scholarly journals Combined inhibition of Bruton’s Tyrosine Kinase (BTK) and Phosphoinositide 3-kinase (PI3K) p110δ improves monocytosis and splenomegaly in a JMML Mouse Model

2018 ◽  
Vol 1 (1) ◽  
Author(s):  
Roshni Patel ◽  
Baskar Ramdas ◽  
Lisa Deng ◽  
Victoria Jideonwo-Auman ◽  
Reuben Kapur

Background and Hypothesis: Juvenile myelomonocytic leukemia (JMML) is an aggressive, childhood leukemic disorder for which there are no efficacious chemotherapeutics. Gain-of-function (GOF) mutations in the SHP2 phosphatase oncogene, Ptpn11, are the most common associated mutations. In hematopoietic cells, these mutations lead to increased AKT and ERK activation, which results in hyperproliferation of myeloid cells. Clinically, this manifests as monocytic leukocytosis and marked splenomegaly with the consequence of severe thrombocytopenia. Previously, this lab has shown that pharmacological inhibition of p110d, the hematopoietic-specific catalytic subunit of PI3K, moderates monocytosis and splenomegaly in a JMML mouse model with a SHP2 GOF mutation (E76K mice). Additionally, BTK has been identified as a potential therapeutic target as it cooperates with PI3K to increase activation of AKT and ERK in myeloid cells. Using a dual-hit approach of targeting both PI3K p110d and BTK may serve as a valuable treatment design for JMML.   Experimental Design or Project Methods: We have treated E76K mice with a combination of 20mg/kg ACP319, a PI3K p110d-specific inhibitor, and 20mg/kg acalabrutinib, a BTK-specific inhibitor, via oral gavage twice daily for 21 days and performed hematopoietic analysis including the degree of splenomegaly, monocytosis, and thrombocytopenia.  Results: The combination treatment scheme significantly decreased monocytosis and ameliorated thrombocytopenia compared to vehicle treated mice. Furthermore, splenomegaly was significantly reduced in the combination-treated mice compared to vehicle.   Conclusion and Potential Impact: Combination chemotherapy with PI3K p110d- and BTK-specific inhibitors profoundly corrects disease state hallmarks of JMML, and may warrant further clinical investigation of efficacy.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4602-4602
Author(s):  
Yuichiro Nakata ◽  
Takeshi Ueda ◽  
Akiko Nagamachi ◽  
Linda Wolff ◽  
Ogawa Seishi ◽  
...  

Abstract Myelodysplastic syndromes (MDS) are disorders originated from hematopoietic stem cells (HSCs), which are characterized by ineffective hematopoiesis, dysplasia mainly in the myeloid lineage, and high progression ratio to acute myeloid leukemia (AML). Recently, we identified mutations of the c-CBL (Casitas B cell lymphoma, a cellular homologue of v-CBL) gene in patients with MDS and myeloproliferative neoplasms (MPN). The mutations are detected in about 8% of the patients with the highest frequency in chronic myelomonocytic leukemia (CMML) cases with acquired uniparental disomy (aUPD) at 11q. c-CBL encodes a RING finger-based E3 ubiquitin ligase that negatively regulates receptor-mediated intracellular signaling. c-CBL is highly expressed in HSCs, strongly suggesting that it functions as a fine regulator of hematopoietic homeostasis. In fact, c-CBL knockout (KO) mice showed a myeloproliferative phenotype, owing to the hyper-responsiveness of HSCs to cytokine stimulation and subsequent augmented hematopoietic progenitor pool. In addition, c-CBL knockin (KI) mice harboring a mutation in the RING finger domain in one allele and a null mutation in the other allele exhibit an MPD-like disease and eventually progress to AML. These findings indicate that dysfunction of c-CBL perturbs normal hematopoietic development and contributes to hematopoietic abnormalities, but the precise leukemogenic mechanism(s) remains elusive. To gain insights into this issue and to create a novel animal model for mutated c-CBL-harboring leukemia, we generated conditional knock-in (cKI) mice that express wild-type c-CBL at steady state and inducibly express c-CBLQ367P, which was identified in patients with chronic myelomonocytic leukemia (CMML). After induced expression of c-CBLQ367P, the cKI mice exhibited a rapid and sustained increase in myelomonocytic cells with dysplasia in the peripheral blood and splenic enlargement with proliferation of myeloid cells, which closely resemble to the phenotype of human CMML. The bone marrow (BM) was hypercellular with predominance of myeloid cells, and increased number of HSC subpopulations and early myeloid progenitors were observed. In addition, phosphorylation of AKT, STAT3 and STAT5 was detected in long-term hematopoietic stem cells (LT-HSCs) of c-CBLQ367P cKI mice, indicating that PI3K/AKT and JAK/STAT signaling pathways are activated in c-CBLQ367P LT-HSCs. Moreover, competitive repopulation assays revealed that mice transplanted with c-CBLQ367P LT-HSCs showed significantly higher donor-derived chimerism than those transplanted with control LT-HSCs and displayed expansion of myelomonocytic cells as observed in c-CBLQ367P cKI mice, indicating that c-CBLQ367P conferred a proliferative advantage to LT-HSCs and that the phenotypes observed in c-CBLQ367P cKI mice were hematopoietic cell-intrinsic. CMML is known to progress to AML, possibly with additional genetic aberrations. To investigate the mechanism(s) underlying the disease evolution, we performed retroviral insertional mutagenesis using MOL4070A, a derivative of Moloney murine leukemia virus capable of inducing myeloid diseases. Almost all MOL4070A-infected c-CBLQ367P cKI mice developed AML, while no disease was observed in virus-injected control mice. Inverse PCR method identified Evi1 gene as a common integration site in the diseased mice and high Evi1 expression was detected in Evi1-integrated tumors. Mice transplanted with Evi1-transduced c-CBLQ367P cKI c-kit-positive BM cells developed AML at a high frequency and in a shortened period as compared to those transplanted with Evi1-transduced control cells. Taken together, we demonstrated that acquired expression of c-CBLQ367P plays a causative role in the development of CMML by activating PI3K/AKT and JAK/STAT pathways in HSCs and found that Evi1 overexpression cooperates with c-CBLQ367P to develop AML. Our mouse model provides a powerful tool for understanding of the pathogenesis of CMML and for developing novel therapeutic strategies. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 4 (14) ◽  
pp. 3246-3251
Author(s):  
Zhigang Cai ◽  
Chi Zhang ◽  
Jonathan J. Kotzin ◽  
Adam Williams ◽  
Jorge Henao-Mejia ◽  
...  

Abstract Mutations in PTPN11, which encodes the protein tyrosine phosphatase SHP2, contribute to ∼35% of cases of juvenile myelomonocytic leukemia (JMML). A common clinical picture in children with JMML is that it presents as a constitutive hyperinflammatory syndrome, partially reminiscent of chronic myelomonocytic leukemia in adults. Thus, a component of JMML is associated with a hyperinflammatory state and abundant innate immune cells such as neutrophils and monocytes. Recently, we showed that the evolutionarily conserved mouse lncRNA Morrbid is specifically expressed in myeloid cells and uniquely represses the expression of the proapoptotic gene Bim to regulate the lifespan of myeloid cells. However, its role in JMML has not been investigated. In this study, we characterized the role of Morrbid and its target Bim, which are significantly dysregulated in Shp2E76K/+-bearing myeloid cells, in driving JMML. Loss of Morrbid in a mouse model of JMML driven by the Shp2E76K/+ mutation resulted in a significant correction of myeloid and erythroid cell abnormalities associated with JMML, including overall survival. Consistently, patients with JMML who had PTPN11, KRAS, and NRAS mutations and high expression of MORRBID manifested poor overall survival. Our results suggest that Morrbid contributes to JMML pathogenesis.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2623-2623
Author(s):  
Roshni Patel ◽  
Baskar Ramdas ◽  
Lisa Deng ◽  
Victoria Jideonwo-Auman ◽  
Rebecca J Chan ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) is an aggressive childhood myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN). It has no effective treatments and the only treatment that prolongs survival is allogeneic hematopoietic stem cell transplant (HSCT). However, even with this aggressive intervention, approximately 50% of patients relapse with JMML within five years. The most commonly mutated gene in JMML patients is PTPN11, which encodes for the non-receptor protein tyrosine phosphatase SHP2. Gain-of-function (GOF) mutations in SHP2 lead to hyperactive RAS signaling and PTPN11 is recognized as a well-established oncogene in various leukemia's. Our lab has shown that p110δ, the hematopoietic-specific catalytic subunit of phosphoinositide 3-kinase (PI3K), plays an important role downstream of SHP2-signaling. Recently, we have shown that in vivo treatment with a specific pharmacologic p110δ inhibitor significantly prolongs the overall survival and reduces the splenomegaly seen in GOF Shp2-expressing mice. Having observed the effectiveness of PI3K p110δ inhibition in correcting the mutant Shp2-induced leukemia phenotype in vitro and in prolonging survival of mice in vivo, we next explored signaling molecules with which p110δ may be interacting to promote the aberrant Shp2 signaling in myeloid cells. In recent years, a key player in B cell receptor (BCR) signaling, Bruton's tyrosine kinase (BTK), has come under intense study in the field of lymphocytic leukemia and lymphoma research. Ibrutinib, a small molecule inhibitor targeting BTK, has proven to be very effective and has received FDA-approval for the treatment of a variety of B cell malignancies. Although research on BTK up to this point has focused on B cell malignancies, BTK is also highly expressed in myeloid cells and mice lacking BTK are defective in many myeloid cell functions. Thus, it is possible that BTK signaling may also play an important role in myeloid malignancies, such as JMML. Given the collaboration of BTK and p110δ in BCR signaling, the key role of p110δ in GOF Shp2-induced leukemia, and the high expression of BTK in myeloid cells, we hypothesized that BTK and p110δ function cooperatively in GOF Shp2-expressing myeloid cells to promote MPN. To test this hypothesis, we examined the potential collaboration of GM-CSF-stimulated BTK and p110δ in GOF Shp2-expressing myeloid cells and tested the inhibitor combination in vivo. We show that BTK cooperates with p110δ to promote GOF Shp2-induced leukemia. We show a role for B cell adaptor for PI3K (BCAP) in BTK upregulation of PI3K activity. In mutant Shp2 macrophages, BCAP phosphorylation is increased specifically in the larger isoforms regulating PI3K activation, and BTK inhibition results in a dose-dependent reduction in this phosphorylation. Our findings show that the MPN caused by GOF Shp2 is due to cooperative signaling between p110δ and BTK, which forms a positive feedback loop with BCAP, thus leading to more Akt/Erk hyperphosphorylation and hyperproliferation in response to GM-CSF. Given these in vitro observations, we treated GOF Shp2 bearing mice with a combination of a PI3K p110δ-specific inhibitor and, a BTK-specific inhibitor, or single agents alone and performed hematopoietic analysis. The combination treatment scheme completely rescued monocytosis and uniquely ameliorated thrombocytopenia compared to single agent treatment of diseased mice. In addition, splenomegaly was also completely rescued in all three treatment groups compared to vehicle. Detailed flow cytometric analysis of bone marrow hematopoietic stem and progenitor cell populations revealed significant repression of LSK, HPC1 and granulocyte macrophage progenitors (GMPs) with a concomitant increase in the megakaryocyte precursors (MKPs) in drug combination treated mice vs. other groups. Consistent with these observations, a significant reduction in mature myeloid cells was noted in the spleen and a complete rescue in the production of peripheral blood platelets was observed in the mutant mice treated with a combination of the two drugs. Thus, combination therapy with PI3K p110δ- and BTK-specific inhibitors profoundly rescues disease state hallmarks of JMML, and may warrant further clinical investigation. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 4 (15) ◽  
pp. 3728-3740
Author(s):  
Nitza Vara ◽  
Yuqing Liu ◽  
Yan Yan ◽  
Shelly Y. Lensing ◽  
Natalia Colorado ◽  
...  

Abstract It is not clear whether disrupted age-specific hematopoiesis contributes to the complex manifestations in leukemia patients who carry identical mutations, particularly in pediatric and adult patients with similar clinical characteristics. By studying a dual-age–specific mouse model, we demonstrate that (1) loss of Pten during the fetal-to-adult hematopoiesis switch (hematopoiesis switch) causes sustained fetal hematopoiesis, resulting in death in juvenile leukemia; (2) myeloid-biased hematopoiesis in juvenile mice is associated with the sustained fetal properties of hematopoietic stem cells (HSCs); (3) the age specificity of juvenile myelomonocytic leukemia depends on the copy number of Pten and Nf1; (4) single-allelic Pten deletion during the hematopoiesis switch causes constitutive activation of MAPK in juvenile mice with Nf1 loss of heterozygosity (LOH); and (5) Nf1 LOH causes monocytosis in juvenile mice with Pten haploinsufficiency but does not cause lethality until adulthood. Our data suggest that 1 copy of Pten is sufficient to maintain an intact negative-feedback loop of the Akt pathway and HSC function in reconstitution, despite MAPK being constitutively activated in juvenile Pten+/ΔNf1LOH mice. However, 2 copies of Pten are required to maintain the integrity of the MAPK pathway in juvenile mice with Nf1 haploinsufficiency. Our data indicate that previous investigations of Pten function in wild-type mice may not reflect the impact of Pten loss in mice with Nf1 mutations or other genetic defects. We provide a proof of concept that disassociated age-specific hematopoiesis contributes to leukemogenesis and pediatric demise.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1471-1471
Author(s):  
Santhosh Kumar Pasupuleti ◽  
Baskar Ramdas ◽  
Kai Yang ◽  
Chujing Zhang ◽  
Elliot Stieglitz ◽  
...  

Abstract Tumor-associated macrophages (TAMs) are a key component of tumor-infiltrating immune cells. Macrophages are largely characterized as M1 or M2 types, and TAMs have been shown to express an M2-like phenotype. TAMs endorse tumor progression and contribute to resistance to chemotherapies. However, it is unclear what the composition of M2 macrophages is in patients with Juvenile myelomonocytic leukemia (JMML) and how do these cells mechanistically contribute to JMML and/or relapse after bone marrow transplantation. To study the role of M2- TAMs in JMML development, we first examined the bulk RNA-sequence data in 90 JMML patients. These data demonstrated a significant increase in the expression of arginase-1 (Arg-1) and programmed cell death-1 (PD-1). Furthermore, single cell RNA-sequencing analysis of monocytes/macrophages from 4 JMML patients revealed higher expression of M2- macrophage markers/genes such as IL-10, CD163, MRC1/CD206, TGF-β1 and IL-1R1 compared to M1 macrophage (CD80, CCR7, IL-6, CXCL10, CXCL11 and TNF) expression. We hypothesized that in JMML, inflammatory myeloid cells including neutrophils and M2-macrophages express higher levels of arginase and PD-1, which may contribute to the local suppression of immune responses and damage the bone marrow microenvironment (BME) leading to poor engraftment of normal donor cells, resulting in relapse. To study how alterations in bone marrow (BM) macrophages (M1/M2) contribute to JMML development and relapse, we utilized a mouse model bearing Shp2 E76K mutation (Ptpn11 E76K/+) driven by lysosome-cre (Ptpn11 E76K/+; LysM-Cre+, indicated as Shp2* mice hereafter). This model is frequently used to study JMML as it manifests cardinal features of human JMML. In a competitive transplantation experiment using, Shp2* + Boy/J BM cells (1:1 ratio) transplanted into lethally irradiated Shp2* recipient mice, we show that Shp2* mutant cells out compete WT BoyJ cells and result in rapid growth of CD45.2+ Shp2* mutant mature myeloid cells, hematopoietic stem and progenitors (HSC/Ps) and M2- macrophages (F4/80+/CD206+) in the BM and spleen leading to leukemia relapse. To determine if modulating Arg-1 and PD-1/PD-L1 levels in the background of Shp2* mutant leukemic stem cells in Shp2* recipients would alter the overall engraftment and JMML development and relapse, we again performed a competitive transplantation experiment using, Shp2* + Boy/J (BM cells, 1:1 ratio) into Shp2* and WT recipient mice. After 8 weeks post transplantation, we investigated the role of Arg-1 and PD-L1 in Shp2* recipients using pharmacological inhibitors, CB-1158 (Arg-1 inhibitor; 100 mg/kg, orally) + anti-PD-L1 antibody (10 mg/kg, i.p) for 30 days. The Arg-1 + PD-L1 treatment significantly reduced the number of white blood cells, neutrophils, monocytes and improved RBC and platelet counts. The spleen and liver weights were significantly rescued as well. Interestingly, CD45.1 WT donor cells in the PB, BM, and spleen were significantly increased and a significant reduction of Shp2* mutant CD45.2+ mature myeloid cells in the PB, BM, and spleen was observed. Importantly, the frequency and absolute number of leukemic blasts, LSK (Lin-/Sca1+/c-KIT+) cells, short term hematopoietic cells (ST-HSCs), common myeloid progenitors (CMP), granulocyte macrophage progenitors (GMP) and megakaryocyte erythroid progenitors (MEP) were significantly reduced. Furthermore, the M2- TAMs were significantly reduced in the BM and spleen of Arg-1 + PD-L1 drug treated group compared to vehicle treated mice. Notably the CD8+ T-cells (IFN-γ+ and TNF-α+) were significantly improved in the drug treated mice. These data suggest that the suppression of arginase-1 allows for the arginine levels to increase, which promotes the proliferation of T-cells. Increasing arginine levels also promotes an anti-tumor immune response resulting in the emergence of CD45.1 WT HSCs as opposed to mutant CD45.2 HSCs, suggesting that Arg-1 + PD-L1 treatment is a novel therapeutic approach to treat patients with JMML and for preventing leukemia relapse after BM transplantation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1550-1550
Author(s):  
Yan Yan ◽  
Natalia C Colorado ◽  
Shelly Lensing ◽  
Delli R Robinson ◽  
Nicholas J Baltz ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) and chronic myelomonocytic leukemia (CMML) are mixed myelodysplastic syndromes (MDS)/myeloproliferative neoplasms (MPNs) that share similar clinical manifestations, but differ in prognosis and ages at diagnosis. Both are characterized by monocytosis and monocytic infiltration in vital organs. JMML is extremely aggressive, with death occurring within one year after diagnosis in children (<6 Yrs) without therapy, whereas CMML presents in adults with a more indolent course. A dysregulated RAS pathway is the key mechanism of JMML and CMML, but the number of non-synonymous mutations per tumor in JMML patients is ranked among the least of all human cancers. Previously, we reported that mice with somatic Pten deletion and germline mutant Nf1 (PtenΔ/ΔNf1wt/-) mimic human MPNs with features of a pediatric mixed MDS/MPNs, in terms of age of onset and organ infiltration with myelomonocytic cells, but lack a key feature of JMML with respect to GM-CSF hypersensitivity (Blood 2016; 127:1912). In order to simulate the molecular dynamics observed in JMML, we introduced Nf1 Loss-Of-Heterozygosity (Nf1LOH) by somatically deleting the second copy of Nf1 in mice on postnatal day 8 (PND8, equivalent to a full-term newborn age in humans).We generated mice with PtenfloxP/floxPNf1Fcr/floxPMx1-Cre+on a C57BL6/129 genetic background, and conditionally deleted Pten and the second copy of Nf1 myeloid-specifically by intraperitoneal injection of Poly(I:C). Mice with bi-allelic Pten deletion and Nf1LOH (PtenΔ/ΔNf1LOH) were born healthy but revealed signs of sickness in the 2nd week of life, and all died by age 4 weeks (equivalent to 1-3 years old in humans); whereas mice with heterozygous Pten deletion and Nf1LOH (Pten wt/ΔNf1LOH) did not show obvious signs of disease until age 2 months, and possessed a lifespan longer than 3 months. The natural lifespan of PtenΔ/ΔNf1LOH mice (n=8) was significantly shorter than littermates with PtenΔ/ΔNf1wt/Δ (n=18), Pten wt/ΔNf1LOH(n=10), or wild type (ptenwtNf1wt, hereafter referred asWT, n=14, p<0.001). We analyzed 13 PtenΔ/ΔNf1LOH mice at age PND18-21 along withtheir littermates. Substantial hepatosplenomegaly was observed in mice with Pten deletion and correlated with the loss of copy number of Pten and Nf1 inthe following order: PtenΔ/ΔNf1LOH> PtenΔ/ΔNf1wt/Δ > Pten wt/ΔNf1LOH> WT (median spleen weight: 228>180>134>56mg, respectively, p<0.001). Mice with PtenΔ/ΔNf1LOH(n=13) or Pten wt/ΔNf1LOH(n=12) had significantly elevated white blood cells (WBCs) and a lower hemoglobin than PtenΔ/ΔNf1wt/Δ (n=21) or WT littermates (n=15). HE stained tissue sections of formalin-fixed organs revealed increased cellularity in bone marrow (BM), and severe infiltration of mature myeloid cells in the spleens, livers, and lungs of mice with PtenΔ/ΔNf1LOH. Flow cytometry analysis (FACS) revealed that PtenΔ/ΔNf1LOHmice had significantly increased monocyte/macrophages and granulocytes in BM and peripheral blood (PB), but significant reduction in T-cells and B-cells in PB and spleen. A significant increase in Ter119+CD71high cells was also found in PtenΔ/ΔNf1LOH mice, indicating MDS or leukemia. Interestingly, splenic marginal zone B-cells were significantly lower exclusively in PtenΔ/ΔNf1LOHmice. Mice with Ptenwt/ΔNf1LOHhad the similar FACS profile with less disease severity at age PND18-21. They lived longer than 3 months without obvious signs of disease except increased WBCs and mild anemia. BM cells from PtenΔ/ΔNf1LOH mice at an age of 3 weeks were hypersensitive to GM-CSF, but Ptenwt/ΔNf1LOH mice did not display this until an age of 2 months. Based on the Bethesda proposals for classification of non-lymphoid hematopoietic neoplasms in mice, we have developed a mouse model of a mixed MDS/MPNs with features of JMML and CMML, in terms of age of onset and organ infiltration with myelomonocytic cells, and GM-CSF hypersensitivity. In conclusion, our data demonstrate that bi-allelic Pten deletion and Nf1LOH at an early age results in JMML in mice, whereas single-allelic Pten deletion with Nf1LOH induces CMML. This is the first mouse model that differentially resembles pediatric and adult mixed MDS/MPNs, and closely mimics the true genetic and epigenetic dynamics in humans. It provides a novel tool for studying the mechanisms underlying pediatric and adult mixed MDS/MPNs. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 225 (03) ◽  
Author(s):  
S Fluhr ◽  
T Witte ◽  
CF Krombholz ◽  
C Plass ◽  
CM Niemeyer ◽  
...  

Author(s):  
William A. Lambert ◽  
Joseph A. DiGiuseppe ◽  
Tatiana Lara-Ospina ◽  
Markus J. Bookland ◽  
Jonathan E. Martin ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document