In this issue of Blood, Kim et al1  investigated the preclinical therapeutic potential of targeting precursor B-cell receptor (pre-BCR) signaling and its mechanism of action in acute lymphoblastic leukemia (ALL). Ibrutinib, a US Food and Drug Administration (FDA)–approved inhibitor of Bruton tyrosine kinase (BTK), was demonstrated to interfere with pre-BCR signaling and specifically suppress in vitro and in vivo cell proliferation of B-ALL cells that express a functional pre-BCR. The synergistic activity with conventional chemotherapeutic agents corroborates ibrutinib as a new therapeutic opportunity for pre-BCR+ ALL.

Simplified schema of pre-BCR signaling and its inhibition by ibrutinib. (A) Pre-BCR assembles with Igα and Igβ to signal through spleen tyrosine kinase (SYK), BTK, phospholipase C-gamma2, SRC-homology-2-domain-containing leukocyte protein of 65 kDa (SLP65), and with SRC-family protein tyrosine kinases, such as LYN, to activate downstream pathways (AKT/ERK/mechanistic target of rapamycin [mTOR]), which promote cell proliferation. Ibrutinib interferes with pre-BCR signaling by inhibiting multiple targets, including BTK and B-lymphocyte kinase (BLK); impairs phosphorylation of the downstream effectors AKT/ERK/mTOR; and reduces BCL6 expression. (B) Schematic representation of bone marrow niche. Kim et al showed that ibrutinib inhibits chemotaxis of pre-B-ALL cells toward CXCL12. P, phosphorylation sites; PI3Kδ, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit δ. Professional illustration by Patrick Lane, ScEYEnce Studios.

Simplified schema of pre-BCR signaling and its inhibition by ibrutinib. (A) Pre-BCR assembles with Igα and Igβ to signal through spleen tyrosine kinase (SYK), BTK, phospholipase C-gamma2, SRC-homology-2-domain-containing leukocyte protein of 65 kDa (SLP65), and with SRC-family protein tyrosine kinases, such as LYN, to activate downstream pathways (AKT/ERK/mechanistic target of rapamycin [mTOR]), which promote cell proliferation. Ibrutinib interferes with pre-BCR signaling by inhibiting multiple targets, including BTK and B-lymphocyte kinase (BLK); impairs phosphorylation of the downstream effectors AKT/ERK/mTOR; and reduces BCL6 expression. (B) Schematic representation of bone marrow niche. Kim et al showed that ibrutinib inhibits chemotaxis of pre-B-ALL cells toward CXCL12. P, phosphorylation sites; PI3Kδ, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit δ. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

Pre-B-cell ALL is a hematological malignancy that arises from an oncogenic transformation of an early B-lymphocyte progenitor. It comprises multiple subtypes with distinct constellations of somatic genetic alterations. Despite cure rates exceeding 80% in children, disease-specific and treatment-related toxicity remain important causes of morbidity and mortality in children and adults.2  Therefore, development of targeted therapies that focus on specific oncogenic mechanisms of ALL cells are of great interest to improve outcomes. In this issue of Blood, the investigators explored the activity and mechanisms of action of the BTK inhibitor ibrutinib in preclinical models of pre-B-ALL.

The basic premises for this study were that: (1) targeting BCR signaling has been proved a successful therapeutic strategy in mature B-cell malignancies3-5 ; (2) signaling of the mature BCR is largely similar to that of pre-BCR, which plays a critical role during early B-cell development, promoting survival and expansion of progenitor cells with productively rearranged pre-BCRs6 ; and (3) a distinct subset of pre-B-ALL (∼15%) is selected for expression and activity of a functional pre-BCR (pre-BCR+).7  This group includes TCF3-PBX1+ ALL that is selectively sensitive to BCR signaling inhibitors.8  In the remaining ALL cases (eg, BCR-ABL1+ ALL),9  the dominant leukemic clones lack expression of a functional pre-BCR because of nonproductive immunoglobulin heavy chain (IGH) gene rearrangement and/or deregulation of other pathway components.

During early B-cell development, functional pre-BCRs are expressed for a short period after productive IGH rearrangement, allowing progenitor cells to transition into the pool of mature peripheral B cells. Survival signals from the pre-BCRs are transduced by the immunoglobulin-α (Igα) and Igβ signaling chains and include activation of downstream kinases (eg, LYN, SYK, BTK, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit δ)10  and transcriptional changes (eg, expression of B-cell CLL/lymphoma 6 [BCL6]) (see figure). Given the critical role of BTK for B-cell development and lymphomagenesis, BTK inhibitors have been extensively investigated. To date, ibrutinib is the only BTK inhibitor that is FDA-approved for treatment of mature B-cell lymphoproliferative malignancies, specifically mantle cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma, and Waldenstrom macroglobulinemia.

To explore the functional role of BTK in ALL, Kim et al began their study by screening BTK expression and activity (autophosphorylation at residue Y223) in a panel of B-ALL cell lines. The results indicated activated BTK in BCR-ABL1+ and pre-BCR+ ALL cells; however, only pre-BCR+ cells were sensitive to ibrutinib at clinically relevant concentrations. Ibrutinib acted through the inhibition of cell proliferation rather than through the induction of apoptosis. The exclusive sensitivity of pre-BCR+ ALL led the investigators to further explore the effects of ibrutinib on the activity of the pre-BCR signaling cascade. These studies revealed that ibrutinib abrogates induced and autonomous pre-BCR signaling, resulting in reduced calcium release and suppression of AKT and its downstream targets, including FOXO transcription factors, ERK, and mTOR signaling pathways, in pre-BCR+ ALL cells.

Kim et al, using gene expression profiling and western blot, observed that prolonged treatment with ibrutinib induces a feedback compensatory mechanism characterized by suppression of negative regulators (CD22, CD72, and PTPN6) and upregulation of downstream targets of BTK such as protein kinase C β. Similar results were previously reported for pre-BCR+/TCF3-PBX1+ ALL cells treated with the dual SRC/ABL kinase inhibitor dasatinib. A key defining feature of pre-BCR+ B-ALL is the constitutive expression of the transcription factor BCL6, which is induced following induction of pre-BCR signaling to sustain cell survival7 ; therefore, it is reasonable to expect reduced BCL6 expression upon treatment with ibrutinib, which was clearly demonstrated by the authors.

Because signals transduced by BTK regulate tissue homing, Kim et al explored the potential of ibrutinib to disrupt the interactions of leukemia cells with their microenvironment. Similar to its effects in mature B-cell malignancies, ibrutinib compromised the interactions of leukemic cells with their supportive microenvironments, as shown by the inhibited chemotaxis of ALL cells toward the chemokine C-X-C motif chemokine ligand 12 (CXCL12), independent of pre-BCR status (see figure).

Having characterized the mechanisms of action of ibrutinib in leukemic cells, Kim et al further investigated the importance of alternative targets for the activity of ibrutinib in pre-BCR+ ALL cells. Using CRISPR-Cas9–mediated genome editing, the authors nicely showed that both BTK and B-lymphocyte kinase are relevant targets of ibrutinib in pre-BCR+ ALL cells.

As a compelling follow-up to the FDA approval of ibrutinib for mature B-cell malignancies, these discoveries suggest that inhibition of pre-BCR signaling with BTK inhibitors may represent an effective therapeutic option for pre-BCR+ ALL patients. Kim et al provided another important step toward clinical translation by demonstrating that single-agent ibrutinib significantly prolonged survival in mouse xenografts models of pre-BCR+ ALL. Moreover, its combinations with conventional ALL chemotherapeutic agents, dexamethasone or vincristine, synergistically reduced viability and proliferation of pre-BCR+ ALL cells.

Overall, the work by Kim et al exploring new insights in the activity and mechanisms of actions of ibrutinib represents a significant therapeutic advance in ALL and provides a strong rationale for the clinical development in the ALL subset with functional pre-BCR expression. Establishment of diagnostic approaches to accurately detect pre-BCR+ B-ALL will help identify patients that may benefit from inhibition of pre-BCR signaling.

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

1.
Kim
E
,
Hurtz
C
,
Koehrer
S
, et al
.
Ibrutinib inhibits pre-BCR+ B-cell acute lymphoblastic leukemia progression by targeting BTK and BLK
.
Blood
.
2017
;
129
(
9
):
1155
-
1165
.
2.
Hunger
SP
,
Mullighan
CG
.
Acute lymphoblastic leukemia in children
.
N Engl J Med
.
2015
;
373
(
16
):
1541
-
1552
.
3.
Byrd
JC
,
Brown
JR
,
O’Brien
S
, et al
;
RESONATE Investigators
.
Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia
.
N Engl J Med
.
2014
;
371
(
3
):
213
-
223
.
4.
Wang
ML
,
Rule
S
,
Martin
P
, et al
.
Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma
.
N Engl J Med
.
2013
;
369
(
6
):
507
-
516
.
5.
Smith
MR
.
Ibrutinib in B lymphoid malignancies
.
Expert Opin Pharmacother
.
2015
;
16
(
12
):
1879
-
1887
.
6.
Müschen
M
.
Rationale for targeting the pre-B-cell receptor signaling pathway in acute lymphoblastic leukemia
.
Blood
.
2015
;
125
(
24
):
3688
-
3693
.
7.
Geng
H
,
Hurtz
C
,
Lenz
KB
, et al
.
Self-enforcing feedback activation between BCL6 and pre-B cell receptor signaling defines a distinct subtype of acute lymphoblastic leukemia
.
Cancer Cell
.
2015
;
27
(
3
):
409
-
425
.
8.
van der Veer
A
,
van der Velden
VH
,
Willemse
ME
, et al
.
Interference with pre-B-cell receptor signaling offers a therapeutic option for TCF3-rearranged childhood acute lymphoblastic leukemia
.
Blood Cancer J
.
2014
;
4
:
e181
.
9.
Trageser
D
,
Iacobucci
I
,
Nahar
R
, et al
.
Pre-B cell receptor-mediated cell cycle arrest in Philadelphia chromosome-positive acute lymphoblastic leukemia requires IKAROS function
.
J Exp Med
.
2009
;
206
(
8
):
1739
-
1753
.
10.
Saijo
K
,
Schmedt
C
,
Su
IH
, et al
.
Essential role of Src-family protein tyrosine kinases in NF-kappaB activation during B cell development
.
Nat Immunol
.
2003
;
4
(
3
):
274
-
279
.
Sign in via your Institution