New Approach to the Mechanism of Hyperleukocytosis in Patients with APL After Treatment with ATRA.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3585-3585
Author(s):  
Guosheng Jiang ◽  
Kehong Bi ◽  
Chuanfang Liu ◽  
Peie Wen

Abstract Abstract 3585 Poster Board III-522 Objective To detect the new approach to the mechanism of hyperleukocytosis during induction therapy with ATRA in patients with acute promyelocytic leukemia. Methods Diagnosis of acute promyelocytic leukemia (APL) was performed according to the FAB cytological classification criteria and cytogenetic criteria. The collected leukemia cells were re-suspended in ATRA at 37°C for 96 h. Differentiation of leukemia cells was assessed by NBT reduction assay and indicated as percent of CD11b positive cells. Constant MTT assay and cell number count were used to detect the proliferation of leukemia cells after treatment with ATRA. For FACS analysis, cells were incubated with CD11b, CD54, CD106 and Ki67 mAb or isotypic control IgG1 antibody. L-CFU assay was taken to measure the colony formation. GM-CFU assay was used to detect the granulocyte-macrophage colony-forming unit. The mRNA expression of MMP-9, TIMP-1 was detected by RT-PCR. RESULTS (1) The results showed that the total number of WBC was up-regulated after treatment with ATRA in patients with APL, which was chiefly due to the up-regulation of myelocytes and more matured granulocytes. (2) The primary APL cells, unlike normal promyelocyte, have little requirement for survival factors, Its plating efficiency was also lower and could be up-regulated by exposure to ATRA. MTT assay showed that the OD value of primary APL cells was lower than that of NB4 clone cells or normal hematopoitic cells. Otherwise, the proliferation of primary APL cells, post exposure to ATRA, was up-regulated and more cytokine-dependent or more sensitive to G-CSF stimulation. (3) Most importantly, the division hypothesis should be taken into consideration, because the leukocytosis chiefly contributed to the up-regulation of differentiated myelocytes and more matured neutrophils, which should be the key proliferation pool as the normal myelocytes. The primary APL cells had a low proliferation potential, the mitotic cells are often scarcely detected even for 4 d in culture. Most of primary APL cells had a growth arrest in G0/G1 or G0 phase. The APL cells occur to switch from G0 into cell cycle that was indicated by the up-regulation of Ki67 antigens. In our opinion, most of APL cells enter into cell cycles after exposure to ATRA or arsenic trioxide in vivo, with more mitotic cells. (4) The rheological hypothesis was taken into consideration. The up-regulation of adhessive ability of APL cells, which resulted in more release from bone marrow into peripheral blood. The results indicated that ATRA can induce differentiation of the malignant cells, most remarkable feature was the progressive change of malignant cells with signs of their terminal differentiation and with Auer rods being sometimes observed in mature cells. Usually, the percent of promyelocyte was down-regulated and the percent of myelocyte, metamyelocyte and more matured myeloid cells was obviously up-regulated, especially for the percent of myelocyte or myelocyte-like cells. These results suggest that an asynchronism between rheological and morphological maturation in each APL cell might explain the occurrence of hyperleukocytosis in some patients during ATRA therapy. For example, the serum sICAM1 and sVCAM1level was up-regulated, the secretion up-regulation of adhesive molecules ICAM1 and VCAM1 in primary APL cells was also observed, as inagreement with the up-regulation of CD11b, CD54 and CD106. The adhessive coefficiet was also up-regulated. Conclusion The leukocytosis or hyperleukocytosis chiefly contributed to the up-regulation of myelocyte-like cells, which chiefly contributed to the more myelocytes divisions and more sensitive to cytokine stimulation. Partly due to the up-regulation of adhesive index and adhesion molecules of differentiationed leukemia cells, which could easily resulted in the release of APL cells from bone marrow to peripheral blood. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 246-246 ◽  
Author(s):  
Bruno Nervi ◽  
Matthew Holt ◽  
Michael P. Rettig ◽  
Gary Bridger ◽  
Timothy J. Ley ◽  
...  

Abstract CXCR4/SDF-1 axis regulates the trafficking of normal stem cells to and from the bone marrow (BM) microenvironment. SDF-1 is a chemokine widely expressed by many tissues especially BM stromal cells and osteoblasts. AMD3100 (AMD) is a novel bicyclam molecule that is a competitive inhibitor of SDF-1/CXCR4 binding and has been used to enhance stem cell mobilization when combined with G-CSF in mouse, dog and man. We are interested in evaluating whether leukemic cells “mobilize” similar to normal stem cells after treatment with AMD, and if so, whether this mobilization increases the efficacy of chemotherapy. Therefore, we utilized a mouse model of human acute promyelocytic leukemia (APL) in which the PML-RARα transgene was knocked into a single allele of the murine cathepsin G locus. To more efficiently track the leukemic cells, we transduced banked APL tumors with a dual function reporter gene that encodes a fusion protein comprised of click beetle red (CBR) luciferase, a bioluminescence imaging (BLI) optical reporter gene, and EGFP for ex vivo cell sorting (CBR/EGFP). We generated large numbers of CBR/EGFP+ APL cells by isolating EGFP+ cells using a MoFlo cell sorter, and passaging them in secondary syngeneic recipients. Importantly, the secondary recipients developed a rapidly fatal acute leukemia after intravenously (iv) or intraperitoneal injection, which displayed an APL phenotype (CD34/GR1 co-expression) and exhibited luciferase activity. Upon iv injection into syngeneic recipients, the CBR/EGFP+ APL cells rapidly migrated to the BM microenvironment, as evidenced by the significantly increased BLI signal in the femurs, spine, ribs, and skull of recipients at 4 days after injection. Over the next 2–3 days the CBR/EGFP+ cells migrated to the spleen followed rapidly by widespread dissemination and death due to leukostasis by 14–16 days. To our knowledge, this represents the only mouse leukemia model in which leukemia cells home preferentially to the BM microenvironment in a manner that is similar to what is seen in human AML. Therefore, we used this model to study the effect of AMD on the “mobilization” of APL cells into the peripheral blood (PB) and on their sensitivity to chemotherapeutic agents that are known to affect the proliferation of these cells. Surprisingly, injection of AMD (5 mg/kg) immediately at the time of APL infusion had no impact on the engraftment (short term or long term) of either normal BM stem cells or the leukemic cells. However, we observed rapid mobilization of the leukemic cells when AMD was administered 11 days after APL injection. In fact, 40% of mice that received a single dose of AMD on day +11 after APL injection died 2 to 4 hours after AMD injection as a result of the rapid and massive mobilization of blasts. Overall, we found that AMD treatment on day +11 induced a 3-fold increase in total WBC counts with a 10-fold increase in the leukemic blasts into PB. Interestingly, the administration of AMD concomitant with cytarabine (AraC) (200 mg/kg) on day +11 significantly prolonged the overall survival of mice, compared with mice treated only with AraC. In summary, we developed a mouse model to study the APL cell trafficking, and we have shown leukemia cell mobilize from the BM into PB after AMD administration. We propose that CXCR4/SDF-1 is a key regulator for leukemia migration and homing to the BM. In these preliminary results, we observed that AMD sensitizes APL cells to AraC.


2014 ◽  
Vol 32 (15_suppl) ◽  
pp. 7079-7079
Author(s):  
Mariana Scaranti ◽  
Jose Mauricio Segundo Correia Mota ◽  
Ana Silvia Gouveia de Lima ◽  
Barbara Amelia Santana Lemos ◽  
Antonio Roberto Lucena-Araujo ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3756-3756 ◽  
Author(s):  
Koji Sasaki ◽  
Hagop M. Kantarjian ◽  
Raja Luthra ◽  
Keyur P. Patel ◽  
Guillermo Garcia-Manero ◽  
...  

Abstract Introduction: Acute promyelocytic leukemia (APL) is biologically distinct subtype of acute myeloid leukemia characterized by PML-RARA fusion transcripts, and its survival has significantly improved with all-trans retinoic acid (ATRA). Monitoring PML-RARA fusion transcripts with quantitative real-time polymerase chain reaction (RT-PCR) is the standard of care for the evaluation of response in patients with acute promyelocytic leukemia. We sought to determine the correlation between peripheral blood (PB) and bone marrow (BM) samples in patients treated on our frontline studies. Methods: We correlated results from BM and PB samples obtained from patients with newly diagnosed APL treated on three consecutive prospective clinical trials of combination of arsenic trioxide (ATO) and ATRA with or without gemtuzumab ozogamycin (ID01-014; NCT01409161; and NCT00413166) at our institution. Qualitative RT-PCR was performed on reverse-transcribed RNA from PB and BM samples for the short and long isoforms of PML-RARA, and the percent ratios of PML-RARA to ABL1 transcript levels were calculated. The sensitivity of detection with RT-PCR was 1 in 100,000. For this analysis, we selected samples with the time interval of collection within 1 day. Qualitative correlation was assessed using Kendall tau rank correlation coefficient test. Spearman's correlation coefficient was computed for quantitative variables to measure the extent of the association between samples. P values were two-sided and a p value of <0.05 was considered as statistically significant. Results: From July 2002 to May 2015, 184 patients were enrolled in the clinical trials. RT-PCR for PML-RARA was performed in 2077 samples including 1261 BM samples and 816 PB samples. In total, 584 samples (292 sample pairs) from BM and PB were identified within 1-day time interval. PML-RARA levels from PB samples with RT-PCR were strongly qualitatively and quantitatively correlated with those from BM samples (r= 0.831, p<0.001; τ= 0.792, p<0.001, respectively). Minimal qualitative discrepancy was observed in 13 samples from 11 patients: 8 BM samples from 8 patients with PML-RARA/ABL1 ≤0.01% (4 short form; 4 long form), with undetectable PML-RARA from concurrent PB; and 5 PB samples from 4 patients with PML-RARA/ABL1 ≤0.01% (2 short form; 3 long form), with undetectable PML-RARA from concurrent BM samples. Of 8 BM samples, 4 BM samples were obtained after 2 cycles, 3 cycles, 3 cycles, and 4 cycles of ATRA + ATO, respectively; 4, 1 month, 9 months, 9 months, and 10 months after completion of ATO + ATRA. The discrepancy of 4 BM samples during ATRA + ATO was resolved with undetectable PML-RARA level from peripheral blood samples and bone marrow samples after 1 additional cycle of ATO + ATRA. Of 4 BM samples after completion of ATO + ATRA, previous PML-RARA showed undetectable PML-RARA level, and repeat PML-RARA showed undetectable PML-RARA level from bone marrow and peripheral blood samples without intervention. Of 5 PB samples from 4 patients, 3 PB samples from 2 patients were obtained after 3 cycles, 3 cycles, and 4 cycles of ATRA. One patient had persistent PML-RARA/ABL1 <0.01% (short form) from peripheral blood samples with undetectable PML-RARA from concurrent BM samples after 3 cycles and 4 cycles of ATO + ATRA. Two PB samples from 2 patients were obtained 1 month after completion of ATO + ATRA. Conclusions: Strong correlation between PB and BM results indicate that PML-RARA fusion transcript levels in patients with APL can be monitored using PB samples. Figure 1. The correlation between bone marrow and peripheral blood samples for detection of PML-RARA Figure 1. The correlation between bone marrow and peripheral blood samples for detection of PML-RARA Disclosures Verstovsek: Incyte Corporation: Research Funding. Estrov:incyte: Consultancy, Research Funding. Cortes:Novartis: Consultancy, Research Funding; Teva: Research Funding; BerGenBio AS: Research Funding; BMS: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Ariad: Consultancy, Research Funding; Astellas: Consultancy, Research Funding; Ambit: Consultancy, Research Funding; Arog: Research Funding; Celator: Research Funding; Jenssen: Consultancy.


2019 ◽  
Vol 2019 ◽  
pp. 1-7
Author(s):  
Yuta Inagawa ◽  
Yukiko Komeno ◽  
Satoshi Saito ◽  
Yuji Maenohara ◽  
Tetsuro Yamagishi ◽  
...  

A 34-year-old woman was diagnosed with acute promyelocytic leukemia. Chemotherapy was administered following the JALSG APL204 protocol. Induction therapy with all-trans retinoic acid resulted in complete remission on day 49. She developed coccygeal pain from day 18, which spread to the spine and cheekbones and lasted 5 weeks. She had similar bone pain on days 7–10 of the first consolidation therapy and on days 4–12 of the second consolidation therapy. Oral loxoprofen was prescribed for pain relief. On day 33 of the third consolidation, white blood cell and neutrophil counts were 320/μL and 20/μL, respectively. After she developed epigastralgia and hematemesis, she developed septic shock. Gastroendoscopy revealed markedly thickened folds and diffusely damaged mucosa with blood oozing. Computed tomography revealed thickened walls of the antrum and the pylorus. Despite emergency treatments, she died. Bacterial culture of the gastric fluid yielded Enterobacter cloacae and enterococci growth. Collectively, she was diagnosed with phlegmonous gastritis. Retrospective examination of serial bone marrow biopsy specimens demonstrated progressive bone marrow fibrosis, which may have caused prolonged myelosuppression. Thus, evaluation of bone marrow fibrosis by bone marrow biopsy after each treatment cycle might serve as a predictor of persistent myelosuppression induced by chemotherapy.


Sign in / Sign up

Export Citation Format

Share Document