Insights into Therapeutic Mechanisms: Measuring Immature Platelet Fraction (IPF) Describes Response to Treatment in Immune Thrombocytopenic Purpura (ITP).

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1070-1070 ◽  
Author(s):  
Beth Psaila ◽  
Glynis Villarica ◽  
James B. Bussel

Abstract ITP is characterized by accelerated platelet destruction and also decreased platelet production. The inability to reliably quantify both platelet antibodies and platelet turnover has made it difficult to assess mechanisms of therapeutic effect with certainty. Two widely used therapies, IVIG and IV anti-D, are believed to primarily increase platelets by blocking Fcγ mediated platelet destruction; a similar effect in the marrow may also alter platelet production. Treatments for ITP are expanding as two thrombopoeitic agents (AMG 531 and Eltrombopag) complete phase 3 trials and a specific FcγRIII blocking agent (GMA161) is in phase I. One measure of thrombopoiesis is to use RNA-binding fluorochromes to identify RNA present in newly made, ‘reticulated’ platelets using flow cytometers. The Sysmex XE-2100 enables rapid and fully automated quantification of the absolute immature platelet fraction (IPF), equivalent to enumerating reticulated platelets. This study uses the IPF to assess acute response to treatment in 16 patients with ITP, providing insight into mechanisms of effect (table 1). Table 1: IPF & Platelet Acute Response to Treatment Patient Treatment Initial Plts Max Post-Treatment Plts Increase (Plts) Initial IPF Max Post-Treatment IPF Increase (IPF) E/P BSM = Eltrombopag vs placebo blind RCT study medication. Plts = platelet count 1 IVIG 1 121 120 0.7 33.5 32.8 2 IVIG 1 210 209 0.4 12 11.6 3 IVIG 10 157 147 1.7 8 6.3 4 IVIG 15 58 43 2.1 2.3 0.2 5 IVIG 7 171 164 1.5 6 4.5 6 IVIG 21 136 115 1.4 3.1 1.7 7 IVIG + Anti-D 3 160 157 1.8 17.3 15.5 8 Anti-D 7 335 328 2.8 8.4 5.6 9 Anti-D 9 79 70 2 5.5 3.5 10 Anti-D 1 11 10 0 3.9 3.9 11 Anti-D 13 167 154 2 5 3 12 GMA 14 108 94 3.4 13 9.6 13 GMA 7 45 38 1.2 5.1 3.9 14 Rituxan 24 187 163 6.6 14.4 7.8 15 E/P BSM 13 113 100 7.4 66.4 59 16 E/P BSM 16 558 542 5.9 29.4 23.5 In 4/6 patients treated with IVIG (example fig. 1), 4/4 patients treated with anti-D, and 2/2 GMA-treated patients, the IPF did not change appreciably while the platelet counts dramatically increased, substantiating the Fc inhibition mechanism of effect. In 3 patients with large platelet increases (120–209K) in response to IVIG or IVIG plus anti-D, the IPF also substantially increased, demonstrating that IVIG +/− anti-D may increase platelet production in certain cases. All had low pre-treatment IPF and platelet counts, suggesting antibody-mediated inhibition of platelet production. The two patients on the Eltrombopag/placebo blind RCT (example fig. 2) had 2 of the 3 largest increases in IPF, 23.5 and 59 × 10^3/uL. Using IPF to estimate platelet production provides novel insight into pathophysiology and mechanisms of effect of treatments in ITP. The dramatic increase in the absolute IPF seen with the thrombopoietic agent helps validate the clinical utility of this tool and allows discrimination of ITP patient heterogeneity. Fig. 1 (Patient 3) Response to treatment with IVIG Fig. 1. (Patient 3) Response to treatment with IVIG Fig. 2 (Patient 15): Response to Eltrombopag study medication Fig. 2. (Patient 15): Response to Eltrombopag study medication

Blood ◽  
2011 ◽  
Vol 117 (21) ◽  
pp. 5723-5732 ◽  
Author(s):  
Sarah J. Barsam ◽  
Bethan Psaila ◽  
Marc Forestier ◽  
Lemke K. Page ◽  
Peter A. Sloane ◽  
...  

Abstract This study investigated the immature platelet fraction (IPF) in assessing treatment effects in immune thrombocytopenia (ITP). IPF was measured on the Sysmex XE2100 autoanalyzer. The mean absolute-IPF (A-IPF) was lower for ITP patients than for healthy controls (3.2 vs 7.8 × 109/L, P < .01), whereas IPF percentage was greater (29.2% vs 3.2%, P < .01). All 5 patients with a platelet response to Eltrombopag, a thrombopoietic agent, but none responding to an anti-FcγRIII antibody, had corresponding A-IPF responses. Seven of 7 patients responding to RhoD immuneglobulin (anti-D) and 6 of 8 responding to intravenous immunoglobulin (IVIG) did not have corresponding increases in A-IPF, but 2 with IVIG and 1 with IVIG anti-D did. This supports inhibition of platelet destruction as the primary mechanism of intravenous anti-D and IVIG, although IVIG may also enhance thrombopoiesis. Plasma glycocalicin, released during platelet destruction, normalized as glycocalicin index, was higher in ITP patients than controls (31.36 vs 1.75, P = .001). There was an inverse correlation between glycocalicin index and A-IPF in ITP patients (r2 = −0.578, P = .015), demonstrating the relationship between platelet production and destruction. Nonresponders to thrombopoietic agents had increased megakaryocytes but not increased A-IPF, suggesting that antibodies blocked platelet release. In conclusion, A-IPF measures real-time thrombopoiesis, providing insight into mechanisms of treatment effect.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3288-3288 ◽  
Author(s):  
Robert Makar ◽  
Olga S. Zhukov ◽  
Mervyn A. Sahud ◽  
David J. Kuter

Abstract Abstract 3288 INTRODUCTION: Thrombopoietin (TPO) is the major regulator of platelet production. In prior clinical studies, thrombopoietin levels have been shown to vary inversely with circulating platelet mass and with the rate of platelet production. Thus, TPO levels may help distinguish between the various disorders of thrombocytopenia. In addition, the introduction of TPO agonists has created an interest in predicting the response of patients to these agents. Determining TPO levels may help predict such treatment responses. METHODS: Sera from 121 patients with a history of abnormal platelet counts were tested using a novel, commercially available ELISA assay that measures TPO levels. The TPO assay detected TPO levels as low as 7 pg/mL and was linear for levels up to 2000 pg/mL. The coefficient of variation ranged from 27% near the lower limit of detection to 9% at a TPO concentration of 669 pg/mL. The reference range for TPO was established in serum samples from 118 apparently healthy individuals (58 males and 60 females) and was 7–99 pg/mL. The Wilcoxon test was used to compare continuous variables and the Fisher's exact test was used to compare categorical variables. RESULTS: The patient population included 40 patients with a consumptive thrombocytopenia (38 with primary or secondary immune thrombocytopenic purpura (ITP), 2 with thrombotic thrombocytopenic purpura), 34 patients with myeloproliferative disorders (23 with essential thrombocytosis, 9 with polycythemia vera, 2 with an ill-defined myeloproliferative disorder), and 47 patients with hypoproliferative thrombocytopenia (29 with chemotherapy-related thrombocytopenia, 19 with primary or secondary bone marrow failure syndromes). Among the 38 patients with ITP, 11 were taking TPO agonists (9 on romiplostim, 2 on eltrombopag), 19 were taking immunomodulatory agents (16 on steroids alone or in combination with other therapies, 2 on azathioprine, 1 on danazol), and 12 were off ITP-specific therapy when the TPO level was measured. 9 out of 38 (24%) patients with ITP had undergone splenectomy and/or been previously treated with rituximab. The median serum TPO level in patients with consumptive thrombocytopenia was 64.5 pg/mL (interquartile range, 48.5–97.5 pg/mL) and the corresponding median platelet count was 68,000/μL (interquartile range, 27,000–144,500) (Figure). While patients with myeloproliferative disorders had similar TPO levels [median 87.0 pg/mL (38.0–125.5)], their platelet counts were significantly higher than those of patients with consumptive thrombocytopenia [median 549,500/mL (431,250–693,000] (P <0.0001). Contrastingly, comparable platelet counts [median 61,000/μL (31,000–118,000)] were observed among patients with hypoproliferative thrombocytopenia, but serum TPO levels were significantly higher than those of patients with consumptive thrombocytopenia [844 pg/mL (409.5–1551.5), P <0.0001]. Among 22 evaluable patients meeting diagnostic criteria for primary or secondary ITP who had taken a TPO agonist for at least 1 month, serum TPO levels appeared to predict responsiveness to the drug. A clinical response to a TPO agonist was defined as achieving a platelet count ≥50,000/μL after starting the drug and maintaining it at or above that count in ≥50% of subsequent complete blood counts from initiation until discontinuation of the drug, loss to follow-up, or 6 months had passed, whichever was longest, without the need for recurrent rescue therapy. Whereas 14 out of 16 (88%) ITP patients with a TPO level <99 pg/mL met our definition for a clinical response to treatment with a TPO agonist, only 1 out of 6 patients (17%) with a TPO level >99 pg/mL responded (P <0.005 for the difference in clinical response to TPO agents.) CONCLUSIONS: TPO levels may have diagnostic utility in discriminating between patients with hypoproliferative and consumptive thrombocytopenia. High TPO levels among patients with ITP may predict a poor clinical response to treatment with TPO agonists. Further studies are required to confirm these data. Disclosures: Zhukov: Quest Diagnostics: Employment. Sahud:Quest Diagnostics: Employment. Kuter:Quest Diagnostics: Consultancy, Research Funding.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1085-1085
Author(s):  
Koji Miyazaki ◽  
Miyako Taira ◽  
Tomiteru Togano ◽  
Manabu Ohsaka ◽  
Yuhko Suzuki ◽  
...  

Abstract It is sometimes confusing to distinguish idiopathic thrombocytopenic purpura (ITP) from thrombocytopenia due to dysmegakaryopoiesis, as seen in myelodysplastic syndrome (MDS) patients, especially MDS with isolated thrombocytopenia. In this study, we investigated the useful parameters for the different diagnosis of thrombocytopenia. The number of reticulated platelets reflects the rate of thrombopoiesis, and this clinical utility has been established in the laboratory diagnosis of thrombocytopenia due to increased peripheral platelet destruction, such as autoimmune thrombocytopenic purpura (AITP). However, the number of reticulated platelets has not been well investigated in the patients with myelodysplatsic syndrome (MDS), while some of them are misdiagnosed as ITP. The aim of this study is to evaluate the diagnostic utility of the measurements of reticulated platelets as well as other parameters of platelets, such as MPV (mean platelet volume), P-LCR (platelet larger cell ratio) and PDW (platelet distribution width). The reticulated platelets, expressed as the immature platelet fraction (IPF) were determined in 108 ITP and 57 MDS patients using the Sysmex XE-2100 blood cell counter with upgraded software (Sysmex, Kobe, Japan). This system enabled rapid, inexpensive, automated, stable measurements of reticulated platelets compared with the flow cytometry system, of which consensus method has not yet been identified to provide acceptable intra- and inter-laboratory results. The platelet counts in ITP and MDS patients were equivalent (ITP, 7.99 ± 0.40 × 104/μL; MDS, 8.05 ± 0.57× 104/μL). The IPF values in ITP patients (10.4 ± 0.61%) were significantly higher than those in MDS (5.82 ± 0.63%), and the inverse correlation between the IPF and the platelet counts was observed among the ITP patients, but not among the MDS. Both MPV and PDW in MDS (10.6 +/− 0.15 fL and 12.2+/−0.41 fL, respectively) were significantly higher than in ITP (7.7 +/− 0.38 fL and 9.4 +/− 0.48 fL, respectively), while P-LCR in MDS (28.7 +/− 1.2%) and ITP (23.6 +/− 1.3%) were not significantly different. Although MPV was correlated with IPF among either group, the correlation between IPF and either PDW or P-LCR was weak among MDS (IPF × PDW, r=0.673; IPF × P-LCR, r=0.660) compared with ITP (IPF × PDW, r=0.779; IPF × P-LCR, r=0.803). Next we precisely investigated the clinical features of the minor population of MDS with higher IPF. Most of these patients revealed the significantly higher values of PAIgG (Platelet-associated IgG) and/or poor response to the blood transfusion, suggesting the possibility of associated autoimmune mechanisms. The patients of MDS in overt leukemic stage also recorded higher IPF even if they had no or few blood transfusion. The IPF would be a useful parameter to distinguish ITP from MDS with isolated thrombocytopenia, which has been shown to have a favorable prognosis.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4980-4980
Author(s):  
Ondrej Zapletal ◽  
Jan Blatny ◽  
Michaela Selingerova ◽  
Jiri Jarkovsky ◽  
Miroslav Penka

Abstract Abstract 4980 Background Myeloproliferative disorders (MPD) in general result from proliferation of a clone of myeloid cells derived from a neoplastic pluripotent precursor or from connective tissue elements in bone marrow. This leads to increased numbers in one or more blood cell lines in peripheral blood. Some MPDs can be associated with thrombocytosis (MPD-T): essential thrombocythaemia (ET), polycythaemia vera (PV) and early stages of chronic idiopathic myelofibrosis (CIMF). Usually in MPD-T the thrombocytosis is caused by increased platelet production from proliferating mature megakaryocytes, especially in ET. Elevated platelet counts in these patients are often associated with both thromboembolic events and bleeding. One of the goals of MPD treatment is the control of platelet count. Immature platelets mirror the platelet production in bone marrow. In certain automated blood count analyzers it is possible to measure Immature platelet fraction (IPF) from the routine CBC samples sent to haematology lab. Reference range for IPF parameter for method used in this study is 1,1-6,1%. Objective Measurement of IPF parameter by fully automated analyzer (XE-2100, Sysmex, Kobe, Japan) in optical channel and analyzing it ( software IPF Master) in patients treated for Ph-negative MPD. We enrolled 85 pts- 67 ET (79%), 10PV(12%) and 8 CIMF (9%) patients; 57 were women and 28 men; median age 56 years ranging from 20 up to 83 years. We analyzed and evaluated IPF in whole group as well as in subgroups depending on diagnosis, gender, age, JAK-2 mutation and platelet count. Results At the time of assessment the majority of our pts were already commenced on treatment for their MPD. Platelet counts (plt) in whole cohort ranged from 164 up to 2148 ×109/l, with median 374 ×109/l. Thirty eight pts (45%) had plt < 350 ×109/l. Plt <450 ×109/l (WHO 2008 recommended cut off level for thrombocytosis) were found in 59 pts (69%). IPF median in whole cohort was 5,9% (0,7-14,4%). When comparing IPF in subgroups mentioned above statistically significant differences (p<0,05) was found only between subgroups with normal and abnormal plt counts: IPF median 7,45% (0,8-14,4%) resp. 4,6% (0,7-11,9%), (p=0,002) and between subgroups with less and more than 450×109/l plt (IPF median 6,6% (0,8-14,4%) resp. 3,85% (0,7-9,6%), (p<0,001). Fact, that patients with higher plt had lower IPF and vice versa, was confirmed also by Spearman correlation coefficient. When correlating results of plt a IPF in the whole cohort, we found the trend to indirect dependence (rs= –0,386). Conclusions MPD-T patients in our cohort did not have marked elevation of IPF parameter, neither those with high platelet counts (so far untreated pts). Thus we can speculate, that increased number of Plts in peripheral blood is caused by increased number of mature megacaryocytes in bone marrow which produce adequate numbers of platelets without increase of immature fraction rather than increased number of immature platelets released from megacaryocytes appearing in normal numbers in bone marrow Thus it seems that negative feedback of increased Plt counts on releasing of Plts from megacaryocytes is maintained also in patients with MPD-T. Further assessment is needed and should further determine, what is the clinical relevance (if any) of measurement of IPF in patients treated for MPD-T. Disclosures Blatny: Sysmex, Czech Republic: Consultancy.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1024-1024
Author(s):  
Emily M. Harris ◽  
Michele P. Lambert ◽  
Jenny M. Despotovic ◽  
Susan E Kirk ◽  
Abinaya Arulselvan ◽  
...  

Abstract Background: As the treatment options for immune thrombocytopenia (ITP) continue to expand, the choice of which treatment to give to an individual patient has become increasingly complex. Therefore, a laboratory marker to help guide treatment selection would be clinically useful. The immature platelet fraction (IPF), measured clinically by automated Sysmex hematologic analyzers, correlates with bone marrow thrombopoietic activity and may correlate with disease activity in patients with ITP. Objective: To investigate the relationship between pre-treatment immature platelet fraction (IPF) and treatment response among pediatric patients with ITP. Methods: This is an observational cohort study of 148 patients with ITP who received ITP-directed treatment as monotherapy at 3 tertiary academic children's hospitals. Eligibility included a clinical diagnosis of ITP, ITP-directed surgical or pharmacologic treatment given as monotherapy, and available pre-treatment IPF values. The Sysmex XN-series measures IPF by adopted fluorescence flow cytometry using a semiconductor dioxide laser to measure platelets stained with oxazine fluorescent dyes. Demographic and clinical characteristics, laboratory studies, and treatments were collected. Response to treatment was defined as a platelet count ≥30 x 10 9/L and at least 2-fold increase from the baseline platelet count within 3 weeks of first dose of IVIG, Rh(D) immune globulin, or corticosteroid or within 3 months of all other therapies. For second-line treatments, platelet counts within 1 month after a rescue therapy were excluded. Regression analysis was utilized to estimate association between variables; estimated coefficients and p values are reported. Results: The cohort included 148 patients, 52% (n=77) of whom were female. Median age of diagnosis was 8 years (IQR: 3-13). Twenty percent (n=29) of treated patients had secondary ITP including 9 (6%) with Evans syndrome. Median platelet count at time of diagnosis was 5 x 10 9/L, and median IPF at diagnosis was 16.7% (IQR: 7.7-25.8). Median pre-treatment platelet count was 17 x 10 9/L with a median pre-treatment IPF of 16.6% (IQR: 10.0-25.7). There was a significant association between pre-treatment platelet count and pre-treatment IPF (coefficient -0.176, p = 0.003). Increased variation in IPF was seen at lower platelet counts compared to higher platelet counts (p=0.014, Figure 1a). IPF at diagnosis and pre-treatment IPF were not correlated with platelet response to treatment overall (p=0.28 for pre-treatment IPF, p=0.31 for IPF at diagnosis). IPF prior to treatment did not correlate with platelet response to individual medications: IVIG (coefficient 0.001, p=0.78, n=64), corticosteroids (coefficient 0.007, p=0.28, n=43), Rituximab (coefficient -0.01, p=0.52, n=10), and thrombopoietin receptor agonists (coefficient 0.0006, p=0.93, n=26, Figure 1b). Similarly, IPF at diagnosis of ITP did not correlate with platelet response to individual medications. Conclusions: Pre-treatment platelet count and IPF are inversely correlated in children with ITP, although there is significant variability in IPF at low platelet counts. The IPF at ITP diagnosis and prior to treatment does not correlate with platelet response to the most common ITP-directed treatments in children. A normal or elevated IPF should not impact decision-making about initiation of any specific ITP-directed treatments, including thrombopoietin receptor agonists or immunosuppressive therapy, in children with ITP. Figure 1 Figure 1. Disclosures Lambert: Principia: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer: Consultancy; Sysmex: Research Funding; Astra Zeneca: Research Funding; Dova: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; PDSA: Research Funding; Octapharma: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; ClinGen, ISTH, ASH, GW University: Honoraria; Rigel: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Argenx: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Shionogi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Despotovic: Apellis: Consultancy; UpToDate: Patents & Royalties: Royalties; Novartis: Consultancy, Research Funding; Agios: Consultancy. Kirk: Biomarin: Honoraria. Grace: Agios: Research Funding; Novartis: Research Funding; Dova: Membership on an entity's Board of Directors or advisory committees, Research Funding; Principia: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3199-3199
Author(s):  
Marjorie L. Zucker ◽  
Barry S. Skikne ◽  
Jane M. Rachel ◽  
Carol A. Murphy ◽  
Gregory A. Martinez ◽  
...  

Abstract Thrombocytopenia is a frequent complication in patients with hepatitis C. Possible mechanisms include hypersplenism, immune-mediated platelet destruction, and drug-induced impairment of platelet production. The immature platelet fraction (IPF), determined on an automated blood cell analyzer (Sysmex XE-2100), has been shown to be useful in differentiating consumptive and aplastic causes of thrombocytopenia. We studied 31 patients known to have hepatitis C, 21 with thrombocytopenia (platelet count <100,000/uL) and 10 with normal platelet counts (>140,000/uL). None of the patients was taking interferon. Blood samples were drawn for CBC with platelet count, IPF (%), and thrombopoietin (TPO) assay (Quantikine TPO ELISA). The presence or absence of splenomegaly and liver function test results were noted. In the thrombocytopenic group IPF was elevated above the upper limit of the reference range (>7.1%) in 12/21 (57%) patients, suggestive of peripheral platelet consumption. TPO levels (known to correlate inversely with platelet/megakaryocyte mass) were within the normal range (<146pg/mL) in 11/12 (92%) of these patients, implying adequate megakaryocyte mass, which is further evidence of peripheral platelet consumption in this group. Splenomegaly was present in 8 of these 12 patients, suggesting that hypersplenism may be the mechanism for thrombocytopenia in this sub-group. IPF was within normal limits in 9/21 (43%) of the thrombocytopenic patients, implying either decreased platelet production or ineffective thrombopoiesis as the major cause of thrombocytopenia in this group. TPO levels were within the normal range in 8/9 (89%) of these patients, again implying adequate megakaryocyte mass, and suggesting that ineffective or impaired thrombopoiesis, rather than decreased platelet production, may be the major mechanism for thrombocytopenia in this group. Splenomegaly was present in 7 of these 9 patients, thus hypersplenism cannot be ruled out as at least a contributory factor in this sub-group (in spite of normal IPF values). One patient in this group had an elevated TPO level, suggestive of megakaryocyte hypoplasia. In the group of patients with normal platelet counts IPF was within normal limits in 8/10 patients, and minimally elevated in the other 2 patients. TPO levels were within normal limits in this entire group, and no patients in this group had splenomegaly. In conclusion, our findings suggest that peripheral platelet consumption is a major cause of thrombocytopenia in patients with hepatitis C (at least 57% of patients in this study), predominantly secondary to hypersplenism. Other possible causes of peripheral platelet consumption (in those without splenomegaly) include immune-mediated platelet destruction secondary to antibodies, immune complexes or drugs. In the remaining patients (43% in this study) ineffective thrombopoiesis may be the major mechanism of thrombocytopenia, possibly related to the effect of virus, cytokines, drugs or antibodies on thrombopoiesis, in the presence of adequate megakaryocytes in the marrow. Decreased platelet production (megakaryocyte hypoplasia) appears to be an unusual mechanism of thrombocytopenia in these patients.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2103-2103
Author(s):  
Amanda Prislovsky ◽  
Falk Nimmerjahn ◽  
Jeffrey V. Ravetch ◽  
Carl W. Jackson ◽  
Ted S. Strom

Abstract Patients with the Wiskott-Aldrich Syndrome (WAS) have thrombocytopenia and increased platelet consumption rates, and may have reduced platelet production rates. WASP(−) mice have been reported to have only a mild thrombocytopenia. We find that when fully crossed onto the C57Bl/6J background, WASP(−) mice have a >50% reduction in platelet counts. Ex vivo labeled WASP(−)platelets are consumed 2 to 4-fold faster than are WT platelets in WT mice, and with exponential rather than linear kinetics. Clearance rates of WASP(−) platelets in WT mice, and vice versa, indicate that rapid consumption is due to factors both intrinsic and extrinsic to platelets. In vivo biotinylation demonstrates consumption rates comparable to those seen with ex vivo labeling, and shows a normal rate of consumption of WASP(−) reticulated (immature) platelets. Reticulated platelet counts are reduced, indicating that their production rate is reduced. Megakaryocytes are increased in spleen and bone marrow, and in the latter their ploidy distribution is normal, suggesting that impaired platelet production occurs at the level of thrombopoiesis. The absolute turnover rates of mature and reticulated platelets, however, indicate that maturation of the latter can account for only a fraction of the former’s turnover in either WT or WASP(−) mice. A subset of WASP(−) mice show an increased fraction of reticulated platelets and more severe thrombocytopenia, and some members of this subset also express serum anti-platelet antibodies. CMFDA-labeled WASP(−) platelets opsonized with anti-CD61(IgG1) antibody are more susceptible to ex vivo phagocytosis by bone marrow derived macrophages (BMDM) than WT platelets, and as susceptible as CD47(−/−) platelets. After opsonization with 6A6(IgG2b) antibody, WASP(−) platelets are also taken up more rapidly than WT platelets by BMDM, but less rapidly than are CD47(−/−) platelets. The in vivo consumption rate of WASP(−) platelets in WT recipients is more accelerated by opsonization with anti-CD61 antibody than is that of WT platelets. Increased phagocytosis is not due to (A) altered levels of the targeted antigens or of CD47 (B) increased exposure of phosphatidyl serine, or (C) antibody-induced activation as assayed by CD62P expression. Increased phagocytosis is not due to selective sensitivity to IgG1 vs IgG2b antibodies, as opsonization with an engineered 6A6(IgG1) antibody leads to reduced phagocytosis for both CD47(−/−) and WASP(−) platelets. Anti-CD61 opsonized platelets deficient in both WASP and CD47 show markedly increased ex vivo phagocytosis compared to platelets deficient in either protein, suggesting that platelet WASP does not function to amplify signals from platelet CD47 through macrophage SIRP-alpha. These results raise the possibility that the binding of low affinity or low titer antibodies that might have no effect on WT platelets could cause thrombocytopenia when the platelets lack WASP. Alternatively, rapid phagocytosis of opsonized WASP(−) platelets could promote a self-reinforcing cycle of increased host antigen presentation and increased immune responses to host antigens.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2513-2513
Author(s):  
Sarah J Barsam ◽  
James B. Bussel ◽  
Peter A Sloane

Abstract Abstract 2513 The pathogenesis of ITP involves accelerated platelet destruction and decreased platelet production. The immature platelet fraction (IPF) from the Sysmex XE2100 autoanalyzer is a marker of platelet reticulocytes, so we used it to explore the mechanisms of effect of treatments of ITP. First, 67 patients with ITP were compared to 100 healthy adult controls to test the Absolute IPF (A-IPF) as a marker of thrombopoiesis. A second study investigated the plasma glycocalicin levels, a soluble external portion of membrane glycoprotein 1bα, in conjunction with A-IPF and IPF% to assess platelet turnover and survival. The mean baseline A-IPF in both ITP study groups (2.8 ×109/L & 8.67 ×109/L ) were substantially and significantly lower than the controls (7.80 ×109/L & 10.55 ×109/L ) but the IPF% were markedly higher (28.20 ×109/L & 20.83 ×109/L vs 3.20 ×109/L & 4.55 ×109/L. In the first study, A-IPF was further validated clinically by comparing responses to a thrombopoietic agent (TPO-A) and GMA161 (an anti-FcγRIII antibody, inhibiting platelet destruction). A platelet count increment >30 ×109/L or >doubling of baseline and an A-IPF increment >10× 109/L were defined as responses. In the TPO-A sub-group, 19/34 patients had platelet and A-IPF responses; 6/34 patients had platelet responses but no A-IPF response; 8/34 had no platelet or A-IPF responses; 1/34 had an A-IPF response but no platelet response (figure). With GMA161, 2/5 patients had a platelet response without an A-IPF response; 1/5 patients had a response in both platelet count and A-IPF; 2/5 patients had no platelet or A-IPF response. Almost all increases of A-IPF >10×109 and all increases >20×109 were only seen with TPO-A (figure).Figure:Maximum A-IPF Results for Platelet Responders (black diamonds) and Platelet Non-responders (gray squares) in ITP following therapeutic interventions.Figure:. Maximum A-IPF Results for Platelet Responders (black diamonds) and Platelet Non-responders (gray squares) in ITP following therapeutic interventions. These results showed, as expected, that platelet responses to a TPO-A were usually associated with increased A-IPF, whereas those with an inhibitor of FcγR-mediated destruction were usually not. The effects of IV anti-D (Winrho) and IVIG in ITP were studied. 8/8 patients treated with IV anti-D had a platelet response without any A-IPF response. 10/19 patients treated with IVIG had a platelet response without a response in A-IPF, however, 4/19 had platelet and (small) A-IPF responses (figure). These results were consistent when the same patient was studied on different episodes. In a separate study, IPF findings were compared to simultaneous samples of plasma glycocalicin levels (Beer, Blood 1994) in 21 patients. Glycocalicin Index (GCI) is a measure normalized for the platelet count. Mean plasma glycocalicin level was marginally higher in ITP patients than controls: 1.86mcg/ml (0.52–3.91) vs 1.6 mcg/ml (0.79–2.53). However, mean GCI was dramatically higher in the ITP group: 31.36 (0.368–229) vs 1.75 (0.72–2.84). For the separate treatment groups, the mean plasma glycocalicin was greatest in TPO-A treated patients and the mean GCI was greatest in IVIG & prednisolone treated patients. This illustrates the relative importance of effective platelet production and platelet turnover in these therapies. There was a statistically significant (p<0.05) inverse correlation between GCI and platelet count in ITP and there was a positive correlation between plasma glycocalicin and A-IPF, suggesting that the A-IPF was a valid marker of platelet turnover. In summary, the results of A-IPF in ITP patients compared to healthy adult controls and in ITP patients treated with TPO-A and GMA161, demonstrate the utility of this measurement in assessing thrombopoiesis and the mechanism of treatment effect in patients with ITP. Although IVIG and IV anti-D have differing interactions with FcγR, the A-IPF results support the primary effect of anti-D and IVIG to be inhibition of platelet destruction. A small group of patients mounted both platelet and A-IPF increments to IVIG, suggesting that IVIG could also cause an increased effective platelet production. Finally, results with GCI, in a smaller number of patients, further confirmed the validity of the IPF. In the future, A-IPF could be used diagnostically (not explored in this study), for assessing response to treatment, and potentially to select the most effective treatment in individuals. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
pp. 1-6
Author(s):  
Masuho Saburi ◽  
Masao Ogata ◽  
Yasuhiro Soga ◽  
Takako Satou ◽  
Kazuhito Itani ◽  
...  

<b><i>Objective:</i></b> Platelet-associated immunoglobulin G (PA-IgG) refers to IgG attached to the surface of platelets, while the immature platelet fraction (IPF) reflects the state of platelet production in bone marrow. Since PA-IgG and IPF are increased in patients with immune thrombocytopenia (ITP), reflecting amounts of platelet antibodies and compensatory platelet production, respectively, we hypothesized that these laboratory findings may provide useful markers for predicting treatment response in patients with ITP. We therefore retrospectively investigated associations between levels of these markers at diagnosis and response to first-line therapy in patients with ITP. <b><i>Methods:</i></b> Forty-three patients diagnosed with ITP at Oita Kouseiren Tsurumi Hospital between May 2010 and November 2018 were included. Patients were divided into 2 groups based on response to corticosteroid as first-line therapy. Laboratory findings were compared between responders and nonresponders. <b><i>Results:</i></b> Median PA-IgG was 285 ng/10<sup>7</sup> cells (range, 45.5–18,200 ng/10<sup>7</sup> cells), and median IPF was 15.5% (range, 5.4–62.1%). Median levels were higher than the respective upper limits of normal range (PA-IgG, 0–46 ng/10<sup>7</sup> cells; IPF, 1.1–9.5%). First-line therapy was performed using standard-dose prednisolone (0.5–1.0 mg/kg/day) in 32 patients and high-dose dexamethasone (40 mg/day, 4 days) or methylprednisolone (125–1,000 mg/day, 3–4 days) in 11 patients. Twenty-four patients (55.8%) responded to first-line therapy. In univariate analysis, type of corticosteroid (<i>p</i> = 0.17) tended to differ between groups but did not differ significantly, and no difference in IPF level was apparent between responders (15.35%; range, 5.4–41.5%) and nonresponders (16.7%; range, 6.3–62.1%; <i>p</i> = 0.15). PA-IgG was significantly higher among nonresponders (430 ng/10<sup>7</sup> cells; range, 101–18,200 ng/10<sup>7</sup> cells) than among responders (254.5 ng/10<sup>7</sup> cells; range, 45.5–470 ng/10<sup>7</sup> cells; <i>p</i> = 0.004). Multivariate analysis revealed PA-IgG was independently associated with response to first-line therapy (odds ratio, 1.000; 95% confidence interval, 1.000–1.010; <i>p</i> = 0.029). <b><i>Conclusion:</i></b> Our data suggested that PA-IgG at diagnosis could offer a useful predictor of response to first-line corticosteroid therapy for ITP.


2010 ◽  
Vol 103 (05) ◽  
pp. 1016-1021 ◽  
Author(s):  
Hannes Hammer ◽  
Christoph Bührer ◽  
Christof Dame ◽  
Malte Cremer ◽  
Andreas Weimann

SummaryNewly released platelets, referred to as immature platelets, can be reliably quantified based on their RNA content by flow cytometry in an automated blood analyser. The absolute number of immature platelets (IPF#) and the immature platelet fraction (IPF%) reflect megakaryopoietic activity. We aimed to analyse the implication of these parameters in analysing the pathomechanism of early-onset neonatal thrombocytopenia. Platelet counts and IPF were determined at day 1 to 3 (d1 to d3) in 857 neonates admitted to intensive care. In thrombocytopenic patients (platelet counts<150 x 109/l, n=129), IPF# was significantly lower (8.5 ± 2.7 x 109/l), than in non-thrombocytopenic neonates (9.5 ± 3.6 x 109/l, n=682, p<0.05). IPF% was significantly higher in thrombocytopenic (9.3 ± 7.9%) vs. non-thrombocytopenic neonates (4.1 ± 1.8%, p<0.001). While neonates with early-onset infection (n=134) had lower platelet counts (199 ± 75 x 109/l) compared to controls (230 ± 68 x 109/l, n=574, p<0.01), there were no differences in IPF# or IPF%. Likewise, “small for gestational age” infants (SGA, n=149) had lower platelet counts at d1 (199 ± 75 x 109/l, p<0.001) than controls, but no differences in IPF. A trend towards lower IPF# was detected if SGA infants with platelet counts <100 x 109/l (5.4 ± 3.9 x 109/l, n=11) and thrombocytopenic neonates with infection (9.9 ± 7.3 x 109/l, n=10, p=0.11) were compared. The evaluation of IPF# indicates that thrombocytopenia in neonates is likely due to a combination of increased platelet consumption and inadequate megakaryopoietic response by the neonatal bone marrow. Furthermore, SGA neonates with moderate and severe thrombocytopenia might have a pronounced suppression of megakaryopoiesis compared to neonates with infection.


Sign in / Sign up

Export Citation Format

Share Document