Preclinical Evaluation of CBP/β-catenin Inhibition as a New Strategy for Drug Resistant Acute Lymphoblastic Leukemia.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1596-1596
Author(s):  
Yong-Mi Kim ◽  
Eugene Park ◽  
Colin Lorentzen ◽  
Brian De La Torre ◽  
Yao-Te Hsieh ◽  
...  

Abstract Despite advances in chemotherapeutic treatment of acute lymphoblastic leukemia (ALL), 20% of children relapse with high death rates, so that new treatment modalities are needed. Recent studies have demonstrated that survivin, a member of the inhibitor of apoptosis (IAP) family proteins, is upregulated in ALL of relapsed patients but not in drug-sensitive ALL. The expression of survivin depends on the formation of a complex between β-catenin and its co-activator CBP. Selective suppression of CBP/β-catenin signaling using the novel small-molecule inhibitor ICG-001 offers the opportunity to sensitize leukemia cells to conventional treatment. We hypothesize that inhibition of CBP/β-catenin signaling by combining ICG-001 with conventional therapy represents a promising therapeutic principle to eradicate drug resistant ALL. To test this hypothesis, we used a NOD/SCID xenograft model engrafted with drug-resistant human pre-B ALL leukemia cells (1x106 cells/mouse) to first model the outcome of the patient in vivo. When human CD45 engraftment of 1% was detected by flow cytometry on day 26 post-leukemia-injection, VDL (Vincristine, Dexamethasone, L-Asparaginase) (n=7) or with saline as control (n=7) was administered for 4 weeks intraperitoneally (i.p.). Without treatment, all mice died between days 31–38 post-treatment with a median survival time (MST) of 36 days. In contrast, one animal of the VDL group died at day 14 post-treatment, the remaining 6 mice between days 67–77 post-treatment (MST=70 days, p<0.05 compared to control group), demonstrating that our xenograft model can mirror the outcome of the patient. Next, we tested whether ICG-001 in combination with standard chemotherapy can improve survival of mice engrafted with the resistant human pre-B ALL cells (1.5x106 cells/mouse). Leukemic animals were treated i.p. with a combination of VDL and ICG-001 (25mg/kg/d) (n=3) or with VDL only as a control (n=2). The animals in the control group died on day 18 and 62 post-treatment (MST=40). In marked contrast, the animals treated with a combination of VDL+ICG-001 died on day 71, 72, 77 post-treatment (MST =72 days, p<0.05 compared to VDL group). Blood count analysis did not show side effects of ICG-001 on hematopoietic cells. We next determined the effect of ICG-001 on the expression of survivin by real-time (RT) PCR in recipients of human relapse T-ALL. Survivin mRNA expression was found to be downregulated after VPL+ICG treatment compared to treatment with VPL only. A greater number of animals and a higher dose of ICG-001 with optimized delivery via osmotic pump are being evaluated. Although limited by the small numbers of mice studied, the sustained survival of the mice treated with combination of standard chemotherapy and ICG-001 is compatible with the hypothesis that ICG-001 can sensitize drug resistant leukemia cells to treatment with standard chemotherapy and may lead to novel therapeutic options to overcome drug resistance.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2922-2922
Author(s):  
Enzi Jiang ◽  
Min Yu ◽  
Yao-Te Hsieh ◽  
Brian DeLaTorre ◽  
Asha Kadavallore ◽  
...  

Abstract Despite advances in chemotherapeutic treatment of acute lymphoblastic leukemia (ALL), 20% of children relapse with high death rates, and adults show a long-term survival of only 60%, so new treatment modalities are needed. AMD3100 is being tested in clinical trails for use as a mobilizing agent for hematopoieitic stem cells and blocks the interaction of CXCL12 (SDF-1a) and its receptor, CXCR4, which are involved in retaining hematopoietic cells in primary lymphoid organs. CXCR4 expressed on bone marrow fibroblasts is involved in retaining pre-B cells in the bone marrow. Blocking the CXCL12 – CXCR4 interaction makes murine acute lymphoblastic leukemia cells more sensitive to drug treatment in vitro. Therefore, we hypothesized, that AMD3100 may sensitize drug resistant leukemia cells to chemotherapy. To test this hypothesis, we established a pre-clinical xenograft model of B-cell precursor-ALL, allowing us to monitor progression of leukemia in vivo with non-invasive bioimaging. Frozen samples of drug resistant adult patients, who did not respond to chemotherapy (drug-resistant Philadelphia chromosome (Ph) positive (Ph+) or negative (Ph−) pre-B ALL leukemia cells), were engrafted via tail-vein injections into female NOD/SCID or NOD/SCID IL2Rγ−/− mice. Primary passages were serially expanded up to three passages. Analysis of the phenotype by flow cytometry and morphology by histochemistry of the xenografts confirmed that the initial characteristics of the patient were retained or not altered throughout the passages. In an effort to sensitize those drug resistant leukemia cells to chemotherapy, we have next proceeded to test AMD3100 in our established preclinical models. Primary ALL cells of an Imatinib-resistant Ph+ patient were transduced with luciferase and injected into sublethally irradiated NOD/SCIDIL2Rγ −/− mice (0.7×106 cells/recipient). Mice were treated per os with saline (n=3), Imatinib (n=4;75 mg/kg/d), AMD3100 via an osmotic pump (n=4;10 mg/kg/d) or Imatinib plus AMD3100 (n=8) for 28 days, after detection of engraftment by bioluminescent imaging. Survival of the group treated with Imatinib + AMD3100 (Median survival time, MST=49 days) was significantly prolonged compared to the group treated with Imatinib only (MST=38 days) (p<0.05). Next, primary ALL cells of a Ph− patient were transduced with luciferase and injected into NOD/SCID mice (0.4×106 cells/animal). Upon detection of engraftment by bioimaging, xenografted mice were treated with saline (n=2), vincristine-dexamethasone-L-asparaginase (VDL; n=3), AMD3100 (n=3) or AMD3100 plus VDL (n=6). Again, significantly prolonged survival of the group treated with the combination of VDL + AMD3100 (MST=61.5 days) compared to the VDL only treated group (MST=54 days) (p<0.05) was observed. In summary, there was a clear beneficial effect of the combination of AMD3100 with two different chemotherapeutic regimens resulting in a significant improvement of survival. Further preclinical evaluation of AMD3100 as a novel adjuvant has the potential to lead to translation into clinical trials.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3072-3072
Author(s):  
Enzi Jiang ◽  
Eugene Park ◽  
Carlton Scharman ◽  
Yao-Te Hsieh ◽  
Asha Kadavallore ◽  
...  

Abstract Abstract 3072 Poster Board III-9 Despite advances in chemotherapeutic treatment of acute lymphoblastic leukemia (ALL), 20% of children relapse with high death rates, highlighting the need for new treatment modalities. Recent population studies have demonstrated that Survivin, a member of the inhibitor of apoptosis (IAP) family proteins, is expressed in most cancerous cells but has also been implicated in normal erythropoiesis. It is upregulated in ALL of relapsed patients but not in drug-sensitive ALL. The expression of Survivin depends on the formation of a complex between β-catenin and its co-activator CBP. Selective suppression of CBP/β-catenin signaling using the novel small-molecule inhibitor ICG-001 offers a novel mechanism to target Survivin in the sensitization of leukemia cells to conventional drug treatment. We hypothesize that inhibition of CBP/β-catenin signaling by ICG-001 in combination with conventional therapy represents a promising therapeutic principle to eradicate drug resistant ALL while sparing normal hematopoiesis. An in vivo study utilized our bioluminescent model to non-invasively monitor leukemogenesis of a primary ALL, transduced with a lentiviral construct encoding firefly luciferase prior to xenotransplantation. NOD/SCIDIL2R gamma-/- mice were sublethally irradiated prior intravenous injection of 50,000 cells per animal. Leukemic animals were treated with a combination of intraperitoneally administered VDL and ICG-001 (100mg/kg/d) (n=3), which was delivered via subcutaneous osmotic pumps to ensure stable plasma levels, with VDL only (n=4), or PBS only (n=2) as a control for 4 weeks. Bioluminescent imaging on Day 42 post-injection showed a contrast in the containment of leukemia of ICG-001+VDL mice as compared to those of the VDL control group. The animals in the PBS control group and the VDL+PBS Pump control groups had Median Survival Times (MST) of 35 days and 66.5 days post-treatment, respectively. In marked contrast, the animals treated with a combination of VDL+ICG-001 had a significant 14% extension in MST of 76 days post-treatment (p=0.016 compared to VDL group). Survivin mRNA expression was found to be downregulated after VDL+ICG treatment compared to treatment with VDL only. Analysis of peripheral blood showed no effect of ICG-001 on leukocyte or red blood cells compared to control groups. Next, we determined in vitro the ability of ICG-001 to increase sensitivity of patient-derived ALL cells and ALL celllines including BEL-1, REH, 697 and SUPB15 to chemotherapy including VDL or Imatinib. After 4 days we observed significantly increased toxicity assessed by MTT assay and AnnexinV staining as well as downregulation of Survivin confirmed by real-time PCR and Western Blot. To determine if ICG-001 is non-toxic to normal hematopoiesis, we treated normalC57BL/6 mice for 3 weeks with ICG-001 only. At end of treatment, normal blood counts including red blood cell, white blood cells and platelets, normal histology and normal weight gain indicated that ICG-001 is not detrimental to the recipient. In vitro apoptotic studies using normal white blood cells isolated from peripheral blood and co-cultured with a stromal layer confirmed further the non-toxicity of ICG-001 to normal cells. In summary, the sustained survival of the mice treated with combination of standard chemotherapy and ICG-001 is compatible with our hypothesis that ICG-001 can sensitize drug resistant leukemia cells to treatment with standard chemotherapy while sparing normal hematopoiesis and may lead to novel therapeutic options to overcome drug resistance. Disclosures No relevant conflicts of interest to declare.


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 ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 453-453
Author(s):  
Gianluigi Zaza ◽  
Meyling Cheok ◽  
Wenjian Yang ◽  
Pei Deqing ◽  
Cheng Cheng ◽  
...  

Abstract Thioguanine nucleotides (TGN) are considered the principal active metabolites exerting the antileukemic effects of mercaptopurine (MP). Numerous clinical studies have reported substantial inter-patient variability in intracellular TGN concentrations during continuation therapy of acute lymphoblastic leukemia (ALL). To identify genes whose expression is related to the intracellular accumulation of TGN in leukemia cells after in vivo treatment with MP alone (MP) or in combination with MTX (MP+MTX), we used oligonucleotide microarrays (Affymetrixâ HG-U95Av2) to analyze the expression of approximately 9,670 genes in bone marrow leukemic blasts obtained at diagnosis from 82 children with ALL. TGN levels were determined in bone marrow aspirates of these patients 20 hours after mercaptopurine infusion (1 g/m2 I.V). Because, as previously reported, patients treated with MP alone achieved higher levels of intracellular TGN compared to those treated with the combination, we used Spearman’s rank correlation to identify genes associated with TGN levels separately for the 33 patients treated with MP alone and the 49 with the combination (MP: median TGN: 2.46 pmol/5x106 cells, range: 0.01–19.98; and MTX+MP: median TGN: 0.55 pmol/5x106 cells, range: 0.005–3.31). Hierarchical clustering using these selected probe sets clearly separated the 33 patients treated with MP alone into two major groups according to TGN concentration (< 2.46 and > 2.46 pmol/5x106 cells; n=60 genes) and two major branches were also found for patients treated with the combination (< 0.55 and > 0.55 pmol/5x106 cells; n=75 genes). Interestingly, there was no overlap between the two sets of genes, indicating that different genes influence the accumulation of TGN when this drug is given alone or in combination with MTX. The association between gene expression profiles and TGN levels determined by leave-one-out cross-validation using support vector machine (SVM) based on Spearman correlation, was rho=0.60 (p<0.001) for MP alone and rho=0.65 (p<0.001) for MTX+MP, with false discovery rate (FDR) computed using Storey’s q-value (MP: 50% true positive, MTX+MP: 70% true positive respectively). Genes highly associated with the post-treatment TGN level in ALL patients treated with MP alone encode transporters, enzymes involved in the MP metabolic pathway and cell proliferation. Genes associated with post-treatment levels of TGN after combined therapy have been implicated in protein and ATP biosynthesis. Together, these in vivo data provide new insights into the basis of inter-patient differences in TGN accumulation in ALL cells, revealing significant differences between treatment with MP alone or in combination with MTX.


2011 ◽  
Vol 35 (6) ◽  
pp. 800-807 ◽  
Author(s):  
Johan Jansson ◽  
Yu-Chiao Hsu ◽  
Igor I. Kuzin ◽  
Andrew Campbell ◽  
Craig A. Mullen

Blood ◽  
2015 ◽  
Vol 125 (2) ◽  
pp. 273-283 ◽  
Author(s):  
Duohui Jing ◽  
Vivek A. Bhadri ◽  
Dominik Beck ◽  
Julie A. I. Thoms ◽  
Nurul A. Yakob ◽  
...  

Key Points The glucocorticoid receptor coordinately regulates the antiapoptotic BCL2 and proapoptotic BIM genes in pediatric ALL cells in vivo. GR binding at a novel intronic region is associated with BIM transcription and dexamethasone sensitivity in pediatric ALL cells in vivo.


1984 ◽  
Vol 8 (4) ◽  
pp. 587-595 ◽  
Author(s):  
Peter Dörmer ◽  
Giovanni Ucci ◽  
Barbara Lau ◽  
Rainer J. Haas ◽  
Gritta E. Janka

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2765-2765 ◽  
Author(s):  
Yongsheng Ruan ◽  
Eun Ji Gang ◽  
Hye-Na Kim ◽  
Chintan Parekh ◽  
Hisham Abdel-Azim ◽  
...  

Abstract Background. Even though remarkable progress has been made in the treatment of childhood acute lymphoblastic leukemia (ALL), salvage of relapse patients remains a challenge. The role of the bone marrow (BM) microenvironment is critical to protect leukemia cells from chemotherapy. The BM microenvironment promotes cell adhesion-mediated drug resistance (CAM-DR) in ALL.We and others have shown that the adhesion molecule integrin α4, referred to hereafter as α4, mediates drug resistance of B-ALL. In our previous studies, we showed that both α4 blockade by natalizumab and inhibition by the small molecule α4 antagonist TBC3486 can sensitize relapsed ALL cells to chemotherapy. However, no α4 targeting therapy is currently clinically available to treat leukemia. Here, we preclinically evaluate a novel non-peptidic small molecule antagonist, AVA4746, which has been safely used in clinical studies, as a potential new approach to combat drug resistant ALL. Method. Six refractory or relapsed primary pre-B ALL cases were used for in vitro studies. Viability was assessed by trypan blue counts or annexin V/7AAD flow cytometric analysis and metabolic activity was evaluated by Cytoscan WST-1 assay. For in vivo evaluation a NOD/SCID IL2Rγ-/- xenograft model of primary pre-B ALL (LAX7R) was used.AVA4746 (15mg/kg) was administered by oral gavage twice a day continuously for 14 days, and vincristine, dexamethasone, L-asparaginase (VDL) was given intraperitoneally (weekly) for 4 weeks. Overall survival was determined by Kaplan-Meier Survival analysis. Results. AVA4746 caused a significant decrease in mean fluorescence intensity (MFI) of α4 expression in six out of six ALL cases at doses of both 5μM and 25μM after 24 hours and 96 hours compared to DMSO control. Interestingly, decreased protein expression of α4 was also observed by Western Blot analysis all six ALL cases. We tested next in two cases (LAX53, ICN13), if AVA4746 de-adheres ALL cells from its counter receptor VCAM-1. The percentages of adherence after treatment with AVA4746 (25μM) were significantly lower than after DMSO treatment (10.3%±4.9% vs. 99.9%±7.6%, p= 0.00007 for LAX7R; 8.1%±1.0% vs. 100.1%±13.6%, p= 0.0003 for LAX53; 9.0%±1.6% vs. 100.0%±14.0%, p=0.0004 for ICN13). AVA4746 was not associated with apoptosis in vitro alone or in combination with chemotherapy (VDL). Metabolic activity as assessed by WST-1 assay was markedly decreased by AVA4746 in two of two ALL cases. AVA4746 also decreased ALL proliferation in two out of two ALL samples tested. In vivo, AVA4746 in combination with VDL chemotherapy treatment led to significant prolongation of overall survival (n=6) compared with the VDL only treated group (n=6) (MST= 78.5 days vs MST= 68 days; P<0.05). There was no significant difference in survival between the PBS control group (n=5) and the AVA4746 mono-treatment group (n=5) (MST=38days vs MST= 38days). Conclusion. We have identified α4 as a central adhesion molecule in CAM-DR of ALL and have shown that AVA-4746, an orally available and specific α4 antagonist, which has been safely used in clinical studies, downregulates α4 in primary ALL and functionally de-adheres them from VCAM-1. Critically, we demonstrated that inhibition of α4 in combination with standard chemotherapy can prolong the survival of NSG mice bearing pre-B ALL. These data support further study of inhibition of α4 using AVA4746 as a novel strategy to treat drug resistant B lineage ALL. Disclosures Bhojwani: Amgen: Other: Blinatumumab global pediatric advisory board 2015. Wayne:Spectrum Pharmaceuticals: Honoraria, Other: Travel Support, Research Funding; Kite Pharma: Honoraria, Other: Travel support, Research Funding; Pfizer: Consultancy, Honoraria, Other: Travel Support; Medimmune: Honoraria, Other: Travel Support, Research Funding; NIH: Patents & Royalties. Kim:Antisense Therapeutics Ltd: Patents & Royalties.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1522-1522
Author(s):  
Ksenia Matlawska-Wasowska ◽  
Dennis Cook ◽  
Samuel R. Stevens ◽  
Elizabeth K. Ward ◽  
Ronald Herbst ◽  
...  

Abstract Abstract 1522 Precursor-B acute lymphoblastic leukemia (pre-B ALL) is the most common malignancy in children and can be cured in a majority of patients. However, cure remains elusive in approximately 20% of patients for reasons that are not well understood. Importantly, survivors commonly develop morbidities that result from dose-intensified treatment with cytotoxic drugs. Here, we investigate the tumoricidal effects of a novel humanized anti-CD19 monoclonal antibody (Medi-551). The a-fucosylated form of this antibody has increased affinity to human FcgammaRIII (CD16) receptor, present on the surface of NK cells and macrophages, mediating antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). Medi-551/CD19 complexes internalize slowly and thus remain accessible for effector cells for prolonged periods. We evaluated in vitro ADCC and ADCP activities of primary human NK cells and macrophages (effector cells) against four pre-B ALL cell lines (697, Nalm 6, MHH-Call 3, RS 4;11), as well as freshly isolated patient blasts. We report results of live cell fluorescent imaging studies, characterizing the formation of immunological synapses between Medi 551-bound target leukemia cells and effector cells, as well as the kinetics of both NK-mediated killing and macrophage phagocytosis. The number of the CD19 receptors present on the cell surface is shown to be a factor in effector-mediated killing of Medi-551 targeted leukemia cells. Further, genetic polymorphisms in FcgammaRIII (158 F/V, V/V or F/F) affected in vitro ADCC and ADCP activities with FcgammaRIII 158 V homo- or heterozygotes showing the strongest activity. We also evaluated the efficacy of Medi-551 in a human pre-B ALL murine xenograft model. SCID mice were engrafted with 697 pre-B ALL cells and received either vehicle alone or Medi-551 (3 mg/kg; twice weekly for a total of 5 doses); treatment was started at day 5 after engraftment. Medi-551 treatment markedly lowered disease burden in blood, liver and bone marrow. The lack of cure is consistent with impaired roles for NK cells in this model, since murine NK cells lack FcgammaRIV. Experiments are in progress to improve the model through adoptive transfer of human NK cells. Taken together, the in vitro and in vivo data show that Medi-551 has strong activity against pre-B ALL and support a move forward to early phase trials in this disease. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document