scholarly journals Rational drug combinations with CDK4/6 inhibitors in acute lymphoblastic Leukemia

Haematologica ◽  
2021 ◽  
Author(s):  
Karen L. Bride ◽  
Hai Hu ◽  
Anastasia Tikhonova ◽  
Tori J. Fuller ◽  
Tiffaney L. Vincent ◽  
...  

Despite improvements in outcomes for children with B and T-cell acute lymphoblastic leukemia (B-ALL and T-ALL), patients with resistant or relapsed disease fare poorly. Previous studies have demonstrated the essential role of cyclin D3 in T-ALL disease initiation and progression and that targeting of the CDK4/6-cyclin D complex can suppress T-ALL proliferation, leading to efficient cell death in animal models. Studies in leukemia and other malignancies, suggest that schedule is important when combining CDK4/6 inhibitors (CDKis) with cytotoxic agents. Based on these observations, we broadened evaluation of two CDKis, palbociclib (PD-0332991, Pfizer) and ribociclib (LEE011, Novartis) in B and T-ALL as single agent and in combination with conventional cytotoxic chemotherapy, using different schedules in preclinical models. As monotherapy, CDKis caused cell cycle arrest with a significant decrease in S phase entry and were active in vivo across a broad number of patient-derived xenograft samples. Prolonged monotherapy induces resistance, for which we identified a potential novel mechanism using transcriptome profiling. Importantly, simultaneous but not sequential treatment of CDKis with conventional chemotherapy (dexamethasone, L-asparaginase and vincristine) led to improved efficacy compared to monotherapy in vivo. We provide novel evidence that combining CDKis and conventional chemotherapy can be safe and effective. These results led to the rational design of a clinical trial.

2015 ◽  
Vol 8 (1) ◽  
Author(s):  
Ilaria Iacobucci ◽  
Andrea Ghelli Luserna Di Rorà ◽  
Maria Vittoria Verga Falzacappa ◽  
Claudio Agostinelli ◽  
Enrico Derenzini ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 860-860
Author(s):  
Inge M. Appel ◽  
Karin M. Kazemier ◽  
Anjo J.P. Veerman ◽  
Elisabeth van Wering ◽  
Monique L. Den Boer ◽  
...  

Abstract L-Asparaginase is an effective drug for treatment of children with acute lymphoblastic leukemia. The effectiveness is generally thought to result from a rapid depletion of asparagine in serum and cells. Several studies have shown that in vitro resistance to this drug is an independent prognostic factor in ALL. We investigated the clinical response of one in vivo dose of 1000 IU/m2 PEG-Asparaginase and its pharmacokinetic and pharmacodynamic effects in children with newly diagnosed ALL before the start of combination chemotherapy. 57 children (36M / 21F) were enrolled in the study: 2 pro B-ALL, 38 common/ pre B-ALL and 17 T-ALL. Genotyping of precursor B-ALL revealed 11 hyperdiploid, 8 TELAML1 positive, 2 BCRABL positive, no MLL rearrangement, 8 normal, 11 others. The clinical response to PEG-Asparaginase on day 0 (5 days after the PEG-Asparaginase infusion) was defined as good when the number of leukemic cells of peripheral blood was < 1 × 109/L, as intermediate when leukemic cells were 1-10 × 109/L, and as poor when leukemic cells were > 10 × 109/L. The in vivo window response was significantly related to immunophenotype and genotype: 26/38 common / pre B-ALL cases, especially those with hyperdiploidy and TELAML1 rearrangement, demonstrated a good clinical response compared to 8/17 T-ALL (p=0.01). Both BCRABL positive ALL cases showed a poor response (p=0.04). A poor in vivo clinical window response was related to in vitro resistance to L-Asparaginase (p=0.02) and both in vitro as well as in vivo response were prognostic factors for long-term event-free survival (Hazard ratio 6.4; p=0.004, and Hazard ratio 3.7; p=0.01, respectively). The L-Asparaginase activity in the serum was >100 IU/L for at least 15 days. The asparagine levels remained below the detection limit of 0.2 mM for at least 26 days with a concomitant rise in serum aspartate and glutamate. These findings confirm that PEG-Asparaginase will yield its pharmacodynamic effects for 2-4 weeks. After administration of one in vivo dose of 1000 IU/m2 PEG-Asparaginase no changes in apoptotic parameters or changes in intracellular levels of twenty amino acids in leukemic cells could be measured, in contradiction to the changes found after in vitro exposure. This may be explained by the rapid removal of apoptotic cells from the circulation in vivo. Otherwise it is possible that in vivo mesenchymal cells from the bone marrow supply leukemic blasts with asparagine in response to treatment with L-Asparaginase. Conclusion: The clinical response to one dose of 1000 IU/m2 PEG-Asparaginase intravenously is related to phenotype and genotype and predicts outcome. These results suggest that children with ALL with a poor clinical response to PEG-Asparaginase might benefit from a more intensive antileukemic therapy.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2927-2927
Author(s):  
David Teachey ◽  
Cecilia Sheen ◽  
Alix Eden Seif ◽  
Valerie I. Brown ◽  
Stephan A. Grupp

Abstract HMG-CoA reductase inhibitors (statins) are commonly used, FDA-approved agents for the treatment of hypercholesterolemia. In addition to reducing serum cholesterol, statins have been shown to have anti-inflammatory properties and anti-proliferative effects on a number of cell types, including lymphocytes. Many of these effects occur because inhibition of HMG-CoA reductase results in depletion of a number of important cellular intermediates required for signal transduction through the AKT, Ras, and Erk pathways. Statins have been demonstrated to have anti-proliferative effects in a number of malignancies, including acute myeloid leukemia using preclinical models; however, there are no published reports evaluating the efficacy of these agents against acute lymphoblastic leukemia. Large population studies have also suggested that statins may reduce the incidence of a number of cancers, furthering interest in the use of these agents in malignancies, especially since the dose-limiting toxicities (hepatitis and rhabdomyolysis) do not overlap with the majority of commonly used cytotoxic agents. We hypothesized that ALL cells are dependent on the function of HMG-CoA reductase for survival. We tested this hypothesis by studying the efficacy of statins against ALL cells. We studied the effects of 5 different compounds (mevastatin, pravastatin, fluvastatin, lovastatin, and simvastatin) against 7 ALL cell lines (4 human and 3 murine). We found that the more potent 2nd generation statins (fluvastatin, lovastatin, and simvastatin) had marked effects on ALL cells, whereas the less potent first generation statins (mevastatin and pravastatin) had less significant effects. We found that fluvastatin, lovastatin, and simvastatin inhibited proliferation of all 7 ALL cell lines using MTT assay (p &lt;0.05). We also found these three statins induced apoptosis, leading to profound cell death in all 7 cell lines (p&lt;0.05) as assessed by flow cytometry for Annenix-V staining and 7-AAD. IC50 dosing for the three agents ranged between 500nM and 5uM depending on the cell line, levels easily obtainable in humans. The most likely explanation for this remarkable, single-agent effect on ALL is that ALL cells are indeed dependent on cholesterol biosynthesis. Other potential explanations include the possibility that ALL cells are dependant on one of the signal transduction pathways affected by targeting HMG-CoA reductase, or the formal possibility of an off-target effect of statins separate from the inhibition of cholesterol biosynthesis. To determine if the effects of the statins were due to a direct effect on HMG-CoA reductase, we treated cells with melavonolactone, the product of conversion of HMG-CoA by HMGCoA reductase. We found that the addition of melavonolactone completely reversed the effects of all statins even at high doses (&gt;10uM) in all cell lines, making the explanation of an off-target, non-HMG-CoA reductase-based mechanism unlikely. Ongoing work includes the testing of statins in NOD/SCID xenograft models of primary human ALL models, assessing the effects of statins on AKT, Ras, and Erk in ALL cells, and assessing the combination of statins with cytotoxic agents in ALL. In conclusion, we found that ALL cells are dependent of HMG-CoA reductase for survival. Since statins are safe and well-tolerated, this class of agents should be further explored in patients with ALL.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3089-3089 ◽  
Author(s):  
Yao-Te Hsieh ◽  
Enzi Jiang ◽  
Carlton Scharman ◽  
Ella Waters ◽  
Eugene Park ◽  
...  

Abstract Abstract 3089 Poster Board III-26 Novel treatment strategies for pediatric acute lymphoblastic leukemia (ALL) have turned a rapidly deadly diagnosis into a highly treatable entity, but we are still failing 25% of our pediatric ALL patients who die of recurrent ALL. Definitive studies have demonstrated that adhesion of leukemia and lymphoma cells to extracellular matrices or stromal cells protects them against the toxicity of cytoreductive chemotherapy drugs. In this context, a specific role for CD49d, a dominant adhesion molecule for normal lymphocytes, was demonstrated for acute myeloid leukemia (AML) and other malignant hematopoietic cells. The finding that CD49d blockade sensitizes AML cells to chemotoxicity may be of therapeutic potential, as is suggested by recent findings for AML cells engrafted in NOD/SCID mice. CD49d is and is similarly expressed on acute lymphoblastic leukemia (ALL) cells, but our knowledge about CD49d adhesion-mediated chemoprotection of B-ALL is limited. We hypothesized whether similar to primary AML blasts, xenografted ALL cells resistant to chemotherapy can be sensitized to chemotherapy by disrupting their CD49d-mediated adhesive interaction with stroma. To test our hypothesis we used as a CD49d inhibitor the humanized anti-human CD49d antibody natalizumab, or Tysabri®, which is in clinical use for the treatment of relapsing or refractory Multiple Sclerosis. To determine the potential of Tysabri as a single agent to decrease leukemia progression, we engrafted 5-7 weeks old NOD/SCID mice with primary drug resistant B-ALL labeled with lentiviral luciferase to allow monitoring of leukemia using noninvasive bioluminescent imaging. Tysabri administered upon detection of engraftment on Day15 post-injection of leukemia in the dose of either 1 mg (n=3) or 6 mg (n=3) led to remarkably slower leukemia progression regardless of the dose compared to the control group treated with saline only (n=2). Additional administration of Tysabri on day 29 and day 37 did not result in further containment of leukemogenesis but still showed a marked reduction in progression compared to the saline treated control group. In addition, we determined in vivo that a weekly administration of Tysabri in the dose of 5mg/kg/d resulted in prolonged survival compared to the treated control (p<0.05). Next, we assessed the effect of adjuvant anti-CD49d antibody-mediated dislodgement of ALL cells of drug resistant patients in combination with chemotherapy. The group treated for 4 weeks with chemotherapy including Vincristine, Dexamethasone and L-Asparaginase (VDL) in combination with Tysabri (5mg/kg/d) admistered once weekly showed decreased progression of leukemia and significantly prolonged survival (p<0.05) compared to the VDL only treated control group. No toxicity of Tysabri treatment was observed. Taken together, our data indicates the potential of Tysabri as a novel adjuvant therapy for treatment of drug resistant B-ALL. Given the availability of clinical-grade CD49d blocking antibody, clinical studies can follow immediately, should our hypothesis be confirmed in further in vitro an in vivo studies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (5) ◽  
pp. 2020-2023 ◽  
Author(s):  
David T. Teachey ◽  
Cecilia Sheen ◽  
Junior Hall ◽  
Theresa Ryan ◽  
Valerie I. Brown ◽  
...  

Abstract We have previously demonstrated that mTOR inhibitors (MTIs) are active in preclinical models of acute lymphoblastic leukemia (ALL). MTIs may increase degradation of cyclin D1, a protein involved in dihydrofolate reductase (DHFR) synthesis. Because resistance to methotrexate may correlate with high DHFR expression, we hypothesized MTIs may increase sensitivity of ALL to methotrexate through decreasing DHFR by increasing turn-over of cyclin D1. We tested this hypothesis using multiple ALL cell lines and nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice xenografted with human ALL. We found MTIs and methotrexate were synergistic in combination in vitro and in vivo. Mice treated with both drugs went into a complete and durable remission whereas single agent treatment caused an initial partial response that ultimately progressed. ALL cells treated with MTIs had markedly decreased expression of DHFR and cyclin D1, providing a novel mechanistic explanation for a combined effect. We found methotrexate and MTIs are an effective and potentially synergistic combination in ALL.


2012 ◽  
Vol 30 (30_suppl) ◽  
pp. 95-95
Author(s):  
Yelena Yuriy Janjigian ◽  
Christopher M. Gromisch ◽  
Gregory Carbonetti ◽  
Laura H. Tang ◽  
David Paul Kelsen ◽  
...  

95 Background: Gastric cancer is a heterogeneous disease that may be subdivided into distinct subtypes—proximal/gastroesophageal (GE) junction, diffuse/signet ring type, and distal gastric cancer/intestinal type—based on histopathologic and anatomic criteria. Each subtype is associated with unique epidemiology and gene expression. Human epidermal growth factor receptor (HER2) is a validated treatment target in gastric cancer. For patients with metastatic disease, the available cytotoxic agents are applied indiscriminately to all disease subtypes, and with only modest success. The purpose of this study is to establish xenograft models from gastric cancer subtypes to improve our understanding of disease heterogeneity and develop therapies geared for each subtype of gastric cancer. Methods: Fresh specimens obtained from resected primary or metastatic tumors under aseptic conditions. 1 g tumor samples injected SQ into flanks of NSG mice. Xenografts established after 5 passages and maintained by serial transplantation into new mice. Cell cultures established after 5 in vitro passages; cell lines after 15 passages Results: To date, 66 tumor samples have been implanted from which 16 xenografts have been established. The table below summarizes the results. Single-agent afatinib (pan-ErbB inhibitor) demonstrated antitumor activity in an HER2-positive xenograft established from MSKCC patient’s tumor harvested from a skin metastasis. Conclusions: We have established xenograft models of gastric cancer. In vivo testing of afatinib showed a reduction of tumor growth of HER2-positive gastric cancer. These models provide a platform to study potential therapeutics for esophagogastric cancer to further validate difference in their biology and guide rational design of clinical trials. [Table: see text]


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3766-3766 ◽  
Author(s):  
Michelle L. Churchman ◽  
Luke Jones ◽  
Kathryn Evans ◽  
Jennifer Richmond ◽  
Irina M Shapiro ◽  
...  

Abstract Introduction: BCR-ABL1+ B-progenitor acute lymphoblastic leukemia (Ph+ B-ALL) is a highly aggressive disease that is often refractory to currently available therapies. Our previous genomic profiling studies have identified loss-of-function or dominant negative mutations in IKZF1, encoding the lymphoid transcription factor Ikaros, in over 80% of Ph+ ALL. In addition, deletion of CDKN2A, which encodes the INK4A and ARF tumor suppressors, is observed in approximately half of all cases (Mullighan et al., 2008). Alterations of IKZF1 are associated with poor outcome despite the use of tyrosine kinase inhibitors (TKIs). Ikzf1 alterations, including Ikaros isoform 6 (IK6), result in the acquisition of stem cell-like features, enhanced self-renewal, expression of adhesion molecules, and transcriptional upregulation of focal adhesion kinase (FAK), resulting in increased adhesion in vitro and in vivo, and decreased sensitivity to TKIs (Churchman, Cancer Cell, in press). VS-4718 is a potent, selective, and orally bioavailable FAK inhibitor currently under evaluation in a phase 1 clinical trial in subjects with various solid tumors, however in vivo efficacy in hematological malignancies had not been evaluated. Targeting FAK with VS-4718 is an attractive approach to abrogate the adhesive phenotype of IKZF1-altered leukemic cells potentially enhancing the effects of dasatinib in the treatment of high-risk BCR-ABL1 B-ALL. Methods: We examined the efficacy and mechanisms of FAK inhibition using VS-4718 as a single agent and in combination with dasatinib in vitro and in vivo in a range of xenograft and genetically engineered mouse models of BCR-ABL1 ALL. Each model had concomitant deletion of Arf which is observed in approximately 50% of human cases. Results: A pre-clinical in vivo trial of dasatinib and VS-4718 combination therapy in a murine C57Bl/6 Arf-/- BCR-ABL1 pre-B cell model resulted in a marked increase in survival in both IK6-expressing and non-IK6 cohorts of mice, and one complete long-term remission in the IK6-expressing group. Further, we showed increased efficacy of VS-4718 and dasatinib, compared to either agent alone, against two highly aggressive human Ph+ IK6-expressing B-ALL xenografts in vivo, with decreased infiltration of leukemic cells in bone marrow and spleens demonstrating a synergistic effect of the VS-4718/dasatinib combination. In vitro cell viability was reduced with induction of apoptosis at increasing concentrations of VS-4718 as a single agent, and further potentiated the effects of dasatinib in cytotoxicity assays using human xenografted and murine leukemic cells. VS-4718 profoundly diminished the ability of BCR-ABL1-expressing cells to form cell-matrix adhesions in vitro, as evident by the reduced adherence to fibronectin monolayers and bone marrow stromal cells. VS-4718 almost completely abolished the colony-forming potential of BCR-ABL1-expressing murine pre-B cells with and without Ikzf1 alterations at drug concentrations that do not affect cell viability suggestive of a reduction in self-renewal. Calvarial imaging of mice transplanted with Ikzf1-altered BCR-ABL1 leukemic cells and treated with VS-4718 alone in vivo revealed a discernible reduction in adhesion in the intact bone marrow niche of Prrx1-Cre; LSL-tdTomato recipient mice. VS-4718 treated leukemic cells localized to Prrx1-expressing perivascular endothelial cells and exhibited round morphology in contrast to the typical spindle-like appearance of Ikzf1-altered pre-B cells adhering to the bone marrow stroma, suggesting that VS-4718 treatment abolished the aberrant leukemic cell-stromal adhesion induced by Ikaros alterations in vivo. Conclusions: Direct inhibition of FAK with VS-4718 attenuates the adhesive, stem-like properties of IKZF1-altered BCR-ABL1 leukemic cells that contribute to the poor prognosis of patients treated with currently available therapies. Targeted FAK inhibition is thus a promising avenue for improving the response of BCR-ABL1 ALL to dasatinib, particularly in refractory cases harboring IKZF1 alterations. These data support the clinical development of VS-4718 in combination with dasatinib in Ph+ B-ALL. Disclosures Shapiro: Verastem: Employment, Equity Ownership. Pachter:Verastem: Employment, Equity Ownership. Weaver:Verastem: Employment, Equity Ownership. Mullighan:Amgen: Honoraria, Speakers Bureau; Cancer Science Institute: Membership on an entity's Board of Directors or advisory committees; Loxo Oncology: Research Funding; Incyte: Consultancy, Honoraria. Off Label Use: The FAK inhibitor VS-4718 for the treatment of BCR-ABL1 acute lymphoblastic leukemia in preclinical models.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 46-46
Author(s):  
Zhongbo Hu ◽  
Michael Sporn ◽  
John Letterio

Background: Pediatric acute lymphoblastic leukemia (ALL) is the most common childhood cancer and the most frequent cause of death from cancer before 20 years of age. Survival rates for patients with relapsed ALL remain less than 50% due to the emergence of chemoresistance, particularly steroid resistance. Several lines of evidence suggest the importance of therapy-related activation of the NLRP3 inflammasome in ALL treatment resistance. The NLRP3 inflammasome has been shown to convey steroid resistance, through epigenetic mechanisms leading to NLRP3 promoter hypomethylation and upregulation of NLRP3 expression, and through inflammasome-dependent modulation of the expression of the glucocorticoid receptor. Objectives: The primary objective of this study is to define the kinetics of activation of the NLRP3 inflammasome in ALL patients following systemic chemotherapy. A secondary objective is to determine whether inhibition of the NLRP3 inflammasome enhances the ALL response to conventional chemotherapy in established, patient-derived xenograft (PDX) preclinical models of pediatric ALL. Specifically, we will investigate whether a newly developed synthetic triterpenoid, CDDO-2P-Im (which we have shown to inhibit activation of NLRP3), can suppress the in vivo growth of steroid-resistant human ALL PDX lines and augment their response to the steroid dexamethasone (DEX), and other conventional chemotherapy agents. Methods: We obtained 1-3 ml of peripheral blood from newly diagnosed ALL patients (0-40 years of age) before and after induction chemotherapy and from normal controls without significant inflammatory or infectious diseases under the UH Hospital Cleveland Medical Center IRB-approved protocol STUDY20190453. Either the Ella Simple Plex protein assay or ELISA were used to determine the expression levels of inflammasome associated proteins such as IL-1β, IL-18, caspase-1 and ASC. Western blot was used to analyze the inflammasome signaling proteins such as casepase-1, IL-1β, IL-18, ASC, NLRP3, AIM2, and LonP1 in the cell lysates. Cell viability assay with Cell-Titer-Glo was performed with different leukemia cell lines, such CEM-1, CEM-7, NALP-3 to determine effects of CDDO-2P-Im and DEX on the leukemia growth. Flow cytometry with Annex-V combined with 7-AAD staining was used for apoptosis assay to detect the effect of CDDO-2P-Im and chemotherapy reagents such as DEX. Leukemia PDX models were established to evaluate the in vivo effects of DEX combined with CDDO-2P-Im. Results: Currently 22 ALL patients and 13 controls were included, with plasma samples from 7 ALL patients. The post chemotherapy expression levels of IL-18, which is one of the important inflammasome proteins in the plasma, are significantly elevated compared with pre-chemotherapy. The expression of NLRP3 mRNA and that of downstream proteins caspase-1 and IL-18 in ALL culture supernatants are upregulated after leukemia cells are exposed to doxorubicin and dexamethasone at 12 and 18 hours. The newly synthesized triterpenoid analog CDDO-2P-Im, which inhibits the activation of NLRP3 inflammasome, inhibited the in vitro growth of ALL cells in culture, induced apoptosis of both steroid-sensitive and steroid-resistant leukemia cells, enhanced the anti-leukemia effect of dexamethasone in steroid-resistant leukemia cells and significantly prolonged the survival of mice bearing human PDX lines. Conclusions: Our preliminary data indicate that chemotherapy activates the NLRP3 inflammasome and downstream signals in patients undergoing therapy for ALL. Importantly, the novel triterpenoid analog, CDDO-2P-Im blocks NLRP3 activation, suppresses ALL viability and exhibits significant single agent activity in steroid-resistant ALL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1432-1432
Author(s):  
Christopher Daniel Chien ◽  
Sang Minh Nguyen ◽  
Haiying Qin ◽  
Elad Jacoby ◽  
Terry J. Fry

Abstract Despite 5-year survival rates nearing 90%, leukemia is the most frequent cause of death from cancer in children with treatment failure primarily caused by relapse after remission. Therefore, it is critical that we identify new therapies that specifically address leukemic persistence during therapy. A cohort of acute lymphoblastic leukemia (ALL) patients that have an increased risk of relapse and consequently poorer overall survival have leukemia that overexpresses cytokine receptor-like factor 2/thymic stromal lymphopoietin receptor (CRLF2/TSLPR). CRLF2/TSLPR overexpressing patients have rates of relapse nearly double the rate of non-overexpressing patients despite comparable remission rates to low-risk patients and account for half of the cases of high risk Philadelphia-like ALL. We hypothesize that TSLP/TSLPR axis may promote relapse of ALL through overexpressed TSLPR, which sensitizes leukemic blasts to low levels of TSLP in bone marrow (BM) niches promoting survival. To test this theory we generated a TSLPR overexpressing syngeneic murine leukemia (TSLPRhigh) through viral transduction of a transplantable pre-B cell ALL line (TSLPRlow). This TSLPRhigh leukemia has comparable TSLPR expression levels to what is found on human ALL that overexpress TSLPR and the transduced TSLPR is functional with increased phosphorylation of STAT5 protein in response to TSLP stimulation. When ALL lines were injected into immunocompetent mice, we observed an 8 fold difference in the percentage of TSLPRhigh vs. TSLPRlow ALL in the BM 5 days after injection, when leukemia accounted for less than 5% of the BM corresponding to an early stage of ALL progression. Interestingly, in vitro and in vivo cell growth and late-stage lethality were no different between the TSLPRhigh and TLSPRlow ALL indicating that TSLP does not alter ALL proliferation. From this data we can infer that TSLP/TSLPR signaling is likely most critical at early stages of leukemia development when BM stromal niches are intact. Hypothesizing that these niches are the source of TSLP ligand we sought to identify the cells responsible for secreting TSLP in the BM. Indeed, basal levels of TSLP mRNA are present in the BM, but the expression is quite low. Interestingly, TSLP mRNA and protein were markedly induced in murine BM stromal cell lines and primary BMSCs by the inflammatory cytokines IL-1a and TNF-a as has previously been reported for other types of TSLP-producing human cells. Since it is well established that cytotoxic chemotherapeutics can mediate an inflammatory response in patients, we investigated whether cytotoxic agents can cause release of inflammatory cytokines from BM cell populations. We treated primary murine BMSCs, BM hematopoietic cells, and ALL lines with low doses of chemotherapy and observed that the pyrimidine analog cytarabine (Ara-C) was particularly potent in upregulating IL-1a expression from BM hematopoietic cells and ALL and not in BMSCs suggesting that inflammatory cytokine release by hematopoietic cells may induce TSLP production by BMSCs. We set out to determine if IL-1a administration to mice could enhance the early progression of ALL in vivo. Indeed, we found that TSLP mRNA and protein were elevated in the BM and serum of mice injected with IL-1a respectively and that there was significant increase in the percentage of early ALL infiltration in the BM of mice bearing TSLPRhigh ALL but not TSLPRlow ALL suggesting a dependence on high levels of expression of TSLPR to respond to IL-1a -induced TSLP production. In addition, we confirmed the necessity of TSLPR-driven early ALL progression on the presence of the TSLP ligand as we observed no significant increase in TSLPRhigh ALL in TSLP deficient mice after stimulation with IL-1a. Furthermore, the early increase in TSLPRhigh leukemia burden can be reversed by using a TSLP blocking antibody demonstrating that targeting the TSLP/TSLPR axis may be therapeutically relevant. These data demonstrate that TSLP secreted in the BM induced by inflammatory cytokines such as IL-1a can drive accelerated early progression of ALL. These inflammatory cytokines can be induced by cytotoxic agents suggesting that chemotherapy can indirectly provide an unintended advantage to TSLPR overexpressing ALL. Finally, we postulate that therapies targeting the TSLP signaling axis would decrease the risk of relapse in TSLPRhigh ALL particularly in the context of standard therapy. Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 530-530
Author(s):  
Jun J. Yang ◽  
Shuyu E ◽  
Shuhong Shen ◽  
Deepa Bhojwani ◽  
Huining Kang ◽  
...  

Abstract Abstract 530 Although cure rates of childhood acute lymphoblastic leukemia (ALL) have improved dramatically, a substantial portion of children still relapse and the prognosis of relapsed ALL is extremely poor. Therefore, a better understanding of molecular determinants of drug resistance in ALL is imperative for the development of more efficacious and individualized therapy, particularly in the context of relapsed disease. In a recent genome-wide association study of 2,534 children with ALL, we identified that genetic variation in PDE4B– phosphodiesterase 4B–strongly influenced the risk of ALL relapse across various ALL treatment regimens (Nat Genet 2011: 43:237). While PDE4B is the predominant phosphodiesterase in lymphoid tissue and a major regulator of cyclic AMP, its role in ALL pathobiology is largely unknown. To this end, we sought to characterize the molecular mechanisms by which PDE4B modulates antileukemic drug sensitivity in ALL. We first characterized PDE4B expression in ALL blasts at diagnosis and its relationship with drug response in vivo in 3 independent cohorts of children with ALL. In 191 children with newly diagnosed ALL enrolled on the COG P9906 protocol, PDE4B expression in ALL blasts was positively correlated with minimal residual disease status at the end of remission induction (P=0.0096). Higher PDE4B expression was also associated with slower early response to induction therapy in COG 1961 (N=82, P=0.019). In 275 children with newly-diagnosed ALL enrolled on the Shanghai Children's Medical Center ALL05 study, we determined that PDE4B2 was the predominant isoform of PDE4B in ALL blasts (P<0.0001); there was also a trend that children with poor in vivo response to the upfront single-agent prednisone treatment had higher PDE4B2 expression in the diagnostic blasts (P=0.042). In parallel, shRNA-mediated knock-down of PDE4B in a glucocorticoid-sensitive (i.e., Nalm6) and a glucocorticoid-resistant (i.e., UOCB1) ALL cell line significantly potentiated cytotoxic effects of prednisolone, whereas Nalm6 and CEM ALL cells over-expressing PDE4B2 were significantly more resistant to prednisolone compared to cells transduced with empty vectors. Sensitization to glucocorticoid was further amplified by forskolin, a stimulator of cAMP synthesis, and was concomitant with activation of PKA as determined by CREB phosphorylation, suggesting that the effects of PDE4B on glucocorticoid sensitivity involve signaling of the cAMP-PKA cascade. Importantly, PDE4B knockdown by shRNA and pharmacologic inhibition by rolipram in Nalm6 and UOCB1 cells consistently led to upregulation of BIM, a key apoptosis regulator and a critical mediator of glucocorticoid sensitivity in lymphoid cells. In both cells lines, BIM upregulation following PDE4B inhibition was enhanced by forskolin but suppressed by PKA inhibitor H89, indicating that BIM might act as a downstream effector of cAMP-PKA signaling in ALL. Finally, we evaluated effects of small molecule regulators of the PDE-cAMP pathway (namely, forskolin and rolipram) on glucocorticoid response in primary ALL cells (N=23) in vitro. Measuring IC50 by MTT assay, increased cytotoxicity was observed in 16 (70%), 8 (35%), and 17 (74%) cases, when forskolin, rolipram, or both were added to prednisolone, respectively. In conclusion, tumor expression of PDE4B at diagnosis was associated with poorer early treatment response in ALL, particularly resistance to glucocorticoids in vivo. Inhibition of PDE4B sensitized both cultured and primary ALL cells to glucocorticoids via activating the cAMP-PKA pathway and subsequent upregulation of BIM. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document