Myelodysplasia and Acute Myeloid Leukemia during Treatment of Chronic Myeloid Leukemia with Dasatinib

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4284-4284
Author(s):  
Monika Conchon ◽  
Israel Bendit ◽  
Patricia Ferreira ◽  
Lucia Dias ◽  
Cristina Kumeda ◽  
...  

Abstract The incidence and significance of Philadelphia negative aberrant clones in patients treated with the second generation of tyrosine kinase inhibitors, including dasatinib, are infrequent. Here we report 2 patients with myelodysplastic syndrome associated with monosomy 7 in Philadelphia negative cells during consecutive imatininb and dasatinb therapy. No previous exposure to chemotherapeutic agents was reported in both patients. In case 1, retrospective assessment of the bone marrow sample collected prior to treatment with dasatinib did not show any abnormal clone, which became evident only after dasatinib was commenced. In case 2, monsomy 7 were found already in 15% Philadelphia negative cells during imatinib treatment and persisted during dasatinib therapy. In the 2 cases described, we assumed that the consecutive imatinib/dasatinib therapy, could abrogate the proliferation of the Philadelphia positive clone and allow pre-existing monosomy 7 clone to emerge.

2017 ◽  
Vol 2017 ◽  
pp. 1-6 ◽  
Author(s):  
Yasuhiro Tanaka ◽  
Atsushi Tanaka ◽  
Akiko Hashimoto ◽  
Kumiko Hayashi ◽  
Isaku Shinzato

Myelodysplastic syndrome (MDS) terminally transforms to acute myeloid leukemia (AML) or bone marrow failure syndrome, but acute myeloid leukemia with basophilic differentiation has been rarely reported. An 81-year-old man was referred to our department for further examination of intermittent fever and normocytic anemia during immunosuppressive treatment. Chromosomal analysis showed additional abnormalities involving chromosome 7. He was diagnosed as having MDS. At the time of diagnosis, basophils had not proliferated in the bone marrow. However, his anemia and thrombocytopenia rapidly worsened with the appearance of peripheral basophilia three months later. He was diagnosed as having AML with basophilic differentiation transformed from MDS. At that time, monosomy 7 was detected by chromosomal analysis. We found that basophils can be confirmed on the basis of the positivity for CD203c and CD294 by flow cytometric analysis. We also found by cytogenetic analysis that basophils were derived from myeloblasts. He refused any chemotherapy and became transfusion-dependent. He died nine months after the transformation. We should keep in mind that MDS could transform to AML with basophilic differentiation when peripheral basophilia in addition to myeloblasts develops in patients with MDS.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2939-2939
Author(s):  
Robin L. Perry ◽  
Patricia Vanessa Sanchez ◽  
Jean-Emmanuel Sarry ◽  
Alexander Perl ◽  
Adam Bagg ◽  
...  

Abstract Xenotransplantation of human acute myeloid leukemia (AML) in immunocompromised animals has been critical for the definition of leukemic stem cells. However, existing immunodeficient strains such as NOD/SCID and NOD/SCID/b2mnull have short life spans, age dependent leakiness of humoral immunity and low levels of AML cell engraftment making long-term evaluation of primary human AML biology difficult. A recent study suggested that the nonobese diabetic/severe combined immondeficient/IL2Rgnull (NOG) mouse has enhanced ability to engraft AML cells but this study relied on neonatal injections that are technically challenging. We performed an extensive analysis of AML engraftment in adult NOG mice using intravenous tail vein injection. Thirty-six different AML samples were analyzed including 2 samples of acute promyelocytic leukemia (APML). We used a threshold for AML engraftment of >0.5% human CD45+33+ cells in the murine bone marrow. Based on this threshold, 22 samples (61%) showed engraftment in NOG mice. Of these samples, 14 (64%) showed high levels of engraftment (greater than 10% of murine marrow replaced with human CD45+CD33+ cells). Engraftment did not correlate with FAB subtype or cytogenetic abnormalities to a statistically significant degree, however we noted that one sample with an 11q23 translocation and several samples with Flt3 ITD mutations showed consistent high level engraftment. Several samples demonstrated engraftment as high as 95% of the murine marrow with total AML cell expansion of 2-30 fold. Evaluation of AML stem cell frequency and expansion is ongoing. Engraftment in spleen was variable and in general significantly lower than in bone marrow. For most samples, peripheral blood engraftment was barely detectable. In contrast to NOD/SCID mice, both APML samples engrafted well in the NOG mouse with high levels of peripheral blood involvement. Some samples occasionally showed engraftment of a population of cells expressing CD2 and other T cell associated markers by flow cytometry, however this observation was inconsistent even between mice injected with the same sample. All samples tested (n=5) showed consistent engraftment in secondary and tertiary recipients with most samples tested showing further expansion of total AML cells in subsequent transplants. Importantly, a number of animals developed organomegaly and a wasting illness consistent with advanced leukemic disease. Several such animals showed extramedullary leukemic infiltration into non-hematopoietic tissues. Etoposide monotherapy (40 mg/kg in divided doses) of heavily engrafted mice did not induce a significant response in terms of leukemia regression. Studies of other chemotherapeutic agents are ongoing. We conclude that the NOG xenotransplantation model is a robust model for studying human AML cell engraftment which will allow for better characterization of AML biology and testing of new therapies


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5170-5170
Author(s):  
Bradley A Patay ◽  
Andrew Carson ◽  
Timothy J Martins ◽  
Sylvia Chien ◽  
Mary-Elizabeth M. Percival ◽  
...  

Abstract Context: The 5-year survival rate for acute myeloid leukemia (AML) is 26.6%. The prognosis of patients with adverse events such as older age or unfavorable risk cytogenetics remains poor, even for those who undergo allogeneic hematopoietic cell transplant. AML is a heterogeneous disease and novel N-of-1 clinical trial designs may offer benefit to individuals compared to conventional clinical trials by offering improved utilization of investigational chemotherapy regimens. Precision medicine based assays that reveal a deeper understanding of the cancer biology and potential for novel therapeutics may improve survival in the future. Objective: The goal of the clinical trial on which this patient was enrolled, Individualized Treatment for Relapsed/Refractory Acute Leukemia Based on Chemosensitivity and Genomics/Gene Expression Data (ClinicalTrials.gov Identifier:NCT01872819) was to determine feasibility of a study that utilized results of comprehensive mutation analysis and an in vitrohigh throughput functional assay to choose treatment for individual patients with refractory AML. Feasibility was defined as initiating chosen treatment within 21 days. A secondary objective was to achieve a response (cytoreduction or at least partial response) greater than that expected for comparable refractory patient populations with other salvage regimens. Design, Setting, and Patient: A single center enrolled individuals who had failed at least 2 inductions at initial diagnosis or >1 salvage regimen for relapsed AML. Patients could receive any FDA approved drug or combination regimen based on molecular analysis and high throughput drug sensitivity assay. A 58 year old female was enrolled into this protocol with MECOM (EVI1) rearranged, Monosomy 7 refractory AML. Methods and Main Outcome Measures: The patient had various assays performed on her samples, including next generation sequencing and a high throughput in vitro assay that analyzed enriched blast samples for sensitivity to 150 drugs or drug combinations. MyAML™, a next generation sequencing panel, analyzed 194 genes including breakpoint hotspot loci with long paired end sequencing and high depth that optimized detection of large insertion and deletions and other structural variants found in AML at low variant allele frequency. Results: The MyAML assay detected multiple variants including: NRAS:c.38G>A; p.Gly13Asp VAF = 100%, RUNX1:c.494_495ins; p.165_R166ins VAF = 42%, WT1:c.1149_1150ins; p.E384Pfs*5 VAF=38%. Fusions were also detected: t(13;17)(q12.2;q11.2), t(8;13)(q21.13;q12.2), and t(9;12)(q32;p13.2) which involved FLT3 novel fusions. FLT3 internal tandem duplications (ITD) or tyrosine kinase domain (TKD) variants were not detected. The assay also detected the Monosomy 7 and confirmed the t(2;3) as a THADA-MECOM (EVI1) fusion to the nucleotide breakpoint. These variants involved activated signaling, myeloid transcription factor and DNA demethylation pathways. The high throughput drug sensitivity assay identified sensitivity to kinase inhibitors such as the MEK inhibitor selumetinib (IC50 8.1 X 10e-9 M), Flt3 inhibitor staurosporine (IC50 1.4 X 10e-8 M) and Abl kinase inhibitor ponatinib (IC50 2.7 X 10e-8 M). Insurance coverage for these agents was not able to be obtained as this indication would be considered off label. However, based on the FLT3 fusion we identified using MyAML, we decided to treat the patient with the tyrosine kinase inhibitor sorafenib that we were able to obtain through a patient assistance program. This novel case is the first demonstration of a FLT3 fusion detection with a drug sensitivity assay suggesting that kinase inhibitors with multiple targets might be suitable for this type of variant. Conclusion: Specialized assays designed to identify clonal and subclonal architecture of genes associated with specific diseases can reveal variants that present therapeutic options not currently utilized for high risk patients, suggesting broader use of this approach could improve current clinical outcomes. Disclosures Patay: Invivoscribe, Inc: Consultancy. Carson:Invivoscribe, Inc: Employment. Becker:GlycoMimetics: Research Funding.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 276-276 ◽  
Author(s):  
Kazuko Kudo ◽  
Seiji Kojima ◽  
Ken Tabuchi ◽  
Eisaburo Ishii ◽  
Hiromasa Yabe ◽  
...  

Abstract Purpose: Recent multi-institutional studies have reported that children with Down syndrome (DS) and acute myeloid leukemia (AML) have a favorable outcome with less intensive chemotherapy. Based on our previous trial (Kojima, et al: Leukemia; 14:786,2000), the Japanese Childhood AML Cooperative Study Group conducted the AML-Down protocol study designed for children with DS and AML. Patients and Method: Between February 2000 and June 2004, 72 children (44 boys, 28 girls; median age, 1 year; range, 7 months to 7 years) were enrolled in this study. The median white blood cell count was 5,800/10−9 L. The median follow-up period was 3 years (range, 9 months to 5 years). Acute megakaryocytic leukemia (M7) was diagnosed most often (90%). The treatment regimen consisted of 5 cycles of Ara C 100mg/m2 (1-hour infusion) x 7 days, THP-ADR 25mg/m2 x 2 days, and etoposide 150mg/m2 x 3days. No prophylaxis against CNS leukemia was included. Results: Among the 72 children, 69 achieved complete remission (CR) after 1 to 2 cycles of induction therapy, with no deaths occurring during the induction period. One of 3 patients with induction failure achieved CR after another intensified chemotherapy. Eight patients relapsed during chemotherapy. One relapsed while off therapy and successfully entered a second remission after an intensified chemotherapy, followed by an allogeneic bone marrow transplant. There was no CNS relapse alone, although 1 patient relapsed in the bone marrow and CNS simultaneously. Eight relapsed patients and 2 refractory patients died without achieving a remission. The cause of death was pneumonia in 4 patients and disease progression in 7 patients. One patient died from pneumonia during the first CR. The CR rate, 3-year survival rate, and event-free survival (EFS) rate were 97.2%, 84.4%, and 83.0%, respectively. In a univariate analysis of factors that predict EFS, we found that the presence of monosomy 7 cytogenetic abnormality at diagnosis, and response to induction therapy were predictive factors for EFS. Neither age older than 2 years nor higher white blood cell count at diagnosis were statistically significant risk factors. Children with monosomy 7 had more adverse outcomes than those without monosomy 7 (41.7% vs 86.4%, p=0.02). Discussion: Our AML protocol specified for children with DS and AML does not include high-dose Arac and is much less intensive than other protocols used for treatment of these children. However, this less intensive regimen leads to an excellent outcome. In contrast to a previous study reported from CCG (Children’s Cancer Group) in the United States, age was not a significant risk factor. However, monosomy 7 is a poor prognostic factor in children with AML, whether or not they have DS. Our study strongly suggests that children with DS and AML can be treated successfully with a less intensive chemotherapy regimen that does not include high-dose Arac.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4557-4557
Author(s):  
Sze F. Yip ◽  
Thomas S.K. Wan ◽  
Herman S.Y. Liu ◽  
Michael L.G. Wong ◽  
Li C. Chan

Abstract The Philadelphia chromosome (Ph) is present in less than 3% of the acute myeloid leukemia (AML) and associated with poor prognosis. There are only few reports on the use of imatinib in Ph+ AML. We describe an adult female patient presenting with AML (Hb 3.6 g/dl, Wbc 137×109/L, Plt 295,000/uL and marrow blast 74%, with no basophilia or splenomegaly). Cytogenetic studies showed 45,XX,inv(3)(q21q26),-7,t(9;22)(q34;q11.2) [12] and RT-PCR showed p190BCR-ABL transcripts. The leukemia failed to respond to 2 cycles of daunorubicin and ara-C and imatinib 600mg daily was then started. Fluorescence in-situ hybridization (FISH) analysis of 300 marrow cells using dual fusion BCR-ABL translocation probes (D-FISH, Vysis, Downers Grove, IL, USA) showed that before imatinib, 78% of the cells were positive for BCR-ABL (at diagnosis, it was 83%), which after 6 weeks of imatinib treatment, became BCR-ABL negative (sensitivity 0.3%). However, marrow biopsy still showed 15% blasts. Repeated cytogenetics showed poor growth, but one metaphase showed 45,XX,inv(3)(q21q26),-7. With FISH, 39% of the marrow cells, including neutrophils, were positive for monosomy 7. At 4 months of imatinib, when the marrow blasts count increased to 40%, BCR-ABL remained negative by FISH, whereas monosomy 7 positive cells had risen to 82%. However, by nested RT-PCR with sets of BIOMED primers (sensitivity of 10−5), the p190BCR-ABL transcript was still present, suggesting persistence of a small and quiescent population of BCR-ABL positive leukemic stem cells. In summary, our case study demonstrates the need for careful cytogenetic and molecular analysis in cases of acute leukaemia with Ph chromosome to exclude the possibility of Ph chromosome being a secondary event, in which case durable leukemia remission will be unlikely if treatment is by BCR-ABL inhibition alone.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1630-1630
Author(s):  
Akira Shimada ◽  
Shelley Orwick ◽  
Hiroyuki Fujisaki ◽  
Dario Campana ◽  
Sharyn D. Baker

Abstract There is an increasing body of evidence indicating that the bone marrow microenvironment can generate drug resistance in acute leukemia. The mechanisms underlying this effect have not yet been elucidated; signals triggered by direct contact with extracellular matrix components and by mesenchymal cell (MSC)-secreted factors have been implicated. The protective effect of the microenvironment has been primarily observed for classical chemotherapeutic drugs. Recent reports, however, indicate that this can also occur with molecularly targeted therapies. Thus, it was shown that interleukin (IL)-7 desensitizes BCR-ABL+ leukemic cells to imatinib (Williams RT et al. Genes Dev 2007) and MSC-conditioned media protects BCR-ABL+ cells to imatinib and nilotinib (Weisberg E at al. Mol Cancer Ther, 2008). Several tyrosine kinase inhibitors are in clinical development for the treatment of acute myeloid leukemia (AML). The aim of this study was to determine whether bone marrow MSC affected the sensitivity of AML cells to 3 promising tyrosine kinase inhibitors (sorafenib, sunitinib, and midostaurin) and, if so, to begin to elucidate the underlying mechanisms. Using proliferation assays, we found that 3 AML cells lines (MV4-11, U937, and THP1) were significantly less sensitive to the tyrosine kinase inhibitors when cultured in the presence of bone marrow-derived MSC for 24h before exposure to drugs for 72h. In experiments with MV4-11, IC50 increased from 4.7 nM to 55 nM for sorafenib, from 10 nM to 110 nM for sunitinib, and from 28 nM to 135 nM for midostaurin; in experiments with U937, IC50 increases were 5.1 μM to 11 μM, 6.2 μM to > 10 μM, and 230 to > 1000 nM for each drug; and in experiments with THP1, they were 6.3 μM to 11 μM, 2.2 μM to > 10 μM, and 211 nM to 996 nM. Coculture with MSC also reduced sorafenib- and sunitinib-induced apoptosis by > 60%. Interestingly, drug resistance increased even further after coculturing the cell lines with MSC for 4 weeks or longer: sunitinib had virtually no effect on the proliferation of MV4-11 cells at concentrations of up to 100 nM, and on THP-1 cells at 10 μM. To determine whether the induction of drug resistance was dependent on the direct contact of AML cells with MSC, we tested sensitivity to sorafenib after separating MV4-11cells from MSC with transwell inserts. Under these conditions, the protective effect of MSC was lessened but not abrogated. These results indicated that direct contact with MSC was not an absolute requirement for induction of drug resistance and that MSC-secreted soluble factors might be, at least in part, involved. We therefore determined the soluble factors secreted by MSC using a multiplex assay and tested whether their secretion was augmented by contact with AML cells. MSC secreted IL-6 (230 pg/mL), IL-8 (1880 pg/mL), and VCAM-1 (30 pg/mL). When cocultured with MV4-11, U937 and THP-1 cells for 24h, IL-6 secretion increased 1.3 to 1.8-fold, IL-8 increased 1.5 to 2.6-fold, and VCAM-1 increased 2.2 to 5.6-fold; after 72 of coculture, dramatically elevated levels of IL-6 (2140–3869 pg/mL), IL-8 (4296–8068 pg/mL), and VCAM-1 (5109–6389 pg/mL) were observed. The effects of these and other MSC-derived factors on the sensitivity of AML cells lines and primary AML cells to tyrosine kinase inhibitors are being tested. These results indicate that the anti-AML effect of tyrosine kinase inhibitors is strongly inhibited by bone marrow MSC cells, and support the concept that the microenvironment is an important determinant of resistance to these agents in leukemia. We suggest that the development of agents that interfere with the interaction between AML cells and MSC, and with the molecular mechanisms underlying this protective effect of MSC is a crucial step to improve cure rates.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3944-3944 ◽  
Author(s):  
Yao-Te Hsieh ◽  
Enzi Jiang ◽  
Jennifer Pham ◽  
Hye-Na Kim ◽  
Hisham Abdel-Azim ◽  
...  

Abstract Despite aggressive chemotherapy and early allogeneic transplantation, acute myeloid leukemia (AML) frequently relapses, so that over-all disease-free survival remains below 50%. Strategies to overcome the chemoresistance of relapse-initiating residual AML blasts are, therefore, warranted. Evidence has been provided that AML cells are sheltered from the insult of chemotherapeutic agents by interacting with bone marrow stroma. Integrin alpha4beta1 (VLA4) mediates adhesion of hematopoietic cells to bone marrow stroma cells and extracellular matrix and has been implicated in cell adhesion-mediated chemotherapy resistance. Based on the evidence thereof provided for ALL blasts, VLA4 is here proposed as a therapeutic target for refractory AML. For this purpose, VLA4 was functionally blocked in vitro and in vivo on patient-derived AML cells using an anti-functional humanized VLA4 antibody, Natalizumab (NZM). VLA4-positive (>90%) patient-derived (primary) AML cells were plated on immobilized human VCAM1 or human stromal cell line HS-5 and treated with control (IgG4) or Natalizumab (NZM) for 2 days. NZM de-adhered 94.0%±7.6 AML cells from its counter receptor VCAM-1, yet only 31.3%±13.8 from HS-5, indicating that stroma cells offer ligands for a wider panel of adhesion receptors besides VLA4. We tested also whether VLA4 blockade is beneficial against AML when combined with chemotherapy. For this purpose, primary AML cells were incubated with NZM and incubated on uncoated tissue culture plates or HS-5 stromal layers in the presence or absence of Ara-C (1µM) for two days. AML cells showed higher viability under Ara-C therapy when incubated with HS-5 cells compared to controls, indicating the chemoprotective effect of the stromal layer. The viability of the AML cells treated with combined Ara-C and NZM was similar to the controls, indicating that HS-5-mediated chemo-protection was completely abrogated by NZM. Significantly more AML cells treated with Ara-C+NZM stained AnnexinV+/7AAD- than after Ara-C+control Ig4 treatment (44.4%±5.6 vs. 29.8%±4.8, p=0.03) indicating increased apoptosis of AML cells. On its own, NZM did not induce apoptosis. Next, we tested NZM as a single agent in our NOD/SCIDIL2Rγ deficient (NSG) xenograft model of primary AML. Luciferase-labeled AML cells were intrafemorally injected into NSG mice (1x105 cells / mouse). NZM (5mg/kg) was given intraperitoneally once per week for 4 weeks. NZM-treated animals survived significantly longer than control Ig-treated animals (Median Survival Time, MST=107 days vs. MST=76 days; *p=0.008 by Log-rank Test.To determine effects of NZM on leukemia cell burden/distribution in different organs, primary AML cells were injected into NSG mice and allowed to engraft for 3 days, subsequently treated with a single dose of NZM or Ig control. 72 hours later, AML cell burden in femurs and spleens of NZM-treated animals was significantly decreased compared to control treated mice, however AML cells were not increased into the peripheral blood, so that whether leukemia cells were selectively killed ormobilized and then retained in non-hematopoietic organs remains to be determined. Further studies addressing molecular mechanisms of increased apoptosis after combined VLA4 blockade and chemotherapy are ongoing. Our data suggest that the paradigm of leukemia cell targeting by VLA4 blockade, previously demonstrated by us for ALL, can also be applied to AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3064-3064
Author(s):  
Amina M Abdul-Aziz ◽  
Manar S Shafat ◽  
Matthew J Lawes ◽  
Kristian M Bowles ◽  
Stuart A Rushworth

Abstract Introduction: Acute myeloid leukemia (AML) cells exhibit a high level of spontaneous apoptosis when cultured in vitro but have a prolonged survival time in vivo indicating that the tissue microenvironment plays a critical role in promoting AML cell survival. Knowledge of the complexity of the bone marrow microenvironment is increasing especially with respect to the bone marrow mesenchymal stromal cells [BM-MSC] which are considered a major protective cell type. Other studies have demonstrated the ability of BM-MSC to protect leukemia cells from spontaneous and chemotherapy-induced apoptosis. Increasing evidence suggests the existence of crosstalk between leukemia cells and BM stromal cells to create a leukemia-promoting environment. Recently our group and others have shown that this crosstalk is achieved by a complex communication system that involves multiple bidirectional signals which enhance AML survival and proliferation. Here we report a novel interaction between AML blasts and BM-MSC which benefits AML proliferation and survival. Methods: To investigate the interaction between primary AML blasts and BM-MSC we isolated AML and BM-MSC from the same patient and used an autologous in vitro culture assay to analyze the cytokine profile. Conditioned medium was collected from cultures of primary human AML alone or cultured with autologous BM-MSC and analyzed using Proteome Profiler Human XL Cytokine Array and target specific ELISAs. Real-time PCR was also used to verify the array data. MIF-Receptor inhibitors (SB 225002- CXCR2, AMD3100 - CXCR4 and CD74 blocking antibody - CD74) and signaling kinase inhibitors (LY294002- PI3K/AKT, PD098059 - MAPK, Ro-31-8220 - PKC) were used for initial determination of MIF signaling pathways in BM-MSC. Specific PKC isoform inhibitors (Go6976-PKCα/ß and enzastaurin -PKCß) were then used to determine isoform specific activation. Western blot and siRNA were used to confirm the role of AML derived MIF in regulating downstream BM-MSC signaling pathways including MAPK, PI3K/AKT, and PKC. Results: We initially examined the cytokine profile in cultured human AML compared to AML cultured with autologous BM-MSC or BM-MSC alone and found that MIF was highly expressed by primary AML and that IL-8 was increased in AML/BM-MSC co-cultures. The observed changes in IL-8 were confirmed by ELISA assays. RT-PCR was used to measure MIF and IL-8 gene expression from RNA extracted from primary AML or BM-MSC cultured alone or in combination. Results confirmed that MIF is highly expressed at the RNA and protein level by AML blasts and IL-8 transcription and cytokine release was upregulated in BM-MSC in response to co-culture with AML. Next we found that recombinant MIF increases IL-8 mRNA and protein expression in BM-MSC. Moreover, the MIF inhibition by, ISO-1, inhibits AML induced IL-8 expression and secretion by BM-MSC. Next we sought to determine which kinase signaling cascade is activated by MIF. We used a panel of protein kinase inhibitors and found that the pan-PKC inhibitor Ro-31-8220 completely inhibits AML and MIF induced IL-8 mRNA at sub micromolar concentrations. To further identify the specific PKC isoform responsible for linking AML induced MIF to IL-8 we used PKC isoform specific inhibitors (Go6976 and enzastaurin) which significantly inhibited MIF induced IL-8 expression and protein in BM-MSC. The introduction of PKCß siRNA dramatically inhibited MIF induced IL-8 mRNA expression in BM-MSC confirming that PKCß regulates AML induced BM-MSC derived IL-8 expression. Finally, inhibition of AML/BM-MSC co-cultures with the PKCß inhibitor enzastaurin inhibits BM-MSC induced AML survival in vitro. Conclusions: These results reported here show a novel bidirectional survival mechanism between AML blasts and BM-MSC. Furthermore this work identifies the PKC-ß-IL8 pathway in the BM-MSC of patients with AML as a novel target for future treatment strategies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 87 (12) ◽  
pp. 4979-4989 ◽  
Author(s):  
WG Woods ◽  
N Kobrinsky ◽  
JD Buckley ◽  
JW Lee ◽  
J Sanders ◽  
...  

Timed sequencing of cycles of induction chemotherapy in acute myeloid leukemia (AML) has been proposed as a way to achieve maximal leukemic cell kill through recruitment and synchronization of residual neoplastic cells. Furthermore, whether intensive induction therapy should be continued in the presence of profound myelosuppression is an important question. The Children's Cancer Group (CCG) conducted a prospective randomized trial in which 589 patients with AML were randomized at diagnosis to one of two induction approaches involving a 4-day cycle of five active chemotherapeutic agents, with the second cycle administered either 10 days after the first cycle, despite low or dropping blood counts (intensive timing), or 14 days or later from the beginning of the first cycle, depending on bone marrow status (standard timing). All patients achieving remission received a total of four cycles of induction therapy. They were then allocated to allogeneic bone marrow transplantation (BMT) if a compatible family donor was present or randomized to aggressive nonmyeloablative therapy or to myeloablative therapy with purged autologous BMT rescue. The three postremission arms remain coded. Induction success and median days to complete induction were similar for the 295 patients randomized to the intensive timing arm (75%, 99 days) compared with the 294 patients randomized to the standard timing arm (70%, 105 days; P = .18 for remission). However, a marked improvement in outcome was demonstrated in patients randomized to the intensive timing arm, with an actuarial event-free survival at 3 years of 42% +/- 7% (95% confidence interval [CI]) versus 27% +/- 6% for patients on the standard timing arm (P = .0005). Disease-free survival results at 3 years from the end of induction were superior for patients receiving intensively timed induction therapy (N = 211), 55% +/- 9% versus 37% +/- 9% for standard timing patients (N = 195, P = .0002), with a median follow-up from achieving remission of 28 months. Superior results were documented for patients receiving intensive timing irrespective of the postremission therapy to which they were allocated. Intensively timed induction therapy for patients with AML markedly improves event-free survival, even for patients undergoing myeloablative therapy with BMT rescue. Without controlling for the type of induction therapy received, results of various BMT studies in AML comparing different preparative regimens will be difficult to interpret.


Sign in / Sign up

Export Citation Format

Share Document