PKC412 Inhibits the ZNF198-FGFR1 Fusion Tyrosine Kinase and Is Efficacious in Treatment of t(8;13)(p11;q12) Associated Stem Cell Myeloproliferative Disease.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2549-2549
Author(s):  
Jing Chen ◽  
Daniel J. DeAngelo ◽  
Jeffery L. Kutok ◽  
Ifor R. Williams ◽  
Benjamin H. Lee ◽  
...  

Abstract Human stem cell leukemia-lymphoma syndrome usually presents as a myeloproliferative disease (MPD) that evolves to acute myeloid leukemia and/or lymphoma. The syndrome associated with t(8;13)(p11;q12) results in expression of the ZNF198-FGFR1 fusion tyrosine kinase. Current empirically-derived cytotoxic chemotherapy is inadequate treatment of this disease. We hypothesized that small molecule inhibitors of the ZNF198-FGFR1 fusion would have therapeutic efficacy. We characterized the transforming activity of ZNF198-FGFR1 in hematopoietic cells in vitro and in vivo. Expression of ZNF198-FGFR1 in primary murine hematopoietic cells caused a myeloproliferative syndrome in mice that recapitulated the human MPD phenotype. Transformation in these assays, and activation of the downstream effector molecules PLCγ, STAT5 and PI3K/AKT, required the proline-rich, but not the zinc-finger domains of ZNF198. A small molecule tyrosine kinase inhibitor, PKC412 (N-benzoyl-staurosporine) effectively inhibited ZNF198-FGFR1 tyrosine kinase activity and activation of downstream effector pathways, and inhibited proliferation of ZNF198-FGFR1 transformed Ba/F3 cells. Furthermore, treatment with PKC412 resulted in statistically significant prolongation of survival in the murine model of ZNF198-FGFR1 induced myeloproliferative disease. Based in part on these data, PKC412 was administered to a patient with t(8;13)(p11;q12) and was efficacious in treatment of progressive myeloproliferative disease with organomegaly. Therefore, PKC412 may be a useful therapy for treatment of human stem cell leukemia-lymphoma syndrome.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4142-4142
Author(s):  
Michal Cipok ◽  
Yona Farnoushi ◽  
Sigi Kay ◽  
Igor Grinberg ◽  
Arbel Reis ◽  
...  

Abstract Abstract 4142 There are few in-vivo models for studying human leukemia and its therapy. These include the high cost immune-deficient NOD/SCID mice and large mammalian fetuses, which both require weeks to assess treatment response. We described a rapid and low-cost alternative in-vivo system for human leukemia in the preimmune chick embryo (Taizi et al, Exp Hem 34;1698-708,2006). We recently demonstrated that the turkey embryo provides a more robust model for the preclinical assessment of human leukemia infiltration (Grinberg et al, Leukemia Res 33;1417-26 2009). Here we describe the application of this powerful and inexpensive model for rapid preclinical assessment of anti-cancer therapies and basic research of blood malignancies. BM engraftment was robust occurring in 95% of the embryos using cells lines and 40% using fresh samples. Leukemia cells homed to the BM where they were already detected at 20 hours after injection and reached highest levels on days E19-24, using FACS and RT-PCR. Serial engraftment in secondary recipients of all three human stem cell leukemia lines and one fresh sample was detected in embryos injected with BM harvested 8-10 days after the first inoculation, validating the engraftment of cancer initiating cells. Human stem cell leukemia lines K562 and LAMA, (both BCR/Abl+) and CHRF (c-Kit+), and myeloma cell lines ARH-77 and CAG and fresh patient samples were injected IV into turkeys, on embryonic day E 11, using 5×106 cells from lines or 107 fresh patient cells. Engraftment of human leukemia and myeloma cells (cell lines and fresh samples) was detected 8-14 days later (E19-24), in the BM and in several hematopoietic organs at a frequency of 0.5->20%, by real-time PCR, immunohistochemistry and flow cytometry. CAG and ARH-77 myeloma cells engraftment was also detected by the presence of human monoclonal free light chain (6-10 mg/L) in blood collected from vessels of the chorioallantoic membrane, one week after cell injection. The growth of leukemias treated with doxorubicin or the tyrosine-kinase inhibitor Imatinib and myeloma with Velcade, at levels that were not toxic to the developing embryonic BM, was dramatically inhibited in vivo when the drug was injected together with cells on E11 or 48-72 hours after injection of the cells and homing to the BM. Using flow cytometry analysis the frequency of CHRF cells (detected with anti-human CD33) was reduced from 8% to 0.01% and K562 and LAMA (detected with anti-human CD71) from 1%-3% engraftment to <0.17% following treatment with Imatinib. Q-PCR analysis supported these results showing an average 8 fold reduction of CHRF and a 2-5 fold reduction of K562 and LAMA cells in the Imatinib treated turkey embryos. The ARH-77 cells (detected with anti-human CD38 and CD138) were inhibited from 8.5% to 0.72% after Velcade treatment, with a 16.5 fold reduction determined by Q-PCR analysis compare to untreated embryos. These results prove the efficacy and demonstrate the utility of the turkey embryo as a new complementary in-vivo model for studying cancer initiating cells and the growth of human blood malignancies and their response to treatment. With further improvements, it may provide an affordable, rapid in vivo system for studying the growth blood malignancies and help reduce time and cost of drug development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1586-1586
Author(s):  
Diane Heiser ◽  
S. Haihua Chu ◽  
Li Li ◽  
Ian M Kaplan ◽  
Curt I. Civin ◽  
...  

Abstract Abstract 1586 Internal tandem duplication (ITD) mutations in the receptor tyrosine kinase FLT3 are present in approximately 30% of AML patients and portend poor patient survival. Though there are several small molecule tyrosine kinase inhibitors in clinical trials targeting this constitutively activated receptor, none have produced durable remissions as monotherapy. This, along with high rate of relapse of FLT3-ITD+ blasts, suggests that leukemia-sustaining stem cells harboring the mutant receptor may be escaping inhibitor-induced cytotoxicity. The presence of a constitutively active FLT3 in leukemic stem cells (LSCs) may play a role in the continued survival and proliferation of leukemic blasts and provides an attractive and tractable target for therapy against aberrant LSCs. In order to study the reservoir of stem cells bearing the FLT3-ITD mutation, our laboratory has developed a mouse knock-in model expressing the mutation under the control of its endogeneous promoter. In this model, the FLT3-ITD mice develop a myeloproliferative disorder (MPD) characterized by splenomegaly, anemia, and myeloid expansion. Our studies previously revealed that transplantation of unfractionated bone marrow or lineage depleted (LIN-) marrow from FLT3-ITD mice failed to engraft at high levels, suggesting a hematopoietic stem cell (HSC) defect. After attempting multiple immunophenotypic population transplants, only SLAM-defined HSCs (LIN-CD150+CD48-CD41-) resulted in engraftment levels equivalent to WT littermates. Furthermore, transplantation of SLAM-defined HSCs fully recapitulated the MPD phenotype, indicating that the MPD-initiating cell resides in the SLAM compartment. Interestingly, in this model, the SLAM compartment is depleted 2–10 fold as compared to WT mice despite the expansion of other stem/progenitor compartments (i.e. LIN-, KSLs). We hypothesized that the functional HSC defect observed in our earlier transplantation experiments might be due to the depletion of these long-term HSCs (LT-HSCs). To test this hypothesis, we treated WT and FLT3-ITD transplant recipients with sorafenib, a small molecule inhibitor that has previously been shown to have activity against mutant FLT3. Treating recipients of LIN- marrow had no effect on subsequent engraftment capacity, while treatment of FLT3-ITD mice in utero and during early development (before depletion occurred) was able to fully restore HSC numbers and function. In addition to ameliorating the observed stem cell defect, Sorafenib treatment during development also led to a complete disappearance of all signs of myeloproliferative disease. While primitive LT-HSCs are classically defined as FLT3- by immunophenotype, detectable levels of expression of FLT3 were observed by quantitative PCR (qPCR) in both WT and FLT3-ITD SLAM cells. In fact, FLT3-ITD SLAM cells displayed a 6-fold increase in FLT3 mRNA levels over WT controls. We hypothesize that this expression of FLT3-ITD in the SLAM compartment drives over-proliferation and entry into the cell cycle, leading to depletion of the normally quiescent pool of LT-HSCs. In vivo BrdU incorporation confirmed increased proliferation of FLT3-ITD SLAM cells and cell cycle analysis demonstrated a doubling in the number of SLAM cells in G2/M phase. qPCR revealed increased expression of cell cycle-related genes such as CCND1 and PIM1 within the FLT3-ITD SLAM compartment. We demonstrate for the first time, isolation of SLAM-defined MPD-initiating cells allowing transplantation of traditionally difficult-to-transplant MPD, which may be applicable to other such disease models. Here, we also show that the FLT3-ITD mutation disrupts normal hematopoiesis, leading to a depletion of primitive HSCs coupled to progenitor expansion. The resulting myeloproliferative disease can be completely ablated by treatment with the small molecule inhibitor, Sorafenib. The simultaneous amelioration of disease and restoration of LT-HSC numbers demonstrates an intimate link between stem cell function/homeostasis and disease. By successfully targeting the most primitive pool of HSCs, Sorafenib may provide an avenue for the treatment of FLT3-ITD+ leukemic stem cells in combination with additional therapeutics. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 15 (1) ◽  
pp. 83-91
Author(s):  
Chen Li ◽  
Biao Qian ◽  
Zhao Ni ◽  
Qinzhang Wang ◽  
Zixiong Wang ◽  
...  

AbstractThis study aims to construct recombinant lentiviral vectors containing the human stem cell leukemia (SCL) gene and investigate their in vitro transfection efficiency in Interstitial Cells of Cajal (ICC) of guinea pig bladders. In this study, the human SCL gene was successfully cloned, and the recombinant lentivirus GV287-SCL was successfully constructed. The titer of the recombinant lentivirus was 5 × 108 TU /mL. After transfecting the ICCs with the lentiviral vector at different MOIs, the optimal MOI was determined to be 10.0, and the optimal transfection time was determined to be 3 days. The amplification product of the lentivirus transfection group was consistent with the target fragment, indicating that the SCL gene had been successfully introduced into ICCs. In conclusion, the recombinant lentiviral vector GV287-SCL was successfully constructed and transfected into the in vitro cultured ICCs. The successful expression of SCL in ICCs may provide an experimental basis for the in vivo transfection of the SCL gene.


1989 ◽  
Vol 170 (1) ◽  
pp. 339-342 ◽  
Author(s):  
C G Begley ◽  
P D Aplan ◽  
M P Davey ◽  
J P de Villartay ◽  
D I Cohen ◽  
...  

It has been hypothesized that a rearrangement between the delta recombining element (delta Rec) and a pseudo J alpha gene serves to delete the TCR-delta locus before rearrangement of the TCR-alpha genes. We have now sequenced a direct, site-specific rearrangement between the delta Rec element and a pseudo J alpha gene in a human leukemic stem-cell line. Putative "N-sequence" addition was noted at the site of recombination, suggesting that this event occurred at a time when the enzyme(s) involved in N-region addition were active in this cell. This provides support for the view that deletion of the TCR-delta locus is required before rearrangement of the TCR-alpha chain genes.


Blood ◽  
2009 ◽  
Vol 114 (1) ◽  
pp. 60-63 ◽  
Author(s):  
Kimi Y. Kong ◽  
Elizabeth A. Williamson ◽  
Jason H. Rogers ◽  
Tam Tran ◽  
Robert Hromas ◽  
...  

Abstract In embryonic stem cells, Oct-4 concentration is critical in determining the development of endoderm, mesoderm, and trophectoderm. Although Oct-4 expression is essential for mesoderm development, it is unclear whether it has a role in the development of specific mesodermal tissues. In this study, we have examined the importance of Oct-4 in the generation of hematopoietic cells using an inducible Oct-4 ESC line. We demonstrate that Oct-4 has a role in supporting hematopoiesis after specifying brachyury-positive mesoderm. When we suppressed Oct-4 expression before or after mesoderm specification, no hematopoietic cells are detected. However, hematopoiesis can be rescued in the absence of Oct-4 after mesoderm specification if the essential hematopoietic transcription factor stem cell leukemia is expressed. Our results suggest that, for hematopoiesis to occur, Oct-4 is required for the initial specification of mesoderm and subsequently is required for the development of hematopoietic cells from uncommitted mesoderm.


Genomics ◽  
2019 ◽  
Vol 111 (6) ◽  
pp. 1566-1573 ◽  
Author(s):  
Jeane Silva ◽  
Chang-Sheng Chang ◽  
Tianxiang Hu ◽  
Haiyan Qin ◽  
Eiko Kitamura ◽  
...  

2019 ◽  
Vol 146 (8) ◽  
pp. 2243-2254
Author(s):  
Yating Chong ◽  
Yun Liu ◽  
Sumin Lu ◽  
Baohuan Cai ◽  
Haiyan Qin ◽  
...  

2012 ◽  
Vol 76 (11) ◽  
pp. 2118-2123 ◽  
Author(s):  
Ahmed Magzoub KHALID ◽  
Atsushi ASANO ◽  
Yoshinao Z. HOSAKA ◽  
Masanori OHTA ◽  
Kenji OHYAMA ◽  
...  

Blood ◽  
2004 ◽  
Vol 103 (9) ◽  
pp. 3249-3250
Author(s):  
Malcolm A. S. Moore

Sign in / Sign up

Export Citation Format

Share Document