Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Iskra Pusic is active.

Publication


Featured researches published by Iskra Pusic.


Biology of Blood and Marrow Transplantation | 2008

Impact of Mobilization and Remobilization Strategies on Achieving Sufficient Stem Cell Yields for Autologous Transplantation

Iskra Pusic; Shi Yuan Jiang; Scott Landua; Geoffrey L. Uy; Michael P. Rettig; Amanda F. Cashen; Peter Westervelt; Ravi Vij; Camille N. Abboud; Keith Stockerl-Goldstein; Diane Sempek; A. Smith; John F. DiPersio

The purpose of this article was to examine historic institutional autologous stem cell mobilization practices and evaluate factors influencing mobilization failure and kinetics. In this retrospective study we analyzed clinical records of 1834 patients who underwent stem cell mobilization for autologous transplantation from November 1995 to October 2006 at the Washington University in St. Louis. Successful mobilization was defined as collection of > or =2 x 10(6) CD34(+) cells/kg. From 1834 consecutive patients, 1040 met our inclusion criteria (502 non-Hodgkins lymphoma [NHL], 137 Hodgkins lymphoma, and 401 multiple myeloma [MM]). A total of 976 patients received granulocyte colony-stimulating factor (G-CSF) and 64 received G-CSF plus chemotherapy (G/C) for the initial mobilization. Although the median CD34(+) cell yield was higher in G/C group than in G-CSF alone group, the failure rates were similar: 18.8% and 18.6%, respectively. Overall, 53% of patients collected > or =2 x 10(6) CD34(+) cells/kg during the first apheresis with either mobilization regimen. Regardless of mobilization regimen used, MM patients had the highest total CD34(+) cell yield and required less aphereses to collect > or =2 x 10(6) CD34(+) cells/kg. Mobilized, preapheresis, peripheral blood CD34(+) count correlated with first day apheresis yield (r = .877, P < .001) and 20 cells/microL was the minimum threshold needed for a successful day 1 collection. For the remobilization analysis we included patients from the whole database. A total of 269 of 1834 patients underwent remobilization using G/C, G-CSF, and/or GM-CSF, and G-CSF plus plerixafor. Only 23% of remobilized patients achieved > or =2 x 10(6) CD34(+) cells/kg and 29.7% failed to pool sufficient number of stem cells from both collections. Patients receiving G-CSF plus plerixafor had lowest failure rates, P = .03. NHL patients remobilized with G-CSF who waited > or =25 days before remobilization had lower CD34(+) cell yield than those who waited < or =16 days, P = .023. Current mobilization regimens are associated with a substantial failure rate irrespective of underlying disease. Patients who fail initial mobilization are more likely to fail remobilization. These findings suggest that there is a need for more effective first-line mobilization agents.


The New England Journal of Medicine | 2016

TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes

John S. Welch; Allegra A. Petti; Christopher A. Miller; Catrina C. Fronick; Michelle O’Laughlin; Robert S. Fulton; Richard Wilson; Jack Baty; Eric J. Duncavage; Bevan Tandon; Yi-Shan Lee; Lukas D. Wartman; Geoffrey L. Uy; Armin Ghobadi; Michael H. Tomasson; Iskra Pusic; Rizwan Romee; Todd A. Fehniger; Keith Stockerl-Goldstein; Ravi Vij; Stephen T. Oh; Camille N. Abboud; Amanda F. Cashen; Mark A. Schroeder; Meagan A. Jacoby; Sharon Heath; Kierstin Luber; M R Janke; Andrew Hantel; Niloufer Khan

BACKGROUND The molecular determinants of clinical responses to decitabine therapy in patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) are unclear. METHODS We enrolled 84 adult patients with AML or MDS in a single-institution trial of decitabine to identify somatic mutations and their relationships to clinical responses. Decitabine was administered at a dose of 20 mg per square meter of body-surface area per day for 10 consecutive days in monthly cycles. We performed enhanced exome or gene-panel sequencing in 67 of these patients and serial sequencing at multiple time points to evaluate patterns of mutation clearance in 54 patients. An extension cohort included 32 additional patients who received decitabine in different protocols. RESULTS Of the 116 patients, 53 (46%) had bone marrow blast clearance (<5% blasts). Response rates were higher among patients with an unfavorable-risk cytogenetic profile than among patients with an intermediate-risk or favorable-risk cytogenetic profile (29 of 43 patients [67%] vs. 24 of 71 patients [34%], P<0.001) and among patients with TP53 mutations than among patients with wild-type TP53 (21 of 21 [100%] vs. 32 of 78 [41%], P<0.001). Previous studies have consistently shown that patients with an unfavorable-risk cytogenetic profile and TP53 mutations who receive conventional chemotherapy have poor outcomes. However, in this study of 10-day courses of decitabine, neither of these risk factors was associated with a lower rate of overall survival than the rate of survival among study patients with intermediate-risk cytogenetic profiles. CONCLUSIONS Patients with AML and MDS who had cytogenetic abnormalities associated with unfavorable risk, TP53 mutations, or both had favorable clinical responses and robust (but incomplete) mutation clearance after receiving serial 10-day courses of decitabine. Although these responses were not durable, they resulted in rates of overall survival that were similar to those among patients with AML who had an intermediate-risk cytogenetic profile and who also received serial 10-day courses of decitabine. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT01687400 .).


Current Opinion in Hematology | 2010

Update on clinical experience with AMD3100, an SDF-1/CXCL12-CXCR4 inhibitor, in mobilization of hematopoietic stem and progenitor cells.

Iskra Pusic; John F. DiPersio

Purpose of reviewMobilized peripheral blood stem cells are increasingly used for the reconstitution of hematopoiesis in autologous and allogeneic transplants. New agents and approaches are emerging to improve mobilization efficacy while reducing duration and toxicity of mobilization. The purpose of this review is to overview clinical experience with AMD3100 (plerixafor) and its role in stem cell mobilization. Recent findingsAMD3100 is a bicyclam molecule that selectively and reversibly antagonizes the binding of stromal cell-derived factor-1 (SDF-1) to its receptor CXC motif receptor-4 (CXCR4) with subsequent egress of hematopoietic stem cells to the peripheral blood. AMD3100 safely and rapidly mobilizes stem cells in patients with lymphoma, myeloma and healthy donors, and is synergistic in combination with granulocyte-colony stimulating factor. In addition, AMD3100 disrupts the interaction between mouse and human leukemic blasts and the bone marrow stroma, mobilizing blasts to the peripheral blood and sensitizing them to chemotherapy. SummaryAMD3100 was recently FDA-approved for stem cell mobilization in combination with granulocyte-colony stimulating factor in patients with non-Hodgkins lymphoma and multiple myeloma. Studies are underway testing AMD3100 as an adjunct to chemotherapy in patients with refractory acute myelogenous leukemia (and other hematologic malignancies), as a strategy to sensitize leukemic cells to chemotherapy and improve clinical outcomes.


Blood | 2015

Genomic analysis of germ line and somatic variants in familial myelodysplasia/acute myeloid leukemia

Jane E. Churpek; Khateriaa Pyrtel; Krishna L. Kanchi; Jin Shao; Daniel C. Koboldt; Christopher A. Miller; Dong Shen; Robert S. Fulton; Michelle O'Laughlin; Catrina C. Fronick; Iskra Pusic; Geoffrey L. Uy; Evan M. Braunstein; Mark Levis; Julie A. Ross; Kevin Elliott; Sharon Heath; Allan Jiang; Peter Westervelt; John F. DiPersio; Daniel C. Link; Matthew J. Walter; John S. Welch; Richard Wilson; Timothy J. Ley; Lucy A. Godley; Timothy A. Graubert

Familial clustering of myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML) can be caused by inherited factors. We screened 59 individuals from 17 families with 2 or more biological relatives with MDS/AML for variants in 12 genes with established roles in predisposition to MDS/AML, and identified a pathogenic germ line variant in 5 families (29%). Extending the screen with a panel of 264 genes that are recurrently mutated in de novo AML, we identified rare, nonsynonymous germ line variants in 4 genes, each segregating with MDS/AML in 2 families. Somatic mutations are required for progression to MDS/AML in these familial cases. Using a combination of targeted and exome sequencing of tumor and matched normal samples from 26 familial MDS/AML cases and asymptomatic carriers, we identified recurrent frameshift mutations in the cohesin-associated factor PDS5B, co-occurrence of somatic ASXL1 mutations with germ line GATA2 mutations, and recurrent mutations in other known MDS/AML drivers. Mutations in genes that are recurrently mutated in de novo AML were underrepresented in the familial MDS/AML cases, although the total number of somatic mutations per exome was the same. Lastly, clonal skewing of hematopoiesis was detected in 67% of young, asymptomatic RUNX1 carriers, providing a potential biomarker that could be used for surveillance in these high-risk families.


Biology of Blood and Marrow Transplantation | 2015

Measuring Therapeutic Response in Chronic Graft-versus-Host Disease. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease

Stephanie J. Lee; Daniel Wolff; Carrie L. Kitko; John Koreth; Yoshihiro Inamoto; Madan Jagasia; Joseph Pidala; Attilio Olivieri; Paul J. Martin; Donna Przepiorka; Iskra Pusic; Fiona L. Dignan; Sandra A. Mitchell; Anita Lawitschka; David A. Jacobsohn; Anne M. Hall; Mary E.D. Flowers; Kirk R. Schultz; Georgia B. Vogelsang; Steven Z. Pavletic

In 2005, the National Institutes of Health (NIH) Chronic Graft-versus-Host Disease (GVHD) Consensus Response Criteria Working Group recommended several measures to document serial evaluations of chronic GVHD organ involvement. Provisional definitions of complete response, partial response, and progression were proposed for each organ and for overall outcome. Based on publications over the last 9 years, the 2014 Working Group has updated its recommendations for measures and interpretation of organ and overall responses. Major changes include elimination of several clinical parameters from the determination of response, updates to or addition of new organ scales to assess response, and the recognition that progression excludes minimal, clinically insignificant worsening that does not usually warrant a change in therapy. The response definitions have been revised to reflect these changes and are expected to enhance reliability and practical utility of these measures in clinical trials. Clarification is provided about response assessment after the addition of topical or organ-targeted treatment. Ancillary measures are strongly encouraged in clinical trials. Areas suggested for additional research include criteria to identify irreversible organ damage and validation of the modified response criteria, including in the pediatric population.


Current Pharmaceutical Design | 2008

The use of growth factors in hematopoietic stem cell transplantation.

Iskra Pusic; John F. DiPersio

Mobilized, peripheral blood stem cells (PBSC) are increasingly used for both autologous and allogeneic transplants. Granulocyte-colony-stimulating factor is the most widely used cytokine for mobilization. Several different mechanisms of stem cell mobilization have been proposed including protease-dependent and non-protease- dependent mechanisms. In autologous transplants, the addition of chemotherapy to mobilization can enhance the yield of PBSC collected but with substantial adverse effects, and not necessarily faster engraftment. In allogeneic transplants, the use of mobilized PBSC is associated with faster engraftment and donor chimerism compared to bone marrow. In the majority of studies, the rate of acute graft-versus-host disease (GVHD) has not been shown to be significantly higher with PBSC, but the rate of chronic GVHD appears to be increased. Several different strategies have been proposed for patients and donors who fail initial mobilization, including the use of novel agents. AMD3100 (Plerixafor) works by directly inhibiting the interaction between stromal cell-derived factor-1 and its receptor CXCR4, and mobilizes hematopoietic stem cells within hours. It is being studied alone or in conjunction with growth factors for PBSC mobilization in both autologous and allogeneic settings. Although the use of growth factors after PBSC transplantation results in faster neutrophil engraftment its impact on treatment-related mortality and survival does not appear significant. Here, we review the biology and methods of PBSC mobilization, the effect of growth factors on normal donors and the controversies of growth factor use in the post-transplant setting. We also review the data on novel agents for mobilization of stem cells.


Biology of Blood and Marrow Transplantation | 2015

Maintenance Therapy with Decitabine after Allogeneic Stem Cell Transplantation for Acute Myelogenous Leukemia and Myelodysplastic Syndrome

Iskra Pusic; Jaebok Choi; Mark Fiala; Feng Gao; Matthew Holt; Amanda F. Cashen; Ravi Vij; Camille N. Abboud; Keith Stockerl-Goldstein; Meghan A. Jacoby; Geoffrey L. Uy; Peter Westervelt; John F. DiPersio

Decitabine is a hypomethylating agent that irreversibly inhibits DNA methyltransferase I, inducing leukemic differentiation and re-expression of epigenetically silenced putative tumor antigens. We assessed safety and efficacy of decitabine maintenance after allogeneic transplantation for acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Decitabine maintenance may help eradicate minimal residual disease, decrease the incidence of graft-versus-host disease (GVHD), and facilitate a graft-versus-leukemia effect by enhancing the effect of T regulatory lymphocytes. Patients with AML/MDS in complete remission (CR) after allotransplantation started decitabine between day +50 and +100. We investigated 4 decitabine doses in cohorts of 4 patients: 5, 7.5, 10, and 15 mg/m(2)/day × 5 days every 6 weeks, for a maximum 8 cycles. The maximum tolerated dose (MTD) was defined as the maximum dose at which ≤ 25% of people experience dose-limiting toxicities during the first cycle of treatment. Twenty-four patients were enrolled and 22 were evaluable. All 4 dose levels were completed and no MTD was reached. Overall, decitabine maintenance was well tolerated. Grade 3 and 4 hematological toxicities were experienced by 75% of patients, including all patients treated at the highest dose level. Nine patients completed all 8 cycles and 8 of them remain in CR. Nine patients died from relapse (n = 4), infectious complications (n = 3), and GVHD (n = 2). Most occurrences of acute GVHD were mild and resolved without interruption of treatment; 1 patient died of acute gut GVHD. Decitabine maintenance did not clearly impact the rate of chronic GVHD. Although there was a trend of increased FOXP3 expression, results were not statistically significant. In conclusion, decitabine maintenance is associated with acceptable toxicities when given in the post-allotransplantation setting. Although the MTD was not reached, the dose of 10 mg/m(2) for 5 days every 6 weeks appeared to be the optimal dose rather than 15 mg/m(2), where most hematological toxicities occurred.


Biology of Blood and Marrow Transplantation | 2014

Protective effect of cytomegalovirus reactivation on relapse after allogeneic hematopoietic cell transplantation in acute myeloid leukemia patients is influenced by conditioning regimen.

Shivaprasad Manjappa; Pavan Kumar Bhamidipati; Keith Stokerl-Goldstein; John F. DiPersio; Geoffrey L. Uy; Peter Westervelt; Jingxia Liu; Mark A. Schroeder; Ravi Vij; Camille N. Abboud; Todd A. Fehniger; Amanda F. Cashen; Iskra Pusic; Meagan A. Jacoby; Srinidhi J. Meera; Rizwan Romee

Cytomegalovirus (CMV) reactivation after allogeneic hematopoietic cell transplant (allo-HCT) has been associated with a reduced risk of relapse in patients with acute myeloid leukemia (AML). However, the influence of the conditioning regimen on this protective effect of CMV reactivation after allo-HCT is relatively unexplored. To address this, we evaluated the risk of relapse in 264 AML patients who received T cell-replete, 6/6 HLA matched sibling or 10/10 HLA matched unrelated donor transplantation at a single institution between 2006 and 2011. Of these 264 patients, 206 received myeloablative (MA) and 58 received reduced-intensity conditioning (RIC) regimens. CMV reactivation was observed in 88 patients with MA conditioning and 37 patients with RIC. At a median follow-up of 299 days, CMV reactivation was associated with significantly lower risk of relapse in patients who received MA conditioning both in univariate (P = .01) and multivariate analyses (hazard ratio, .5246; P = .006); however, CMV reactivation did not significantly affect the risk of relapse in our RIC cohort. These results confirm the protective effect of CMV reactivation on relapse in AML patients after allo-HCT reported by previous studies but suggest this protective effect of CMV reactivation on relapse is influenced by the conditioning regimen used with the transplant.


Blood | 2017

Ibrutinib for chronic graft-versus-host disease after failure of prior therapy

David B. Miklos; Corey Cutler; Mukta Arora; Edmund K. Waller; Madan Jagasia; Iskra Pusic; Mary E.D. Flowers; Aaron C Logan; Ryotaro Nakamura; Bruce R. Blazar; Yunfeng Li; Stephen Chang; Indu Lal; Jason A. Dubovsky; Danelle F. James; Lori Styles; Samantha Jaglowski

Chronic graft-versus-host disease (cGVHD) is a serious complication of allogeneic stem cell transplantation with few effective options available after failure of corticosteroids. B and T cells play a role in the pathophysiology of cGVHD. Ibrutinib inhibits Bruton tyrosine kinase in B cells and interleukin-2-inducible T-cell kinase in T cells. In preclinical models, ibrutinib reduced severity of cGVHD. This multicenter, open-label study evaluated the safety and efficacy of ibrutinib in patients with active cGVHD with inadequate response to corticosteroid-containing therapies. Forty-two patients who had failed 1 to 3 prior treatments received ibrutinib (420 mg) daily until cGVHD progression. The primary efficacy end point was cGVHD response based on 2005 National Institutes of Health criteria. At a median follow-up of 13.9 months, best overall response was 67%; 71% of responders showed a sustained response for ≥20 weeks. Responses were observed across involved organs evaluated. Most patients with multiple cGVHD organ involvement had a multiorgan response. Median corticosteroid dose in responders decreased from 0.29 mg/kg per day at baseline to 0.12 mg/kg per day at week 49; 5 responders discontinued corticosteroids. The most common adverse events were fatigue, diarrhea, muscle spasms, nausea, and bruising. Plasma levels of soluble factors associated with inflammation, fibrosis, and cGVHD significantly decreased over time with ibrutinib. Ibrutinib resulted in clinically meaningful responses with acceptable safety in patients with ≥1 prior treatments for cGVHD. Based on these results, ibrutinib was approved in the United States for treatment of adult patients with cGVHD after failure of 1 or more lines of systemic therapy. This trial was registered at www.clinicaltrials.gov as #NCT02195869.


Clinical Cancer Research | 2016

A randomized phase II crossover study of imatinib or rituximab for cutaneous sclerosis after hematopoietic cell transplantation

Sally Arai; Joseph Pidala; Iskra Pusic; Xiaoyu Chai; Samantha Jaglowski; Nandita Khera; Jeanne Palmer; George L. Chen; Madan Jagasia; Sebastian Mayer; William A. Wood; Michael Green; Teresa S. Hyun; Yoshihiro Inamoto; Barry E. Storer; David B. Miklos; Howard M. Shulman; Paul J. Martin; Stefanie Sarantopoulos; Stephanie J. Lee; Mary E.D. Flowers

Purpose: Cutaneous sclerosis occurs in 20% of patients with chronic graft-versus-host disease (GVHD) and can compromise mobility and quality of life. Experimental design: We conducted a prospective, multicenter, randomized, two-arm phase II crossover trial of imatinib (200 mg daily) or rituximab (375 mg/m2 i.v. weekly × 4 doses, repeatable after 3 months) for treatment of cutaneous sclerosis diagnosed within 18 months (NCT01309997). The primary endpoint was significant clinical response (SCR) at 6 months, defined as quantitative improvement in skin sclerosis or joint range of motion. Treatment success was defined as SCR at 6 months without crossover, recurrent malignancy or death. Secondary endpoints included changes of B-cell profiles in blood (BAFF levels and cellular subsets), patient-reported outcomes, and histopathology between responders and nonresponders with each therapy. Results: SCR was observed in 9 of 35 [26%; 95% confidence interval (CI); 13%–43%] participants randomized to imatinib and 10 of 37 (27%; 95% CI, 14%–44%) randomized to rituximab. Six (17%; 95% CI, 7%–34%) patients in the imatinib arm and 5 (14%; 95% CI, 5%–29%) in the rituximab arm had treatment success. Higher percentages of activated B cells (CD27+) were seen at enrollment in rituximab-treated patients who had treatment success (P = 0.01), but not in imatinib-treated patients. Conclusions: These results support the need for more effective therapies for cutaneous sclerosis and suggest that activated B cells define a subgroup of patients with cutaneous sclerosis who are more likely to respond to rituximab. Clin Cancer Res; 22(2); 319–27. ©2015 AACR.

Collaboration


Dive into the Iskra Pusic's collaboration.

Top Co-Authors

Avatar

John F. DiPersio

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Peter Westervelt

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Geoffrey L. Uy

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Amanda F. Cashen

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Camille N. Abboud

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ravi Vij

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Mark A. Schroeder

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Keith Stockerl-Goldstein

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Madan Jagasia

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rizwan Romee

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge