diagnostics stage, it is necessary to consider specificity of the provided taget therapy and its influence on a cell. Objective: to offer stable combinations of antigens to identify B-cell precursors in patients on therapy of blinatumomab. Materials and methods. Clinical observation of patient G. 4 years old with B-cell precursors acute lymphoblastic leukemia (ALL) (pre-pre-B immunosubtype), whom after 3 bloks of reinduction therapy, taking into account MRD-positive status, blinatumomab was appointed as a monotherapy. Tumor immunophenotype was characterized in details by FC protocol according to EuroFlow in debute and relapse of the disease. MRD monitoring was provided by 8-color FC taking into account personalized leukemia-associated aberrant immunophenotypes. Results. In patient with B-cell precursors ALL received blinatumomab, the strategy of MRD monitoring was changed. Due to the lack of CD19 expression, identification of B-cell precursors was based on expression of cyCD22 in combination with nuclear TdT and CD10. Conclusion. In case of blinatumomab’s appointment during B-cell precursors ALL therapy, it is necessary to change the strategy of B-cell precursors identification, due to the lack of CD19 expression. Detection of B-cell precursors should be provided by assessment of other pan-B lineage antigens. First of all, it is cyCD22 or cyCD79a in combination with nuclear TdT and CD10, within the limits of nucleated cells of the sample.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Stephanie L. Rellick ◽  
Gangqing Hu ◽  
Debra Piktel ◽  
Karen H. Martin ◽  
Werner J. Geldenhuys ◽  
...  

AbstractB-cell acute lymphoblastic leukemia (ALL) is characterized by accumulation of immature hematopoietic cells in the bone marrow, a well-established sanctuary site for leukemic cell survival during treatment. While standard of care treatment results in remission in most patients, a small population of patients will relapse, due to the presence of minimal residual disease (MRD) consisting of dormant, chemotherapy-resistant tumor cells. To interrogate this clinically relevant population of treatment refractory cells, we developed an in vitro cell model in which human ALL cells are grown in co-culture with human derived bone marrow stromal cells or osteoblasts. Within this co-culture, tumor cells are found in suspension, lightly attached to the top of the adherent cells, or buried under the adherent cells in a population that is phase dim (PD) by light microscopy. PD cells are dormant and chemotherapy-resistant, consistent with the population of cells that underlies MRD. In the current study, we characterized the transcriptional signature of PD cells by RNA-Seq, and these data were compared to a published expression data set derived from human MRD B-cell ALL patients. Our comparative analyses revealed that the PD cell population is markedly similar to the MRD expression patterns from the primary cells isolated from patients. We further identified genes and key signaling pathways that are common between the PD tumor cells from co-culture and patient derived MRD cells as potential therapeutic targets for future studies.


Blood ◽  
2020 ◽  
Vol 136 (2) ◽  
pp. 210-223 ◽  
Author(s):  
Eun Ji Gang ◽  
Hye Na Kim ◽  
Yao-Te Hsieh ◽  
Yongsheng Ruan ◽  
Heather A. Ogana ◽  
...  

Abstract Resistance to multimodal chemotherapy continues to limit the prognosis of acute lymphoblastic leukemia (ALL). This occurs in part through a process called adhesion-mediated drug resistance, which depends on ALL cell adhesion to the stroma through adhesion molecules, including integrins. Integrin α6 has been implicated in minimal residual disease in ALL and in the migration of ALL cells to the central nervous system. However, it has not been evaluated in the context of chemotherapeutic resistance. Here, we show that the anti-human α6-blocking Ab P5G10 induces apoptosis in primary ALL cells in vitro and sensitizes primary ALL cells to chemotherapy or tyrosine kinase inhibition in vitro and in vivo. We further analyzed the underlying mechanism of α6-associated apoptosis using a conditional knockout model of α6 in murine BCR-ABL1+ B-cell ALL cells and showed that α6-deficient ALL cells underwent apoptosis. In vivo deletion of α6 in combination with tyrosine kinase inhibitor (TKI) treatment was more effective in eradicating ALL than treatment with a TKI (nilotinib) alone. Proteomic analysis revealed that α6 deletion in murine ALL was associated with changes in Src signaling, including the upregulation of phosphorylated Lyn (pTyr507) and Fyn (pTyr530). Thus, our data support α6 as a novel therapeutic target for ALL.


Sign in / Sign up

Export Citation Format

Share Document