Heterodimer-Specific Stimulation Of Toll-Like Receptor 2 Induces Divergent Downstream Effects In Primary Samples Of Precursor B Cell Acute Lymphoblastic Leukemia

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3918-3918
Author(s):  
Nina Rolf ◽  
Amina Kariminia ◽  
Sabine M Ivison ◽  
Gregor SD Reid ◽  
Kirk R. Schultz

Abstract Introduction Reports of spontaneous acute lymphoblastic leukemia (ALL) remissions following severe bacterial infections or administration of Coley's bacteria extract intriguingly suggest that bacterial lipopeptides may trigger effective immune-mediated eradication of ALL. Coley's extract is now known to have delivered a “perfect storm of Toll-like receptor (TLR) agonists” providing potent immune-stimulation. Considerable preclinical and clinical data verified strong anticancer effects for purified TLR ligands. To date, however, TLR2 ligands remain the least studied despite several unique characteristics of TLR2: i) recognition of most bacterial and other pathogens through hetero-dimerization with TLR1 or 6 (all surface-localized, easily ligand-accessible, consistently expressed on most immune cells); ii) mediation of direct cytotoxicity against malignant cells; and iii) distinction of healthy from damaged self by recognizing endogenous ligands from damaged cells. Building on work previously presented at ASH, the aim of this study was to investigate the effects of TLR2 stimulation on primary ALL and to delineate whether heterodimer-specific TLR2 stimulation resulted in divergent functional downstream effects that could contribute to the generation of anti-leukemia immune activity. Methods Pam3CSK4 (Pam3) and Pam2CSK4 (Pam2) are two well-characterized heterodimer-specific synthetic TLR2 ligands not known to cause overwhelming sepsis in vivo. Recently, their exact binding properties were identified by crystallography, offering a reliable in-vitro model to study the potentially divergent functional downstream effects of TLR2/1 (Pam3) vs TLR2/6 (Pam2) stimulation. We investigated whether both TLR2 ligands a) modulate CD40 expression and b) induce death in 4 primary ALL samples and 4 ALL cell-lines. Furthermore, using the best responding cell-line, we investigated which downstream signaling pathways were activated by either ligand. Results TLR1, 2 and 6 expression was confirmed on all primary ALL and ALL cell lines. As shown in Table 1, all 4 primary ALL samples were highly responsive to stimulation with TLR2 ligands as both Pam2 and Pam3 strongly upregulated CD40 expression. However, only Pam3 induced death of leukemic blasts. A similar response pattern was observed with cell lines, albeit stronger and more consistent in primary ALL. Antibody blockade of TLR2 abrogated Pam2-mediated CD40 expression. Blocking of both TLR2 and TLR1 was required to reduce Pam3-mediated CD40 upregulation. Pam3-induced cell death was identified as caspase-mediated apoptosis without ROS activation. Both Pam2 and Pam3 strongly induced NFkB and PI3K pathways, though with distinct signaling kinetics that may underlie their divergent effects on ALL blasts. Conclusions Our results reveal that synthetic TLR2 ligands are potent stimulators of primary ALL blasts and we demonstrate that heterodimer-specific stimulation resulted in distinct functional downstream responses not previously reported. Close association of danger signals with dying cells critically shapes the immune response: although both TLR2 ligands stimulate ALL cells, the ability of Pam3 to augment CD40 expression while simultaneously inducing cell death achieves the conditions recognized as necessary for generating potent immunological responses. Our study, therefore, indicates that the TLR2/1 ligand Pam3 possesses significant potential for generating anti-ALL immune activity through its direct effects on leukemic blasts. Disclosures: No relevant conflicts of interest to declare.

Cancers ◽  
2019 ◽  
Vol 11 (11) ◽  
pp. 1654 ◽  
Author(s):  
Ghelli Luserna Di Rorà ◽  
Bocconcelli ◽  
Ferrari ◽  
Terragna ◽  
Bruno ◽  
...  

Introduction: Screening for synthetic lethality markers has demonstrated that the inhibition of the cell cycle checkpoint kinases WEE1 together with CHK1 drastically affects stability of the cell cycle and induces cell death in rapidly proliferating cells. Exploiting this finding for a possible therapeutic approach has showed efficacy in various solid and hematologic tumors, though not specifically tested in acute lymphoblastic leukemia. Methods: The efficacy of the combination between WEE1 and CHK1 inhibitors in B and T cell precursor acute lymphoblastic leukemia (B/T-ALL) was evaluated in vitro and ex vivo studies. The efficacy of the therapeutic strategy was tested in terms of cytotoxicity, induction of apoptosis, and changes in cell cycle profile and protein expression using B/T-ALL cell lines. In addition, the efficacy of the drug combination was studied in primary B-ALL blasts using clonogenic assays. Results: This study reports, for the first time, the efficacy of the concomitant inhibition of CHK1/CHK2 and WEE1 in ALL cell lines and primary leukemic B-ALL cells using two selective inhibitors: PF-0047736 (CHK1/CHK2 inhibitor) and AZD-1775 (WEE1 inhibitor). We showed strong synergism in the reduction of cell viability, proliferation and induction of apoptosis. The efficacy of the combination was related to the induction of early S-phase arrest and to the induction of DNA damage, ultimately triggering cell death. We reported evidence that the efficacy of the combination treatment is independent from the activation of the p53-p21 pathway. Moreover, gene expression analysis on B-ALL primary samples showed that Chek1 and Wee1 are significantly co-expressed in samples at diagnosis (Pearson r = 0.5770, p = 0.0001) and relapse (Pearson r= 0.8919; p = 0.0001). Finally, the efficacy of the combination was confirmed by the reduction in clonogenic survival of primary leukemic B-ALL cells. Conclusion: Our findings suggest that the combination of CHK1 and WEE1 inhibitors may be a promising therapeutic strategy to be tested in clinical trials for adult ALL.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3245-3245
Author(s):  
Khimara Naidoo ◽  
Waleed Alduaij ◽  
Jamie Honeychurch ◽  
Eleanor Cheadle ◽  
Seema Alexander ◽  
...  

Abstract Abstract 3245 Whilst modern treatment approaches cure a high number of patients with acute lymphoblastic leukemia (ALL), little progress has been made in the treatment of refractory and relapsed ALL and new treatment approaches are needed. We recently demonstrated that anti-HLA-DR class II monoclonal antibody (mAb) L243 induces a novel non-apoptotic mode of cell death in B-cell lymphoma lines, defined by homotypic adhesion (HA), actin reorganisation and lysosomal activity (Ivanov et al. J Clin Invest, 2009). Here, we extend these important observations and examine whether this novel form of mAb induced cell death occurs in pre-B ALL cell lines. Expression of HLA-DR was determined using flow cytometry in a panel of ALL cell lines (REH, SupB15 and SD1). HLA-DR was expressed at high levels on each of the cell lines. The ability of L243 to induce HA and cell death (Annexin V/PI positivity) was assessed using microscopy and flow cytometry respectively. L243 was able to evoke both strong HA and cell death in all of the ALL cell lines (e.g. in SupB15 cells 46±1.7% death versus 7±0.5% in controls, p<0.001 by Student's t-test). Inhibitors of actin polymerization (cytochalasin D, latrunculin B) were used to assess the role of actin in cell death and HA induced by L243. These inhibitors of actin polymerization inhibited both HA and cell death elicited by L243 (e.g. in SupB15 cells 24±0.5% death versus 10.3±0.8% with Latrunculin B, p<0.001), demonstrating the dependence of HA and cell death on actin reorganisation. The importance of cell to cell contact in this form of antibody induced cell death was confirmed by the addition of low-melting point agarose which physically blocked cell to cell contact and markedly attenuated cell death induced by L243. In contrast using the pan-caspase inhibitor QVD OPH had no effect on cell death induced by L243, indicating that this mode of death is non-apoptotic. These findings demonstrate that anti-HLA DR mAb L243 induces a novel model of cell death in ALL cell lines that is independent of caspase activation and dependent on actin reorganization. This data suggests that this novel mAb induced death pathway is independent of apoptosis and potentially exploitable in the clinic in leukemias resistant to chemotherapy apoptosis induction. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1426-1426
Author(s):  
Craig T Wallington-Beddoe ◽  
Stuart M Pitson ◽  
Jason A Powell ◽  
Kenneth F Bradstock ◽  
Linda J Bendall

Abstract Sphingosine 1-phosphate (S1P) is a bioactive lipid with roles in cell proliferation and survival. S1P is produced by the sphingosine kinases, SphK1 and SphK2. SphK1 is over-expressed in a number of malignancies and evidence points overwhelmingly to a pro-survival role. Furthermore, SphK1 has been shown to correlate with the clinical outcome of certain tumors. Here we focus on SphK1 as an important oncogenic target in acute lymphoblastic leukemia (ALL). We have previously shown SphK1 protein to be over-expressed and activated (Ser225 phosphorylation) in ALL cell lines and primary patient samples compared to normal CD34+CD19+ B-cell progenitors. Furthermore, we have reported the importance of SphK1 in the development of ALL by transducing B-cell progenitors isolated from WT or SphK1-/- mice with the ALL associated p185 form of the oncogenic fusion gene BCR/ABL and injecting transduced cells into sub-lethally irradiated WT mice. The absence of SphK1 significantly reduced the incidence of ALL in recipient mice (ASH 2012). Inhibition of SphK1 by the selective inhibitor SK1-I significantly reduced intra-cellular S1P concentrations (p=0.017 and p=0.003 at 24 and 48 hours respectively) in 3 cell lines examined, indicating that the drug targets this enzyme. SK1-I killed ALL cells as determined by annexin V/PI flow cytometric analysis with IC50 values ranging from 12 µM to 18 µM at 72 hours. Furthermore, SK1-I induced cell death in primary patient ALL cells by 16 hours. This agent resulted in virtually no caspase-3 cleavage and cell death was not prevented by the pan-caspase inhibitor Z-VAD-FMK (p=0.45, n=4). Marked cytoplasmic vacuolation was detected by light microscopy, with LC3 processing present by Western blot, consistent with the development of autophagy. However, the autophagy inhibitor 3MA failed to prevent SK1-I-mediated cell death. These results suggest that the cell death associated with inhibition of SphK1 in ALL cells is caspase-independent and cannot be attributed to autophagy. Surprisingly, conventional chemotherapeutic agents such as doxorubicin and vincristine failed to synergize with SK1-I, however, synergistic killing was observed when SK1-I was combined with 500 nM imatinib over 72 hours in Philadelphia-positive (BCR/ABL+) ALL cells. We have developed a novel SphK1 inhibitor, MP8, that targets the enzyme via a different mechanism to SK1-I, since it blocks ATP binding. MP8 reduced intra-cellular S1P in Jurkat cells by 43% compared to untreated controls, and killed Jurkat and SUP-B15 cells over 24 to 48 hours with IC50 values of 8 µM. Additionally, MP8 induced cell death in primary patient ALL cells by 24 hours. This agent resulted in classic apoptotic cell death, which was rescued by Bcl-2 over-expression, resulting in near complete reversal of PARP cleavage. SphK1 has indisputable tumor-promoting properties and lies downstream of a number of signalling pathways known to be dysregulated in ALL. Here we show that SphK1 is over-expressed and activated in ALL cells and targeting SphK1 has potent cytotoxic effects in a wide range of cell lines and patient samples. Furthermore, genetic deletion of Sphk1 significantly reduced the incidence of murine BCR/ABL-driven ALL. These findings suggest further examination of the role SphK1 plays in ALL will uncover novel interactions with oncogenic signalling pathways and paves the way for the inclusion of SphK1 inhibitors in future pre-clinical trials. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 22 (2) ◽  
pp. 651
Author(s):  
Pablo Cruz ◽  
Ulises Ahumada-Castro ◽  
Galdo Bustos ◽  
Jordi Molgó ◽  
Daniela Sauma ◽  
...  

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy whose chemoresistance and relapse persist as a problem despite significant advances in its chemotherapeutic treatments. Mitochondrial metabolism has emerged as an interesting therapeutic target given its essential role in maintaining bioenergetic and metabolic homeostasis. T-ALL cells are characterized by high levels of mitochondrial respiration, making them suitable for this type of intervention. Mitochondrial function is sustained by a constitutive transfer of calcium from the endoplasmic reticulum to mitochondria through the inositol 1,4,5-trisphosphate receptor (InsP3R), making T-ALL cells vulnerable to its inhibition. Here, we determine the bioenergetic profile of the T-ALL cell lines CCRF-CEM and Jurkat and evaluate their sensitivity to InsP3R inhibition with the specific inhibitor, Xestospongin B (XeB). Our results show that T-ALL cell lines exhibit higher mitochondrial respiration than non-malignant cells, which is blunted by the inhibition of the InsP3R. Prolonged treatment with XeB causes T-ALL cell death without affecting the normal counterpart. Moreover, the combination of XeB and glucocorticoids significantly enhanced cell death in the CCRF-CEM cells. The inhibition of InsP3R with XeB rises as a potential therapeutic alternative for the treatment of T-ALL.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 903-903
Author(s):  
Salih Demir ◽  
Galina Selivanova ◽  
Eugen Tausch ◽  
Lisa Wiesmüller ◽  
Stephan Stilgenbauer ◽  
...  

Abstract Mutations of the tumor suppressor gene TP53 have been described to be associated with aggressive disease and inferior prognosis in different types of cancer, including hematological malignancies. In acute lymphoblastic leukemia (ALL), TP53 alterations are infrequently found at diagnosis but have recently been described in about 12% of patients at relapse. This suggests an association with therapy resistance in high risk/relapsed ALL and patients with TP53 mutated ALL have in fact an inferior outcome. Small molecule compounds targeting mutated TP53 such as APR-246, initially described as PRIMA-1MET (p53-dependent reactivation and induction of massive apoptosis) leading to apoptosis induction have shown activity in several types of malignancies with mutated TP53. In ALL, however, mutant TP53 has so far not been addressed as a target for therapeutic intervention. In this study, we investigated a large cohort of patient-derived pediatric B cell precursor (BCP)-ALL primograft samples to identify cases with mutated TP53. Further, we analyzed the effects of APR-246 and evaluated its activity on BCP-ALL cell lines and primografts with mutated (mut) orwild type (wt) TP53. Altogether, 62 BCP-ALL primograft samples established from patients at diagnosis (n=53) or relapse (n=9) by transplantation of primary ALL cells onto NOD/SCID mice were screened for TP53 mutations by denaturating high-performance liquid chromatography (dHPLC) followed by Sanger sequencing of exons 4 to 10 to confirm detected mutations. We identified 4 cases with TP53 mut, 3 obtained from diagnosis (5.6%) and one at relapse (11.1%), corresponding to frequencies described in clinical studies. Mutated cases were further analyzed by fluorescence in situ hybridization (FISH), revealing a 17p deletion in one TP53 mut sample. Similarly, we analyzed 6 BCP-ALL cell lines and identified 2 TP53 mut and 4 TP53 wt lines. Exposure of BCP-ALL primograft (TP53 mut n=4, TP53 wt n=4) and cell line (TP53 mut n=2, TP53 wt n=4) samples to the DNA damaging agent doxorubicin showed, as expected, resistance of TP53 mut leukemia cells for cell death induction, reflected by significantly higher half maximal inhibitory concentrations (IC50; TP53 mut 49 and 143 ng/ml, TP53 wt mean 12 ng/ml) and lower induction of cell death (TP53 mut 16 to 23%, TP53 wt 10 to 60%) in TP53 mut ALL, corroborating the tumor-suppressive function of p53 in ALL. We then investigated the sensitivity of BCP-ALL cell lines for cell death induction by APR-246 (kindly provided by Aprea, Stockholm, Sweden). We observed high sensitivity for APR-246 in TP53 mut (IC50: 5 µM for both cell lines) as compared to TP53 wt ALL (mean IC50: 58 µM). DNA fragmentation and Annexin-V/propidium-iodide (PI) positivity revealed apoptosis as mechanism of APR-246 mediated cell death. Reactive oxygen species (ROS) have recently been described to mediate APR-246 induced cell death in multiple myeloma cells. Therefore, we investigated ROS levels by detection of oxidation-specific fluorescence of dichlorodihydrofluorescein diacetate (DCFDA) in ALL cells. Interestingly, ROS quenching by N-acetyl cysteine abolished induction of cell death in TP53 mut but not TP53 wt ALL cells indicating ROS as a mediator of APR-246 induced cell death in TP53 mut ALL. Furthermore, we addressed p53 activation in response to APR-246 by assessing phosphorylation of p53 (p53pSer15) using phosphoflow cytometry. Most interestingly, APR-246 led to 6-fold increased p53pSer15 levels in TP53 mut compared to no activation in TP53 wt leukemia cells, indicating restoration of p53function upon APR-246treatment in BCP-ALL. Based on these findings, we addressed the effectivity of APR-246on primary, patient-derived primografts and compared sensitivities for cell death induction in TP53 mut (n=4) and TP53 wt (n=4) samples. Importantly, the pattern of responsiveness of TP53 mut ALL was also identified in TP53 mut patient-derived ALL samples with induction of significantly higher cell death rates in TP53 mut ALL (TP53 mut 48%, TP53 wt 18%, 5 µM APR-246, 24 h). Taken together, we showed that TP53 mut BCP-ALL can be targeted by APR-246 leading to re-activation of p53, induction of ROS dependent apoptosis and effective leukemia cell killing. Thus, targeting and re-activation of mutated p53 provides a promising novel strategy for therapeutic intervention in this high-risk subtype of BCP-ALL. Disclosures Selivanova: Aprea: Patents & Royalties: APR-246. Tausch:Gilead: Other: Travel support. Stilgenbauer:Gilead: Honoraria, Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 280-280 ◽  
Author(s):  
Ernesto Diaz-Flores ◽  
Evan Q. Comeaux ◽  
Kailyn Kim ◽  
Kyle Beckman ◽  
Kara L. Davis ◽  
...  

Abstract Acute lymphoblastic leukemia (ALL) is the most common cancer of childhood. Specific genetic subsets, including hypodiploid ALL, are associated with particularly high rates of relapse. Despite the poor outcomes of hypodiploid B-ALL with traditional therapeutic approaches, there have been no known effective alternative therapies or novel candidates tested to improve outcome. We hypothesized that new therapeutic targets could by identified by integrated biochemical and genomic profiling, combined with functional drug assays in order to determine which pathways play an essential role in transformation. For biochemical profiling, we analyzed multiple pathways commonly deregulated in leukemias using phosphoflowcytometry (including receptor tyrosine kinases, JAK/STAT, MAPK, PI3K, PTEN, Bcl-2 survival and pro-apoptotic family members and p53). We subjected hypodiploid cell lines (NALM-16, MHH-CALL2) and patient derived xenograft samples in vitro to inhibitors against each of these pathways (PP2:Src family;Ruxolitinib: JAK/STAT; PD235901/CI1040: MAPK; GDC-0941, PI-90, PI-103, p110 (a, b, g, d): PI3K isoform specific; PP-242:mTOR; ABT-263/ABT-737: Bcl-2/Bcl-xl, and ABT-199: Bcl-2 specific). We found that the Bcl-2 inhibitors (ABT-263, ABT-737 and ABT-199) and to a lesser extent PI3K pathway inhibitors GDC-0941 and PP-242, but not the MAPK or RTK inhibitors, efficiently reduced proliferation of hypodiploid cells. However, only ABT-263/ABT-199 induced high levels of apoptosis at nanomolar concentrations. Based on the consistent efficacy observed with ABT-199 against hypodiploid patient-derived cells and cell lines in culture, we selected eight cryopreserved, previously xenografted (F3 generation) hypodiploid patient samples (4 low hypodiploid, chromosomal number between 32 and 39; and 4 Near Haploid, chromosomal number between 24 and 31) and three non-hypodiploid patient samples (Ph-positive,Ph-Like and Erg+) for a preclinical trial in immunodeficient mice. Each patient sample was engrafted into six mice, which were randomized to receive vehicle or ABT-199 daily over 60 days (Figure 1). Treatment started when the peripheral blood (PB) human CD45 count reached 15%. A rapid decrease in PB blasts was noted at 7 days (Figure 1). Eighty-five percent of the hypodiploid xenografts survived 60 days with either undetectable or low levels of leukemia in the PB. In contrastPh+ andPh-Like xenografts died within 10-20 days regardless of treatment. Importantly, hypodiploid leukemic blasts gradually emerged after discontinuing ABT-199 after 60 days. Additionally, despite low or undetectable levels of leukemic blasts in PB and reduced levels in bone marrow and spleen, all mice had high percentages of leukemic cells in the liver (Figure 2). In conclusion we have identified the survival protein Bcl-2 as a promising molecular target in hypodiploid B-ALL. ABT-199 for dramatically reduced leukemia cells in vitro and in vivo in patient-derived xenograft models of hypodiploid B-ALL. However, the liver represented a protective niche for these leukemias. In addition, our biochemical characterization of the organ infiltrating blasts collected from mice on trial indicate that the sensitivity of hypodiploid ALL to ABT-199 relies not only on high levels of Bcl-2 and deficiency for other survival proteins such as Bcl-xl but also on high levels of proapoptotic proteins, providing two different signatures that correlate with response to ABT-199. Using genome editing (CRISPR/Cas9) we interrogated the necessity for individual proapoptotic genes, including PUMA, NOXA, and BAD, for ABT-199-induced cell death. This study provides encouraging preclinical data that Bcl-2 may be a promising target for the treatment of hypodiploid B-ALL. Our studies identify signature biomarkers that correlate with drug response and identify essential proteins mediating ABT-199-induced cell death. Importantly, this report also identifies the limitations of using ABT-199 as single drug, and provides the rationale for using combinatorial therapies in order to improve the efficacy of the drug. Disclosures Mullighan: Loxo Oncology: Research Funding; Amgen: Speakers Bureau; Incyte: Membership on an entity's Board of Directors or advisory committees. Loh:Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Abbvie: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5108-5108
Author(s):  
Bill H Chang ◽  
Jeffrey W Tyner ◽  
Abdusebur Jemal ◽  
Mathew Thayer ◽  
Brian J Druker

Abstract Abstract 5108 Background: Pediatric Acute Lymphoblastic Leukemia (ALL) remains the most common pediatric malignancy. Despite advances in treatment and outcomes, there continue to be subsets of patients that are refractory to standard intensive chemotherapy and hematopoietic stem cell transplant. Therefore, novel gene targets for therapy are needed to further advance treatment for this disease. Survivin, a member of the chromosome passenger complex and inhibitor of apoptosis has been shown to be over-expressed in malignant cells and in relapsed disease. Therefore, survivin may be a potential target for therapy in pediatric ALL. Further, RNAi technology can be used as a rapid functional screen to identify target genes crucial for viability in ALL cell lines and in primary patient samples. Design/methods: Pediatric lymphoblastic cell lines and fresh primary mononuclear cells from newly diagnosed patients with ALL were used for all studies. Survivin was silenced by transfecting cells (via electroporation or transductin (IDT)) with siRNA SMARTpools obtained from Dharmacon. Protein expression was identified 48 hours after treatment with siRNA. Viability was measured using a standard methane-thiosulfonate viability assay. Activation of apoptosis was identified using the Guava nexin Annexin V binding assay. Result: Survivin is highly expressed in multiple ALL cell lines and in many primary ALL samples. Silencing of survivin expression with siRNA decreases viability and increases apoptosis in ALL cell lines and in primary ALL samples. Decreased protein expression of approximately fifty percent was sufficient to cause an increase in cell death. Finally, inhibition of P53 expression abrogates this phenotype suggesting that the mechanism of cell death induced following survivin silencing involves activation of the P53 pathway. Conclusion: Gene silencing with siRNA can be used as a rapid functional screen to identify potential targets for therapy. Implementing this technique identified survivin as a potential target for therapy in pediatric ALL. The results of these experiments will be used as a foundation to develop a comprehensive understanding of the mechanisms of survivin dependence in pediatric ALL. Future studies will also be designed to develop a model system for targeting of survivin in a therapeutically-relevant manner such that these findings can be translated into improved treatments for patients with ALL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 672-672
Author(s):  
Nadja Kopp ◽  
Christopher J. Ott ◽  
Liat Bird ◽  
Ronald Paranal ◽  
Jun Qi ◽  
...  

Abstract Abstract 672 Long-term disease-free survival is achieved in over 80% of children with B-cell acute lymphoblastic leukemia (B-ALL) but only 40% of adults. Particular genomic alterations in B-ALL, including rearrangements of MLL and the cytokine receptor subunit CRLF2, confer a poor prognosis in both children and adults. In addition, current therapies for B-ALL are associated with significant short- and long-term toxicities, highlighting the critical need for new therapeutics. The novel compound JQ1 inhibits the BET class of human bromodomain proteins from mediating the assembly of macromolecular protein complexes that are required for transcriptional activation and polymerase elongation. In hematologic and epithelial tumors, JQ1 can downregulate the expression of c-MYC and thereby suppress malignant growth and survival. We investigated the therapeutic potential of JQ1 across multiple genetically-defined subsets of B-ALL. JQ1 potently induced apoptotic cell death (IC50∼30–300 nM) in all B-ALL cell lines (697, CEMO-1 NALM-6, MHH-CALL4, MUTZ-5, Reh, RS4;11, SEMK2) studied. Among the most sensitive lines (IC50<50 nM) were MUTZ-5 and MHH-CALL4, which both harbor IGH@-CRLF2 translocations as well as activating mutations in JAnus Kinase 2 (JAK2). CRLF2 heterodimerizes with the IL7 receptor (IL7R) subunit in response to thymic stromal lymphopoietin, which induces JAK/STAT, MAP kinase and AKT signaling. To identify mechanisms through which JQ1 induces cell death in MUTZ-5 and MHH-CALL4 cells, we quantified transcript and protein levels for relevant targets in the presence of JQ1 500 nM or vehicle (DMSO). Chromatin immunoprecipitation was also performed with antibodies against BRD4 followed by PCR to determine the effects of JQ1 on BRD4 binding at relevant promoters. As previously observed, JQ1 induced the downregulation of MYC mRNA, loss of BRD4 at the MYC promoter, and reduced the expression of c-Myc target genes. Immunoblotting with phospho-specific antibodies demonstrated almost complete loss of JAK2 and STAT5 phosphorylation in cells treated with JQ1, suggesting that JQ1 also blocks signaling downstream of CRLF2/IL7R. While the levels of CRLF2 mRNA were unaffected by JQ1 in MUTZ-5 and MHH-CALL4 cells, JQ1 markedly downregulated IL7R mRNA and depleted BRD4 from the IL7R promoter in both lines. The reduction in IL7R mRNA levels led to dramatic decreases in IL7R surface expression. Genome-wide expression profiling demonstrated a highly restricted effect of JQ1, with IL7R and MYC being the 7th and 23rd most downregulated genes, respectively. In fact, IL7R was the only cytokine receptor in both CRLF2-rearranged B-ALL lines that was significantly downregulated by JQ1 treatment. In addition, JQ1 potently reduced IL7R mRNA across other B-ALL cell lines with diverse cytogenetics. To determine whether JQ1 could suppress the growth of human B-ALL in vivo, we xenografted a human CRLF2-rearranged B-ALL primary sample into Nod.SCID.IL2RG−/− mice. Upon the development of >30% bone marrow involvement by human CD45+/CRLF2+ B-ALL cells, mice were randomized to receive JQ1 (50mg/kg intraperitoneally daily) or vehicle (DMSO). After 5 days of treatment, sentinel mice were sacrificed for pharmacodynamic endpoints. Spleens from mice treated with JQ1 had markedly reduced c-Myc expression and STAT5 phosphorylation compared with spleens from vehicle-treated mice. In survival cohorts (n=9 per arm), treatment with JQ1 significantly prolonged overall survival (p=0.0002) compared with vehicle. These results demonstrate that BET bromodomain inhibition is a promising therapeutic strategy for patients with B-ALL, including subsets with high-risk cytogenetics. Moreover, the surprising finding that JQ1 also targets IL7R expression suggests that bromodomain inhibitors may be especially useful in malignant and nonmalignant disorders dependent on IL7R. Disclosures: Bradner: Tensha Therapeutics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Acetylon Pharmaceuticals: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; SHAPE Pharmaceuticals: Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Weinstock:Novartis: Consultancy, Research Funding.


APOPTOSIS ◽  
2006 ◽  
Vol 11 (11) ◽  
pp. 1977-1986 ◽  
Author(s):  
Dong-Joon Min ◽  
Naomi P. Moskowitz ◽  
Carrie Brownstein ◽  
Hokyung Lee ◽  
Terzah M. Horton ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4461-4461
Author(s):  
Greta De Jong ◽  
Sophie E Levie ◽  
Remko Schotte ◽  
Wouter Pos ◽  
Daniel Go ◽  
...  

Despite rapid advances in immunotherapeutic options for precursor B-acute lymphoblastic leukemia (ALL), outcomes remain poor especially for adult ALL and relapsed pediatric ALL. With conventional chemotherapy, remission percentages in adult ALL range from 75 to 90%, but relapse rates are high and long-term leukemia-free survival ranges between 35-70% depending on age and risk group. The introduction of CD19 targeting immunotherapy has significantly improved patient outcomes in (relapsed) B-ALL. However, tumor escape via downregulation of CD19 occurs in a significant number of patients. Therefore an ongoing urgency remains for the identification of additional or alternative immunotherapeutic targets for the treatment of ALL. AT1412 is an antibody that was identified from the peripheral blood memory B cell pool of a patient cured of metastatic melanoma after adoptive T-cell therapy, using a B cell immortalization technology (AIMSelect) with ectopic Bcl-6 and Bcl-xL expression as described previously [Kwakkenbos et al. Nat. Med. 2010]. The antibody was selected based on differential binding to melanoma cells as compared to healthy melanocytes and was shown to be successful in killing melanoma cells in vitro and in vivo [manuscript submitted]. In addition to melanoma, AT1412 binds other tumor types including B-ALL, gastric, colon- and pancreatic cancer. The target of AT1412 is the tetraspanin CD9, which is expressed by more than half of all B-ALL. Expression of CD9 has been correlated with adverse prognosis [Liang et al. Cancer Biomark. 2018]. We assessed binding of this human CD9 antibody to a panel of ALL cell lines using flow cytometry. Binding of AT1412 to the B-ALL cell lines SUP-B15, MHH-CALL-2 and CCRF-SB varied as expected based on the CD9 levels that we detected using a commercial CD9 antibody. AT1412 induced antibody dependent cellular cytotoxicity (ADCC) on these cells, in line with the level of AT1412 binding. No binding was seen to the T-ALL cell line Jurkat. Importantly, these findings were confirmed in primary ALL samples, obtained prospectively at diagnosis from a cohort of patients with T- or B-ALL (n=30). AT1412 showed binding to 61% of B-ALL samples but not to T-ALL samples. The potential of AT1412 to induce ADCC was tested on patient samples from the same panel. Remarkably, AT1412 induced ADCC of all B-ALL samples it bound to (8 out of 14) and of none of the T-ALL samples. Cytotoxicity significantly correlated with the level of AT1412 binding. These findings were supported by the observation that AT1412 induced B-ALL cell death when a freshly drawn whole bone marrow sample from a patient with newly diagnosed B-ALL was cocultured with AT1412. AT1412-induced cell death of B-ALL blasts occurred without affecting the monocytic, granulocytic and lymphocytic populations. This cell death was not observed when this patient's ALL blasts were incubated with AML-targeting antibodies. Remarkably, AT1412 induced cell death in the absence of added effector cells or other (chemo)therapeutic agents, while the bone marrow sample contained over 80% blasts and as little as 3% lymphocytes. We are currently investigating the in vivo efficacy of the antibody in a humanized immune system mouse model with human B-ALL. Taken together, the majority of precursor B-ALL blasts express CD9 and expression of CD9 is associated with a dismal outcome. Our data demonstrate that CD9 can be successfully targeted by the human CD9 antibody AT1412, suggesting that AT1412 has the potential to be developed as a therapeutic antibody for B-ALL. AT1412 is currently being advanced through preclinical development. Disclosures De Jong: AIMM Therapeutics: Employment. Levie:AIMM Therapeutics: Employment. Schotte:AIMM Therapeutics: Employment, Equity Ownership, Patents & Royalties: Patent WO2017119811A1. Pos:AIMM Therapeutics: Patents & Royalties: Patent WO2017119811A1. Go:AIMM Therapeutics: Employment, Patents & Royalties: Patent WO2017119811A1. Yasuda:AIMM Therapeutics: Employment, Equity Ownership. Cercel:AIMM Therapeutics: Employment. van Hal-van Veen:AIMM Therapeutics: Employment. Frankin:AIMM Therapeutics: Employment. Villaudy:AIMM Therapeutics: Employment, Equity Ownership, Patents & Royalties: Patent WO2017119811A1. van Helden:AIMM Therapeutics: Employment, Equity Ownership, Patents & Royalties: Patent WO2017119811A1. van Eenennaam:AIMM Therapeutics: Employment. Spits:AIMM Therapeutics: Employment, Equity Ownership, Patents & Royalties: Patent WO2017119811A1. Hazenberg:AIMM Therapeutics: Other: Employment/equity of partner/spouse.


Sign in / Sign up

Export Citation Format

Share Document