Stromal cell derived factor-1 (SDF-1α) and its receptor, CXCR4 play a role in the trafficking of CD34+ cells. AMD3100, a selective CXCR4 antagonist, can mobilize hematopoietic progenitors from marrow to peripheral blood in healthy human volunteers and in patients with multiple myeloma and non-Hodgkin’s lymphoma (Flomenberg et al, Blood 102, 39a, 2003). Overexpression of CXCR4 on human CD34+ progenitors increases their proliferation and NOD/SCID repopulating capacity (Kahn et al. Blood 103:2942, 2004). Since CXCR4 has been found to regulate the migration and development of AML stem cells in NOD/SCID mice, we studied the effect of AMD3100 on AML cells from the standpoint of proliferation and in vitro transendothelial transmigration utilizing a transwell system. AMD3100 (from AnorMED, Inc.), at concentrations from 0.1 to 1.0 ng/ml did not affect the viability or porliferation of purified AML blasts (n=4). AMD3100 did not influence the adherence of AML blasts to endothelial monolayers. In the presence of 0.1 to 1 ng/ml AMD-3100, the transmigration of normal CD34+ cells stimulated by 100 ng/ml SDF-1α through a human umbilical vein endothelial cell (HUVEC) monolayer was completely inhibited. Likewise, the transmigration of AML blasts through HUVECs was not altered by AMD3100 exposure, but the SDF-1α mediated transmigration was inhibited by AMD3100 from 0.1 to 1 ng/ml. The same effect was noted with AML transmigration through marrow stromal layers. The increase in transmigration through endothelial cells stimulated with G-CSF was not inhibited by AMD3100 whereas the transmigration stimulated by interleukin-8 was inhibited. When AMD3100 was placed in the bottom of the migration chamber, no independent effects on AML transmigration were noted. Co-culture of AML blasts with stromal monolayers protected blasts from apoptosis. This protection was not altered by SDF-1α, AMD3100, nor by the combination. These in vitro results demonstrate that AMD3100 can influence the migratory capacity of AML cells but has no direct effects on their proliferation or survival. Further in vitro and in vivo studies will be required to elucidate the role that this unique chemokine antagonist has in the mobilization potential of AML blasts or progenitors or in the interactions of AML cells with their microenvironment. Such studies have implications for AML autografting and AML blast interactions with extramedullary endothelial cells.

Author notes

Corresponding author

Sign in via your Institution