Investigating the Expression of the MRD Marker CD58 on Leukaemia Initiating Cells in Childhood Acute Lymphoblastic Leukaemia

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1887-1887 ◽  
Author(s):  
Charlotte Victoria Cox ◽  
Paraskevi Diamanti ◽  
Allison Blair

Abstract Abstract 1887 Further improvements in outcome for childhood acute lymphoblastic leukaemia (ALL) will require a better understanding of the underlying biology of this disease and the fundamental mechanisms of drug resistance. The discoveries that a few populations can initiate leukemia in mouse models and that new populations of leukaemia initiating cells (LIC) can be detected following an initial round of transplantation in these models raises important questions about the biology of the leukaemias. If several cell populations have LIC properties, what are the relationships of these populations to each other and which populations are most important to target with therapy? It will also be important to determine whether there is any correlation in the biological properties of LIC identified in the model systems with the response of the patients to therapy. Assessment of minimal residual disease (MRD) levels provides a sensitive measurement of early treatment response and permits detection of the in vivo selected drug resistant population. CD58 (leucocyte function-associated antigen 3; LFA-3) is a useful marker in MRD tracking of B cell precursor (BCP) ALL. CD58 is over expressed in these cases permitting discrimination of leukaemia blasts from normal B cells. In this study we investigated whether CD58 is expressed on LIC populations in childhood ALL. Expression of CD58 and CD34 was assessed in a cohort of 12 diagnostic samples with mixed prognoses and compared to levels detected in 11 normal bone marrow (NBM) samples. Levels of CD58 were significantly higher in the ALL cases (57.4±37.7%) than on NBM cells (21.1±12.2%; p=0.007). Likewise, the CD34+/CD58+ population was larger in ALL cases than in normal cells (22.2±34.7% and 0.25±0.25%, respectively; p=0.05). Cells from eight of the 12 patients, were sorted on the basis of expression or lack of expression of these markers and the functional ability of the sorted subpopulations was assessed in vitro and in vivo. On sorting, the majority of cells were CD34−/CD58− (43.7±39.2%), 20.7±30.7% were CD34−/CD58+, 19±14.3% were CD34+/CD58+ and the CD34+/CD58− population accounted for 16.6±35.3%. Unsorted cells and all 4 sorted populations were set up in long-term culture to assess proliferative capability and the in vivo propagating potential was assessed in NSG mice. All 4 sorted subpopulations proliferated over the 6 week period but the highest levels of expansion were observed in the cultures of CD34+/CD58+ (6–420 fold) and CD34+/CD58− (3–24 fold) cells. Cytogenetic analyses confirmed that leukaemia cells were maintained in the culture system. Results from the in vivo analyses on 5 cases to date indicate that all 4 subpopulations contain LIC. In these cases, higher levels of engraftment were observed with CD34+/CD58+ (up to 20%) and with CD34−/CD58− subpopulations (6.1-98%). Serial transplantation studies will determine whether there are differences in the repopulating and self-renewal abilities of these LIC. These findings suggest that using CD58 alone or in combination with CD34 would be insufficient to track disease progression in ALL. Incorporating additional markers that are commonly used in MRD panels will provide valuable information on LIC populations and facilitate development of improved disease monitoring. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2531-2531
Author(s):  
Paraskevi Diamanti ◽  
William J Barendt ◽  
Benjamin C Ede ◽  
Charlotte V. Cox ◽  
Allison Blair

Abstract Current therapies for the treatment of childhood acute lymphoblastic leukaemia (ALL) have resulted in vastly improved survival rates of around 90% in recent years. Despite these successes, around 15% of patients die of relapse. It is possible that ALL may be maintained by subpopulations of cells, known as leukaemia stem cells (LSC), that are resistant to therapy and subsequent relapses may arise from these cells. Parthenolide (PTL), a naturally occurring sesquiterpene lactone, is an NF-κB inhibitor that kills leukaemia cells by apoptosis and/or increase of reactive oxygen species. PTL has been shown to be remarkably effective against several LSC subpopulations in vivo, with complete ablation of leukaemia. In a minority of cases, leukaemia burden was reduced following PTL treatment but not eliminated. Therefore, it may be necessary to combine PTL with other agents to improve killing of all LSC subpopulations. Another pathway of increasing interest in the treatment of leukaemias is the BCL-2 family. BCL-2 has been shown to be overexpressed in over 66% of B-ALL cases and is associated with tumourigenesis in several cancers. ABT-263 is an inhibitor of BCL-2, BCL-xL and BCL-w, it has been shown to selectively target AML LSC and is in early clinical trials in lymphoid malignancies. ABT-199 is another promising inhibitor that is currently in clinical trials for CLL. ABT-199 is specific for BCL-2 and has minimal risk for thrombocytopenia. In the present study the effects of both ABT-263 and ABT-199 alone or in combination with PTL were assessed in childhood ALL samples to determine whether toxicity to leukaemia cells could be improved in vitro and in vivo. The viability of bulk cells from 11 B cell precursor (BCP) ALL cases and 11 cord blood (CB) samples following drug treatment for 24 hours were assessed using flow cytometry by staining with Annexin V and propidium iodide. Initially, PTL was used at a range of 1 to 10μM, ABT-263 from 0.025 to 1μM and ABT-199 from 0.1 to 10μM. Only PTL and ABT-263 significantly reduced the viability of ALL cells compared to CB with IC50 values of 1.2μM and 0.125μM (P≤0.01 and P≤0.0015), respectively. In vitro drug combination studies demonstrated synergism when combining PTL with ABT-263 in a 9.5:1 ratio using the Chou Talalay model. The viability of ALL cells following combination therapy (1.2μM PTL with 0.125μM ABT-263) was reduced to 38.3±32.5%, while CB viability was unaffected (96.9±29%, P<0.0001). Using this combined dose, toxicity to ALL cells was increased by a further 35% compared to PTL alone and by 25% compared to ABT-263 alone. Even at the highest combined doses tested (9.6μM PTL: 1μM ABT-263) normal CB remained relatively unaffected with 73.3±25% surviving. The effects of these drugs alone and in combination were also assessed in LSC subpopulations in 3 of these cases. Unsorted ALL cells, CD34+/CD19+ and CD34-/CD19+ subpopulations were the most responsive with viabilities ranging from 17.6±4% to 23.9±11% using 1.2μM PTL and 0.125μM ABT-263. The CD34+/CD19- and CD34-/CD19- cells were more resistant with 70.3±40% and 73.3±15% surviving, respectively. However, since we have previously shown that the effects of in vitro drug treatment do not always accurately reflect the response in vivo, it was important to evaluate the effects of these drugs in mice with established leukaemia. NOD/LtSz-scid IL-2Rγc null (NSG) mice were inoculated with 1-1.15x106 unsorted BCP-ALL cells. Once the levels of leukaemia engraftment in murine peripheral blood reached ≥ 0.1%, mice were treated with 100mg/kg ABT-263 or ABT-199 and vehicle by oral gavage for 21 consecutive days and the levels of leukaemia burden were monitored weekly. Results to date demonstrate that leukaemia levels continued to rise in placebo-treated mice, reaching 49.2±7% by day 21, while the levels in ABT-263 and ABT-199 treated mice were significantly lower at 8.6±10% and 23.7±12%, respectively (P<0.0001). The use of ABT-263 and ABT-199 significantly improved the survival of NSG compared to untreated controls (P=0.0001). These data indicate that combining PTL with ABT-263 shows promising results in the killing of bulk and LSC populations in BCP-ALL. Ongoing in vivo studies will assess the potential of using BCL-2 inhibitors in combination with PTL compared to standard chemotherapeutics. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2952-2952
Author(s):  
Paraskevi Diamanti ◽  
Charlotte Victoria Cox ◽  
Allison Blair

Abstract Abstract 2952 Enhanced risk stratification protocols and intensified therapies have improved outcomes and reduced the risk of relapse in childhood acute lymphoblastic leukaemia (ALL). Nevertheless, 20% of patients relapse, due to failure to eradicate the disease. Current chemotherapeutic regimens are designed around the properties of the bulk leukaemia cells, which might differ from those of the leukaemia initiating cell populations (LIC). Therefore, if drugs have no effect on LIC, these cells may expand and eventually cause relapse. Since several populations in childhood ALL have been shown to have LIC properties, developing therapies that are effective against all LIC populations may prevent relapse. We have previously shown that the NF-κB inhibitor parthenolide (PTL) could prevent proliferation and engraftment of unsorted cells and all LIC populations in NSG mice, in most cases examined. Heat shock protein (Hsp) 90 inhibitors are promising targets in cancer therapy. Targeting this protein has a combined impact on many oncogenic pathways involved in malignant progression. In order to investigate whether the ablation of LIC that we observed using PTL could be improved, we examined the effects of two Hsp90 inhibitors on childhood ALL cells in this study. 17-DMAG is used in preclinical and clinical phase I and II testing in breast cancers. It targets the binding site of ATP in Hsp90 and has low toxicity and high oral bioavailability. Celastrol is a novel Hsp90 inhibitor, which has recently been shown to eradicate LIC in AML by inhibiting NF-κB survival signals and inducing oxidative stress. Dose-response effects using 0.01–100nM of each Hsp90 inhibitor were assessed in primary T- and B-ALL cases and on normal haemopoietic cells at 24, 48 and 72h. Cells were stained with Annexin V and PI then viability and apoptosis were assessed by flow cytometry. An IC50 was observed in leukaemia samples using 1nM of both 17-DMAG and Celastrol after 72h. Increasing the dose of 17-DMAG to 100nM and reducing the incubation time to 48 hours for each drug further reduced ALL cell survival, without impacting normal cells. At these doses, 17-DMAG reduced the viability of T-ALL cells to 36±30% and B-ALL cells to 32±13%. In T-ALL cases, 43±15% survived treatment with Celastrol and similar results were observed with B-ALL cells (43±16%). Normal haemopoietic cells were relatively unaffected at these doses (17-DMAG: 81±2% and Celastrol: 86±36%). Subsequently, T-ALL cells were sorted for expression of CD34 and CD7 and B-ALL cells were sorted for CD34 and CD19. The subpopulations were treated with 1nM of each inhibitor and the results compared to those observed using untreated controls. However, at these concentrations the drugs had limited effects on the ALL subpopulations; 31–100% and 28–89% T-ALL subpopulations survived treatment with 17-DMAG and Celastrol respectively. Similar results were observed with B-ALL subpopulations, 9–86% survived treatment with 17-DMAG and 62–100% survived following Celastrol treatment. A number of studies have shown that a regulatory loop may exist between Hsp90 and NF-κB in that downregulation of NF-κB could lead to reduction in Hsp90 protein levels. Therefore, we investigated whether using the Hsp90 inhibitors in combination with a NF-κB inhibitor would be more effective. Samples were treated with 100nM 17-DMAG or 1nM Celastrol for 48h and 7.5mM PTL was added for the last 24 hours. In 3 T-ALL cases, PTL reduced the viability to 28±13%, 17-DMAG to 25±12% and Celastrol to 35±15%. When PTL was used in combination, cell survival was further reduced to only 18±9% (PTL + 17-DMAG) and to 19±10% (PTL + Celastrol). In 3 B-ALL cases, PTL treatment reduced viability to 57±9%, similar results were seen with 17-DMAG (59±6%), while Celastrol appeared to be the most effective of the 3 drugs (38±4%, P=0.02). When PTL was combined with the Hsp90 inhibitors the cell killing was increased by 2 fold compared to PTL or 17-DMAG alone (PTL + 17-DMAG: 31±6%, P=0.04 and PTL + Celastrol: 28±3% P=0.01). Results to date indicate a promising role for the use of Hsp90 inhibitors with PTL and data on the functional ability of treated LIC populations will provide further information on the potential of these drug combinations. In conclusion, these Hsp90 inhibitors were as effective as PTL against childhood leukaemia cells and when used in combination with PTL, cell survival was further reduced by up to 20%. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 28 (6) ◽  
pp. 1341-1346 ◽  
Author(s):  
Elizabeth C. Matheson ◽  
Linda A. Hogarth ◽  
Marian C. Case ◽  
Julie A.E. Irving ◽  
Andrew G. Hall

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3304-3304 ◽  
Author(s):  
Mark Wunderlich ◽  
Fu-Sheng Chou ◽  
Mahesh Shrestha ◽  
Benjamin Mizukawa ◽  
James C. Mulloy

Abstract Abstract 3304 Although significant progress has been made in the treatment of leukemia, relapse continues to be a major problem, particularly in acute myeloid leukemia (AML). The prognosis for relapsed leukemia is poor, indicating an area for potential improvements. However, animal models to study the response of human AML to chemotherapeutics and subsequent relapse are lacking. Recently we developed an improved NOD/SCID mouse with IL2RG knockout and transgenic expression of myelo-supportive cytokines SCF, GM-CSF, and IL-3 (the NSGS mouse). This mouse is remarkable in its ability to accept human AML grafts more efficiently than all other available strains. When coupled with in vitro derived AML cells, the NSGS mouse allows for a more predictable AML model with shorter latency and smaller range of death than in other mouse strains, including NSG mice. Importantly, very low numbers of cells reliably generate fatal AML in roughly 40 days, even in non-irradiated NSGS mice, allowing for rapid experimental conclusions and reduced toxicity. With the benefits of these unique tools, we sought to develop a model system to evaluate the efficacy of chemotherapeutic agents on human AML cells in vivo. Engrafted mice received a chemotherapy regimen over a 5-day treatment period consisting of a daily dose of cytarabine with simultaneous injection of doxorubicin during the first three days. Treated mice experienced striking weight loss during the treatment period with a nadir at days 8–10 post-treatment. Mice recovered body weight within 3 weeks. Serial complete blood counts indicated a rapid transient drop in total white blood cell and neutrophil counts and a delayed transient drop in red blood cell and platelet numbers, reminiscent of the effects observed in patients undergoing chemotherapy. The drugs successfully targeted the cells of the bone marrow, as evidenced by a profound loss of cellularity in treated mice relative to controls. When mice harboring N-Ras(G12D) positive AML cells were treated at early time points post-transplant, a significant reduction of tumor burden was observed in the BM and PB, with the grafts of treated mice essentially undetectable for weeks after treatment cessation. Nevertheless, treated mice inevitably succumbed to disease, although with a significantly prolonged latency compared to mock treated mice. However, when AML cells containing the FLT3-ITD mutation were used, a shift in disease latency was not reproducibly seen. This data correlates well with patient data showing that FLT3-ITD mutant AML has a worse prognosis than AML samples with N-Ras mutations. Importantly, the reappearance of AML within weeks of treatment affords the opportunity to model drug resistance and relapse, as well as the potential synergistic effects of experimental compounds used in combination with traditional chemotherapy. Additionally, the period following treatment may allow for studies of minimal residual disease as well as the testing of potential maintenance therapies. Finally, this approach permits a detailed analysis of the critical few cancer stem cells that remain after induction therapy with the goal of identifying novel compounds capable of targeting these cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1351-1351
Author(s):  
Eleanor L Woodward ◽  
Martin L. Olsson ◽  
Bertil Johansson ◽  
Kajsa Paulsson

Abstract Whilst a great deal is known about acquired somatic aberrations associated with the diagnosis and prognosis of acute leukaemias, relatively little is known about the effects of germline variation. A recent study reported a link between germline variations in the zinc finger (ZnF) binding domain array of the PR domain-containing 9 (PRDM9) gene, regulator of meiotic recombination, with the development of acute lymphoblastic leukaemia (ALL) in children. An excess of rare PRDM9 alleles that affect meiotic recombination events were seen in parents of children affected with B-cell precursor ALL (pre-B-ALL), and in an independent cohort of children with pre-B-ALL; in particular in aneuploid and infant ALL. In the present study, we carried out Sanger sequencing to investigate variation of PRDM9 alleles in a cohort of parents (n=59) with children diagnosed with high hyperdiploid ALL (HeH; 51-67 chromosomes) (n=31) and a control cohort of individuals (n=66) from the south of Sweden. Whereas similar numbers of rare PRDM9 alleles were observed in both groups, a larger number of rare alleles that affect recombination events were observed in the parents from the family cohort with children diagnosed with HeH ALL compared to individuals of the population control cohort (15.3% vs. 4.5%, P=0.0414). Two-thirds of the parents transmitted the rare allele to their affected children. A previously unreported ZnF repeat was also detected in two individuals of the control cohort but it was not detected in the family cohort. A statistically significant difference in frequency of rare alleles affecting recombination events between the two groups indicates a true association between PRDM9 allelic forms and HeH ALL in an independent cohort. Thus, our results confirm the previous findings that PRDM9 may play a role in the development of childhood ALL. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document