Platelet Factor 4 Regulates Platelet Count In Vivo: Implications for Platelet Recovery after Cytotoxic Therapy.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3144-3144
Author(s):  
Michele P. Lambert ◽  
M. Anna Kowalska ◽  
Mortimer Poncz

Abstract Platelet factor 4 (PF4), a platelet-specific CXC chemokine, was the first reported negative autocrine regulator of megakaryopoiesis in vitro. To define the physiological role(s) of PF4, we established mice that either were deficient in murine (m) PF4 (mPF4−/−) or that over-expressed human (h) PF4 (hPF4+/+). These mice had a level of PF4 ~6-fold greater than that present in human platelet controls. All lines studied had been backcrossed onto a C57Bl/6J background for >10 generations. Platelet counts in these animals correlated inversely with PF4 determined expression, beginning with a low platelet count of 702 ± 57 x 103/μL in the hPF4+/+ mice < hPF4+ < wildtype (WT) < mPF4+/− < mPF4−/− mice in which the platelet count was 1,404 ± 117 x 103/μL. The half-life of the platelets from the hPF4+/+ was identical to that of WT mice. Cultured bone marrow mononuclear cells (BMMNC) in serum-free media showed that each line had identical efficiency in growing megakaryocyte colonies, suggesting that megakaryocyte progenitor cells in these different genetic lines were intrinsically normal. Megakaryocyte colony numbers derived from WT BMMNC were reduced by the addition of recombinant PF4 or supernatant from irradiated bone marrow of hPF4+ mice, but not from mPF4−/− mice, suggesting that megakaryocyte lysis in vivo during cytoreductive therapy may contribute to the subsequent thrombocytopenia by releasing PF4. Additionally, a rabbit polyclonal anti-mPF4 antibody (Ab) was able in culture to significantly reverse this inhibitory effect of PF4 on megakaryopoiesis. Preliminary cytoreductive studies using either 600 cGy or 150 mg/kg of 5-fluorouracil (5-FU) intraperitoneally (IP) were performed. In irradiation studies, mPF4−/− mice began to recover on the same day as WT littermates, but they clearly had higher platelet counts at their nadir, with a drop to only 42 ± 7% of baseline vs. 32 ± 6% in the WT mice (n =12 in each arm, p = 0.06). By Day 13, 9 of 12 mPF4−/− mice had recovered to >75% of baseline, while only 3 of 12 WT mice had recovered (p <0.001). hPF4+ mice (n = 7) were studied after 5-FU treatment. Compared to WT littermates (n = 9), the hPF4+ recovered later (15.6 ± 2.2 vs. 11.2 ± 1.5 days, p < 0.0003), and clearly had significantly greater drop to 30 ± 6% vs. 56 ± 9% of baseline (p < 0.00001). By day 15, all of the WT mice had recovered, but only 43% of hPF4+ mice had returned to >75% of baseline platelet count (p = 0.009). To examine if anti-mPF4 Ab was protective of cytotoxic therapy-induced thrombocytopenia, WT mice were treated with 180 mg/kg of 5-FU and were given either anti-mPF4 Ab (25 mg/kg, IV, x 2) or an equal volume of vehicle. By day 5, the Ab-treated group had a platelet count of 45 ± 6% vs. 32 ± 4% in the untreated (n > 13 per arm, p = 0.015). Platelet counts remained higher in the Ab-treated arm throughout the study. By day 10 after intervention, 9 of 16 mice of the Ab-treated arm had platelet counts over 75% of the baseline, while only 3 of 13 control mice did (p < 0.001). Thus, it appears that PF4 is an important negative autocrine regulator of platelet count in vivo. Excessive release of PF4 following cytotoxic therapy may be a mediator of treatment-related thrombocytopenia. Strategies directed to alleviate the consequence of released PF4 may have clinical benefit in ameliorating this thrombocytopenia.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 93-93 ◽  
Author(s):  
Michele P. Lambert ◽  
Lubica Rauova ◽  
M. Anna Kowalska ◽  
Mortimer Poncz

Abstract Platelet Factor 4 (PF4) is a member of the CXC family of chemokines that appears to act as an autocrine regulator of megakaryopoiesis in vitro. To demonstrate that PF4 plays a physiological role in vivo, we examined megakaryopoiesis in animals that over- or under-expressed PF4. We generated PF4−/− knockout animals as well as transgenic animals that overexpress human (h) PF4 to a level that is ~6-fold greater than control human platelet samples (hPF4+). Relative to wildtype (WT) littermate controls (n=26), platelet counts were elevated by ~15% in knockout animals (n=18, p=0.001) and reduced by ~33% in hPF4+ animals (n=13, p=0.0004). Platelet survival studies insured that the observed changes were not due to a consumptive process. The hypothesis that PF4 results in a decrease in megakaryocyte and/or platelet production was supported by in vitro evidence that growth of megakaryocyte-containing colonies was inversely related to the PF4 phenotype. The hPF4+ mice formed 52%±15% fewer megakaryocyte colonies compared to WT (n=8 each arm, p<0.00002). This inhibition was blocked by the inclusion of a rabbit polyclonal anti-hPF4 antibody. Based on these observations, we hypothesized that lysis of megakaryocytes induced by cytolytic chemotherapy with resultant release of PF4 may contribute to post-cytolytic-therapy thrombocytopenia. Exposing mice to intraperitoneal 5-fluorouracil (5-FU) decreased WT platelet counts below normal for 10.8±1.3 days (n=23). PF4−/− mice had a significantly more rapid recovery of 8.2±1.4 days (n=14, p<0.0001), while hPF4+ mice had significantly delayed recovery of 15.3±1.7 days (n=15, p<0.000006). Importantly, the duration of thrombocytopenia in WT animals could by shortened to that seen in PF4−/− mice by administration of 50 mg/kg of F(ab)2 fragments of a polyclonal rabbit anti-murine PF4 antibody, but not by rabbit non-specific polyclonal IgG. Specific anti-PF4 therapy improved recovery of platelet count in WT mice to 8.8±1.5 days vs. 10.7±1.0 days in non-treated WT mice (n=10 and 8, respectively, p<0.008). Forty percent of the specific-treated animals recovered by day 7 post injection of 5-FU, while none of the control animals recovered before day 10. Similar interventions in hPF4+ animals resulted in a marked improvement in platelet count recovery compared to controls with the anti-hPF4 antibody-treated group recovering by 9.0±1 vs. 13.6±1.2 for the control mice (n=8 and 5, respectively, p<0.002). Our data suggest that efforts to block the effect of PF4 during cytotoxic therapy may limit the duration of thrombocytopenia. These observations have clinical implications, particularly for patients who express high endogenous levels of PF4. To that end, we studied 250 well adults and found that 10% of these individuals have 2–4.5 times the average platelet PF4 content. We propose that these individuals with high platelet PF4 levels may be particularly sensitive to developing thrombocytopenia after cytolytic therapy and may specifically benefit from therapy that blocks the effects of released PF4.


Blood ◽  
1996 ◽  
Vol 88 (3) ◽  
pp. 778-784 ◽  
Author(s):  
A Ohwada ◽  
S Rafii ◽  
MA Moore ◽  
RG Crystal

Abstract Thrombopoietin (TPO, c-mpl ligand) has emerged as a major hematopoietic cytokine stimulating megakaryocyte proliferation, endomitosis, and platelet production. This study shows that a single administration of an adenovirus (Ad) vector encoding TPO (AdCMV.TPO) abrogates thrombocytopenia induced in mice by carboplatin and irradiation. Normal Balb/c mice receiving the vector had increased platelet counts peaking at 7 days and returning to baseline by day 15. Mice rendered pancytopenic with 500 rads and 1.2 mg of carboplatin had a nadir platelet count of five percent of the baseline. Mice receiving AdCMV.TPO 3 days before receiving irradiation and chemotherapy achieved a platelet nadir fourfold higher, and had significant reduction in duration of thrombocytopenia, than mice receiving the control Ad vector. Introduction of AdCMV.TPO the same day of chemotherapy and irradiation was equally effective in acceleration of platelet recovery, but administration of AdCMV.TPO 3 days after chemotherapy-radiation had little effect on platelet recovery. At 30 days after therapy bone marrow and spleen of mice treated with AdCMV.TPO were populated with a large number of polyploid megakaryocytes, but there was no evidence of circulating megakaryocytes in the liver or lungs and no pathologic bone abnormalities such as osteosclerosis or myelofibrosis. These observations suggest that an Ad vector may be an excellent delivery system to provide adequate TPO production to maintain platelet levels in circumstances associated with life-threatening thrombocytopenia.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3178-3178
Author(s):  
Zhong Chao Han ◽  
Bin Liu ◽  
Lihua Zhao

Abstract In cancer therapy, specific radioprotection of normal tissue and antiangiogenesis are the ways to increase the therapeutic gain. Here we describe a novel gene therapy, which uses attenuated salmonella SL3261 as oral vectors carrying with cDNA of platelet factor 4 (PF4) or that of a truncated PF4. After oral administrations of attenuated salmonella carrying with cDNA of PF4 or truncated PF4, the survival rate of mice which received sublethal total body irradiation was improved by 50%, In comparison with the control mice, the bone marrow cells obtained from the mice of experimental group increased (13.2±8.3, 15.7±1.5 vs 4.1 ± 2.0 P<0.05) at day 7 after TBI, and the number of HPP-CFC of bone marrow cells also increased significantly (15.7±9, 11.7±5 vs 4.3±4.1 P<0.05) at day 7, suggesting a stimulating effect of PF4 on hematopoietic recovery. This gene therapy also caused significant tumor regression. The microvessel density (MVD) of tumors was significantly decreased in the group of treated mice compared to controls (4.25±0.96, 4.08±0.56 vs 11±0.83 P<0.05). Analysis TUNEL kit revealed an increase in the number of apoptosis cells in tumors of mice treated by SL3261 carrying with cDNA of PF4 or a truncated PF4. GFP expression and gene integration were detected in the liver, kidney, spleens, intestine, peripheral blood, bone marrow and tumors samples of the SL3261 treated mice, and the expression of GFP was higher in tumors than that in other tissues. These data demonstrate for the first time a dual biological function of PF4 against tumor growth and radiation injury. These results also demonstrate that attenuated salmonella can be used in vivo as a DNA delivery vector


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4150-4150
Author(s):  
Michele P Lambert ◽  
Liqing Xiao ◽  
Mortimer Poncz

Abstract We have previously shown that platelet factor 4 (PF4, CXCL4), which is synthesized almost exclusively by megakaryocytes undergoes release intramedullary after which it can undergo reuptake into alpha-granules, but also can be an important negative paracrine regulator of megakaryopoiesis, effecting platelet recovery post-radiation or chemotherapy. Animals that express high levels of human (h) PF4 in addition to their normal levels of murine (m) PF4 (hPF4+) have increased sensitivity to radiation- and chemotherapy-induced thrombocytopenia when compared to wild type (WT) mice or to mice that lack endogenous PF4 (mPF4-/-). Both PF4 reuptake and the negative paracrine effects are at least partially dependent upon the presence of low-density lipoprotein receptor related protein-1 (LRP1) on the surface of megakaryocytes as shown using shRNA suppression of megakaryocyte LRP1 levels. To further understand the role of LRP1 in megakaryopoiesis, we studied LRP1 expressed on primary megakaryocytes in murine models. Homozygous knockout for LRP1 constitutively is embryonically lethal and heterozygous deficiency of LRP1 is insufficient to have an observed effect on PF4 biology. We now established a megakaryocyte-specific knockout of LRP1 using a floxed LRP1 mouse previously described by Rohlman et al, mated to the Cre- PF4 promotor-driven Cre recombinase (Cre+) mice previously described by Tiedt et al. Megakaryocytes from mice that were LRP1fl/fl/Cre+ had no detectable LRP1 mRNA or LRP1 surface protein expression by flow cytometry, while LRP1fl/fl/Cre- mice were essentially identical to WT mice. Baseline platelet counts in LRP1fl/fl/Cre+ and LRP1fl/fl/Cre- mice did not different from each other, and there was no difference in bone marrow derived megakaryocyte ploidy. PF4 available in platelet releasate of LRP1fl/fl/Cre+ platelets was also significantly less than in LRP1fl/fl/Cre- platelets (208 ± 42 vs. 362 ± 47 IU/106 platelets, p=0.002) consistent with a role of LRP1 in PF4 reuptake into megakaryocytes in the steady-state and demonstrating that >42% of PF4 may be released during PF4 megakaryopoiesis and requires megakaryocyte LRP1 expression. In siru cultured LRP1fl/fl/Cre+ megakaryocytes exposed to exogenous hPF4 has a lower level of total PF4 levels than LRP1fl/fl/Cre- megakaryocytes (191 ± 7 vs. 236 ± 17 IU/106 cells, respectively (p=0.03)). A similar effect was seen in liquid bone marrow culture assays. Finally, while LRP1fl/fl/Cre+/hPF4+ mice had similar platelet count recovery after irradiation compared to LRP1fl/fl/Cre+/WT mice, treatment of these mice with a heparin-derivative (ODSH) shown to significantly improve platelet count recovery and animal survival in both WT and hPF4+ mice had no effect on either platelet count recovery or animal survival in animals that were also LRP1fl/fl/Cre+. These data demonstrate that nearly half of the total PF4 in megakaryocytes undergoes recycling in vivo and that LRP1 is important for this phenomenon in the steady-state. LRP1 is also important in the negative paracrine effect of PF4 in stress megakaryopoiesis though LRP1 may affect megakaryocyte biology by non-PF4-dependent pathways as well. Whether the two observations – PF4 uptake and negative paracrine effects – are mechanistically related or are distinct LRP1-dependent pathways now needs to be elucidated. Disclosures Xiao: ECRI Institute: Employment.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4949-4949
Author(s):  
Tao Du ◽  
George F. Atweh ◽  
Yelena Galperin ◽  
Rona Singer Weinberg

Abstract Cord blood is being used at an increasing frequency as a source of stem and progenitor cells in human hematopoietic transplantation. However, very little is known about the kinetics of engraftment of cord blood relative to bone marrow derived stem cells. We have used a murine model of newborn/cord blood transplantation to address this question. Our studies demonstrated that murine NB can provide long-term engraftment in primary, secondary, and tertiary transplant recipients. Although NB engrafts more slowly than BM, at one year, engraftment was similar in both types of recipients. We compared the rate of recovery of blood counts, hematopoietic progenitor counts and putative stem cell [SC] counts (i.e. Sca-1+, c-kit+, Lin- cells) in recipients of NB and BM transplantation. Interestingly, the SC ratio in mononuclear cells from donor NB/donor BM was 3.25 ± 0.8 (range = 2.45 – 4.75, n=15). Similarly, donor NB mononuclear cells contained approximately 30% of the number of megakaryocytic progenitors, 12% of the myeloid progenitors and 5% of the erythroid progenitors of donor BM mononuclear cells. The repopulation kinetics in recipients of donor BM and NB transplantation were analyzed at regular intervals, up to 8 months after transplantation. During the first two weeks, NB recipients had lower hemoglobin, WBC and platelet counts than BM recipients. However, by 1 month, the hemoglobin and WBC counts were at similar levels in NB and BM recipients. In contrast, NB transplantation recipients required 2 to 3 months to achieve platelet counts similar to those in BM recipients. These results are reminiscent of the well-known delayed platelet recovery following human cord blood transplantation. Progenitor cell counts in the bone marrow of recipients paralleled the hematological recovery described above. At 2 weeks post-transplantation, progenitor counts of all lineages in NB recipients were 25 to 35% of those in BM recipients. By 1 month, erythroid and myeloid progenitor numbers were similar in NB and BM recipients. In contrast, the appearance of megakaryocytic progenitors was delayed following NB transplantation and did not reach the same level as BM recipients until about 2 months after transplantation. During recovery, the number of SC in the bone marrow of both types of recipients increased gradually over time. At one month after transplantation, the number of SC in BM recipients was significantly greater than that in NB recipients, with a ratio of 4.2 ± 0.2. This SC ratio decreased gradually during the next several months. At 2, 4, and 8 months following transplantation, the ratios of SC in BM recipients/NB recipients were 3.5 ± 0.4, 2.6 ± 0.5, and 2.2 ± 0.3, respectively (n=5). This gradual decrease in the ratio of BM/NB SC suggests that NB SC increase more rapidly than BM SC. In conclusion, these data demonstrate that the kinetics of hematopoietic and stem cell recovery following NB and BM transplantation are significantly different. Although hematopoietic recovery after NB transplantation is slow at first, final engraftment is similar following NB and BM transplantation. Furthermore, the number of SC in NB recipients increases at a faster rate than the number of SC in BM recipients. These differences in SC recovery may be a reflection of differences either in the homing capacity or in the functional maturity of NB relative to BM SC. Further investigation is required to distinguish between these two possibilities.


Blood ◽  
1996 ◽  
Vol 88 (3) ◽  
pp. 778-784 ◽  
Author(s):  
A Ohwada ◽  
S Rafii ◽  
MA Moore ◽  
RG Crystal

Thrombopoietin (TPO, c-mpl ligand) has emerged as a major hematopoietic cytokine stimulating megakaryocyte proliferation, endomitosis, and platelet production. This study shows that a single administration of an adenovirus (Ad) vector encoding TPO (AdCMV.TPO) abrogates thrombocytopenia induced in mice by carboplatin and irradiation. Normal Balb/c mice receiving the vector had increased platelet counts peaking at 7 days and returning to baseline by day 15. Mice rendered pancytopenic with 500 rads and 1.2 mg of carboplatin had a nadir platelet count of five percent of the baseline. Mice receiving AdCMV.TPO 3 days before receiving irradiation and chemotherapy achieved a platelet nadir fourfold higher, and had significant reduction in duration of thrombocytopenia, than mice receiving the control Ad vector. Introduction of AdCMV.TPO the same day of chemotherapy and irradiation was equally effective in acceleration of platelet recovery, but administration of AdCMV.TPO 3 days after chemotherapy-radiation had little effect on platelet recovery. At 30 days after therapy bone marrow and spleen of mice treated with AdCMV.TPO were populated with a large number of polyploid megakaryocytes, but there was no evidence of circulating megakaryocytes in the liver or lungs and no pathologic bone abnormalities such as osteosclerosis or myelofibrosis. These observations suggest that an Ad vector may be an excellent delivery system to provide adequate TPO production to maintain platelet levels in circumstances associated with life-threatening thrombocytopenia.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 725-725
Author(s):  
Michele Lambert ◽  
Alisa Reznikov ◽  
Yvonne Nguyen ◽  
Lubica Rauova ◽  
Mortimer Poncz

Abstract Abstract 725 Platelet factor 4 (PF4) is a chemokine found almost exclusively in megakaryocytes and platelets. PF4 has been previously shown to be a negative paracrine inhibiting megakaryopoiesis in vitro in humans and mice. We confirmed this finding and also found an inverse correlation between PF4 and steady-state platelet counts in mice. In both chemotherapy- and radiation-induced thrombocytopenia, platelet PF4 levels inversely correlated with platelet count recovery after bone marrow injury, and blocking this effect ameliorated the thrombocytopenia. We now asked whether platelet PF4 levels are of clinical relevance on human platelet biology in patients undergoing chemotherapy. We selected pediatric patients who had completed treatment for standard risk acute lymphoblastic leukemia at the Children's Hospital of Philadelphia, as this was a fairly large population that have reached remission after relatively uniform therapy. Enrolled patients had completed therapy since January 1999. Blood samples were obtained and medical records were retrospectively reviewed for platelet counts, platelet transfusion requirements and duration of therapy during delayed intensification (DI). DI was chosen for investigation as a preliminary study showed that 35% of our patients require platelet transfusion during DI and need for transfusion at that point in therapy is unlikely to be related to primary underlying bone marrow disease. To date, 68 subjects have been enrolled. Sixty-two subjects had evaluable PF4 levels. PF4 levels were independent of age and sex. Leukemia survivors did not have significantly different PF4 levels when compared to a pediatric control population. There was a direct relationship between measured total PF4 level and platelet count (Pearson r 0.36, p<0.006) although contrary to animal studies, there was no correlation between PF4 per platelet and platelet count. Transfusion data from the first 22 patients have been evaluated. Patients who did not require platelet transfusion during DI had markedly lower PF4 per platelet (6.35 ± 1.85 SE) when compared to patients who required transfusion (13.26 ± 1.89 SE, p<0.02). In addition, duration of therapy (for girls) was inversely correlated with PF4 per platelet (r 0.689, p=0.04), consistent with animal data in which platelet count recovery was inversely correlated with PF4 per platelet. These data suggest that PF4 may be an important in vivo regulator of human platelet counts in the setting of bone marrow injury. Further clinical studies will confirm these findings and begin to explore potential interventions to allow for intensified chemotherapy regimens in subjects at risk for more severe chemotherapy-induced thrombocytopenia based on their level of this negative paracrine of megakaryopoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (4) ◽  
pp. 1153-1160 ◽  
Author(s):  
Michele P. Lambert ◽  
Lubica Rauova ◽  
Matthew Bailey ◽  
Martha C. Sola-Visner ◽  
M. Anna Kowalska ◽  
...  

Abstract Platelet factor 4 (PF4) is a negative regulator of megakaryopoiesis in vitro. We have now examined whether PF4 regulates megakaryopoiesis in vivo by studying PF4 knockout mice and transgenic mice that overexpress human (h) PF4. Steady-state platelet count and thrombocrit in these animals was inversely related to platelet PF4 content. Growth of megakaryocyte colonies was also inversely related to platelet PF4 content. Function-blocking anti-PF4 antibody reversed this inhibition of megakaryocyte colony growth, indicating the importance of local PF4 released from developing megakaryocytes. The effect of megakaryocyte damage and release of PF4 on 5-fluorouracil–induced marrow failure was then examined. Severity of thrombocytopenia and time to recovery of platelet counts were inversely related to initial PF4 content. Recovery was faster and more extensive, especially in PF4-overexpressing mice, after treatment with anti-PF4 blocking antibodies, suggesting a means to limit the duration of such a chemotherapy-induced thrombocytopenia, especially in individuals with high endogenous levels of PF4. We found that approximately 8% of 250 healthy adults have elevated (> 2 times average) platelet PF4 content. These individuals with high levels of platelet PF4 may be especially sensitive to developing thrombocytopenia after bone marrow injury and may benefit from approaches that block the effects of released PF4.


Blood ◽  
2001 ◽  
Vol 97 (10) ◽  
pp. 3300-3302 ◽  
Author(s):  
Claire Pouplard ◽  
Sophie Iochmann ◽  
Benoit Renard ◽  
Olivier Herault ◽  
Philippe Colombat ◽  
...  

Abstract The pathogenesis of thrombosis in heparin-induced thrombocytopenia (HIT) was studied by investigating whether antibodies to heparin-platelet factor 4 (H-PF4) induced tissue factor (TF) synthesis by monocytes. Plasma from 5 patients with HIT containing IgG to H-PF4 was incubated with peripheral blood mononuclear cells without or with purified PF4 and heparin. Significant TF-dependent procoagulant activity (PCA) expressed by monocytes, measured with a factor Xa-based chromogenic assay, was induced after incubation of each HIT plasma sample. This monocyte PCA required the presence of PF4 and was inhibited by high concentrations of heparin. Furthermore, purified HIT IgG added to whole blood with PF4 and heparin also provoked significant synthesis of TF mRNA by monocytes, demonstrated by RT-PCR, and this effect was not observed with normal IgG. These findings strongly support the hypothesis that antibodies to PF4 developed in HIT trigger the production of tissue factor by monocytes, and this effect could account in vivo for hypercoagulability and thrombotic complications in affected patients.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2598-2598
Author(s):  
Laura F Newell ◽  
Hu Xie ◽  
John M. Pagel ◽  
Ravinder K Sandhu ◽  
Pamela S Becker ◽  
...  

Abstract Abstract 2598 Background: After initial induction chemotherapy for acute myeloid leukemia (AML), it is commonplace that reinduction or intensified therapy is not indicated if the bone marrow has <5% blasts, even despite persistently low neutrophil (ANC) and/or platelet counts. This practice suggests that complete remission (CR), i.e. ANC >1000/μl and platelet count >100,000/μl per standard criteria (Cheson BD, et al. J Clin Oncol. 1990;8(5):813-9), might still occur and that the lack of blood count recovery may not bear prognostic significance. However, the time to CR after the first induction has been shown to be inversely related to subsequent duration of disease-free survival (DFS) and survival (OS), independent of age, treatment, and cytogenetics (Estey EH, et al. Blood. 2000;95(1):72-7). Additionally, the level of ANC and platelet recovery at time of CR is prognostic, with significantly better DFS among patients with higher counts (Yanada M, et al. Leuk Res. 2008;32(10):1505-9). Newly-diagnosed AML patients often present with below normal neutrophil and platelet counts, suggesting that persistence of such cytopenias after induction may be a clinical indicator of minimal residual disease (MRD) in the marrow. We therefore examined whether blood count recovery affected the probability of subsequent CR in patients with <5% bone marrow blasts. Methods: We included 85 patients who, by day 21 or thereafter of induction therapy for newly-diagnosed AML, had not met blood count criteria for CR despite a bone marrow in the prior week with <5% blasts by morphology. Patients were classified by type of induction therapy based on cytarabine dosing. G-CSF was not systematically administered. Marrows were planned for day 21 after chemotherapy and/or weekly thereafter to assess for disease status and evidence of marrow recovery. Because patients were often managed as outpatients, counts and marrows were not uniformly available and thus “day 28” included days 21–28, “day 35” included days 29–35, etc. If a patient had more than one marrow evaluation after day 21, we included only the first one. Results: Overall cohort CR rate was 64%. Eventual CR rate was significantly affected by platelet count, with 44% eventual CR for patients with platelets <30,000, 66% CR for platelets 30,000–100,000, and 95% CR for platelets >100,000. The effect of ANC recovery on eventual CR was less dramatic, with an OR 0.4 (0.2–1.0, p=0.049), for ANC <0.1 vs. >0.1 in the univariate analysis. By day 28, patients with either ANC or platelet recovery were significantly more likely to obtain CR than patients with neither count recovery (89% vs. 51%), OR 8.05 (2.2–30, p=0.002). In the multivariate analysis, (a) lack of platelet recovery to >30,000 was associated with significantly lower incidence of CR, OR 0.26 (0.1–0.8, p=0.02), and was independent of cytogenetic risk, antecedent hematologic disorder, and induction regimen, and (b) there was a suggested association between earlier count recovery and CR (>28 days vs. day 21–28), OR 0.31 (0.1–1.0, p=0.051). Conclusion: Persistence of low peripheral blood counts, despite the presence of <5% bone marrow blasts, is predictive of low eventual CR rates after induction chemotherapy. These results suggest that initiation of further and possibly different therapy, rather than continued observation, should be investigated in this setting. Disclosures: Becker: Sanofi: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document