Network


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

Hotspot


Dive into the research topics where Alison Walker is active.

Publication


Featured researches published by Alison Walker.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine

William Blum; Ramiro Garzon; Rebecca B. Klisovic; Sebastian Schwind; Alison Walker; Susan Geyer; Shujun Liu; Violaine Havelange; Heiko Becker; Larry J. Schaaf; Jon Mickle; Hollie Devine; Cheryl Kefauver; Steven M. Devine; Kenneth K. Chan; Nyla A. Heerema; Clara D. Bloomfield; Michael R. Grever; John C. Byrd; Miguel A. Villalona-Calero; Carlo M. Croce; Guido Marcucci

A phase II clinical trial with single-agent decitabine was conducted in older patients (≥60 years) with previously untreated acute myeloid leukemia (AML) who were not candidates for or who refused intensive chemotherapy. Subjects received low-dose decitabine at 20 mg/m2 i.v. over 1 h on days 1 to 10. Fifty-three subjects enrolled with a median age of 74 years (range, 60–85). Nineteen (36%) had antecedent hematologic disorder or therapy-related AML; 16 had complex karyotypes (≥3 abnormalities). The complete remission rate was 47% (n = 25), achieved after a median of three cycles of therapy. Nine additional subjects had no morphologic evidence of disease with incomplete count recovery, for an overall response rate of 64% (n = 34). Complete remission was achieved in 52% of subjects presenting with normal karyotype and in 50% of those with complex karyotypes. Median overall and disease-free survival durations were 55 and 46 weeks, respectively. Death within 30 days of initiation of treatment occurred in one subject (2%), death within 8 weeks in 15% of subjects. Given the DNA hypomethylating effect of decitabine, we examined the relationship of clinical response and pretreatment level of miR-29b, previously shown to target DNA methyltransferases. Higher levels of miR-29b were associated with clinical response (P = 0.02). In conclusion, this schedule of decitabine was highly active and well tolerated in this poor-risk cohort of older AML patients. Levels of miR-29b should be validated as a predictive factor for stratification of older AML patients to decitabine treatment.


Leukemia | 2012

DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia.

Klaus H. Metzeler; Alison Walker; Susan Geyer; Ramiro Garzon; Rebecca B. Klisovic; Clara D. Bloomfield; William Blum; Guido Marcucci

DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia


Blood | 2012

Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia.

Parvathi Ranganathan; Xueyan Yu; Caroline Na; Ramasamy Santhanam; Sharon Shacham; Michael Kauffman; Alison Walker; Rebecca B. Klisovic; William Blum; Michael A. Caligiuri; Carlo M. Croce; Guido Marcucci; Ramiro Garzon

Chromosome maintenance protein 1 (CRM1) is a nuclear export receptor involved in the active transport of tumor suppressors (e.g., p53 and nucleophosmin) whose function is altered in cancer because of increased expression and overactive transport. Blocking CRM1-mediated nuclear export of such proteins is a novel therapeutic strategy to restore tumor suppressor function. Orally bioavailable selective inhibitors of nuclear export (SINE) that irreversibly bind to CRM1 and block the function of this protein have been recently developed. Here we investigated the antileukemic activity of KPT-SINE (KPT-185 and KPT-276) in vitro and in vivo in acute myeloid leukemia (AML). KPT-185 displayed potent antiproliferative properties at submicromolar concentrations (IC50 values; 100-500 nM), induced apoptosis (average 5-fold increase), cell-cycle arrest, and myeloid differentiation in AML cell lines and patient blasts. A strong down-regulation of the oncogene FLT3 after KPT treatment in both FLT3-ITD and wild-type cell lines was observed. Finally, using the FLT3-ITD-positive MV4-11 xenograft murine model, we show that treatment of mice with oral KPT-276 (analog of KPT-185 for in vivo studies) significantly prolongs survival of leukemic mice (P < .01). In summary, KPT-SINE are highly potent in vitro and in vivo in AML. The preclinical results reported here support clinical trials of KPT-SINE in AML.


Clinical Cancer Research | 2013

Targeted Delivery of microRNA-29b by Transferrin-Conjugated Anionic Lipopolyplex Nanoparticles: A Novel Therapeutic Strategy in Acute Myeloid Leukemia

Xiaomeng Huang; Sebastian Schwind; Bo Yu; Ramasamy Santhanam; Hongyan Wang; Pia Hoellerbauer; Alice S. Mims; Rebecca B. Klisovic; Alison Walker; Kenneth K. Chan; William Blum; Danilo Perrotti; John C. Byrd; Clara D. Bloomfield; Michael A. Caligiuri; Robert J. Lee; Ramiro Garzon; Natarajan Muthusamy; Ly James Lee; Guido Marcucci

Purpose: miR-29b directly or indirectly targets genes involved in acute myeloid leukemia (AML), namely, DNMTs, CDK6, SP1, KIT, and FLT3. Higher miR-29b pretreatment expression is associated with improved response to decitabine and better outcome in AML. Thus, designing a strategy to increase miR-29b levels in AML blasts may be of therapeutic value. However, free synthetic miRs are easily degraded in bio-fluids and have limited cellular uptake. To overcome these limitations, we developed a novel transferrin-conjugated nanoparticle delivery system for synthetic miR-29b (Tf-NP-miR-29b). Experimental Design: Delivery efficiency was investigated by flow cytometry, confocal microscopy, and quantitative PCR. The expression of miR-29b targets was measured by immunoblotting. The antileukemic activity of Tf-NP-miR-29b was evaluated by measuring cell proliferation and colony formation ability and in a leukemia mouse model. Results: Tf-NP-miR-29b treatment resulted in more than 200-fold increase of mature miR-29b compared with free miR-29b and was approximately twice as efficient as treatment with non-transferrin–conjugated NP-miR-29b. Tf-NP-miR-29b treatment significantly downregulated DNMTs, CDK6, SP1, KIT, and FLT3 and decreased AML cell growth by 30% to 50% and impaired colony formation by approximately 50%. Mice engrafted with AML cells and then treated with Tf-NP-miR-29b had significantly longer survival compared with Tf-NP-scramble (P = 0.015) or free miR-29b (P = 0.003). Furthermore, priming AML cell with Tf-NP-miR-29b before treatment with decitabine resulted in marked decrease in cell viability in vitro and showed improved antileukemic activity compared with decitabine alone (P = 0.001) in vivo. Conclusions: Tf-NP effectively delivered functional miR-29b, resulting in target downregulation and antileukemic activity and warrants further investigation as a novel therapeutic approach in AML. Clin Cancer Res; 19(9); 2355–67. ©2013 AACR.


Blood | 2012

Clinical and pharmacodynamic activity of bortezomib and decitabine in acute myeloid leukemia

William Blum; Sebastian Schwind; Somayeh S. Tarighat; Susan Geyer; Ann-Kathrin Eisfeld; Susan P. Whitman; Alison Walker; Rebecca B. Klisovic; John C. Byrd; Ramasamy Santhanam; Hongyan Wang; John Curfman; Steven M. Devine; Samson T. Jacob; Celia Garr; Cheryl Kefauver; Danilo Perrotti; Kenneth K. Chan; Clara D. Bloomfield; Michael A. Caligiuri; Michael R. Grever; Ramiro Garzon; Guido Marcucci

We recently reported promising clinical activity for a 10-day regimen of decitabine in older AML patients; high miR-29b expression associated with clinical response. Subsequent preclinical studies with bortezomib in AML cells have shown drug-induced miR-29b up-regulation, resulting in loss of transcriptional activation for several genes relevant to myeloid leukemogenesis, including DNA methyltransferases and receptor tyrosine kinases. Thus, a phase 1 trial of bortezomib and decitabine was developed. Nineteen poor-risk AML patients (median age 70 years; range, 32-84 years) enrolled. Induction with decitabine (20 mg/m(2) intravenously on days 1-10) plus bortezomib (escalated up to the target 1.3 mg/m(2) on days 5, 8, 12, and 15) was tolerable, but bortezomib-related neuropathy developed after repetitive cycles. Of previously untreated patients (age ≥ 65 years), 5 of 10 had CR (complete remission, n = 4) or incomplete CR (CRi, n = 1); 7 of 19 overall had CR/CRi. Pharmacodynamic analysis showed FLT3 down-regulation on day 26 of cycle 1 (P = .02). Additional mechanistic studies showed that FLT3 down-regulation was due to bortezomib-induced miR-29b up-regulation; this led to SP1 down-regulation and destruction of the SP1/NF-κB complex that transactivated FLT3. This study demonstrates the feasibility and preliminary clinical activity of decitabine plus bortezomib in AML and identifies FLT3 as a novel pharmacodynamic end point for future trials.


Journal of Clinical Oncology | 2010

Dose Escalation of Lenalidomide in Relapsed or Refractory Acute Leukemias

William Blum; Rebecca B. Klisovic; Heiko Becker; Xiaoxia Yang; Darlene M. Rozewski; Mitch A. Phelps; Ramiro Garzon; Alison Walker; Jason Claud Chandler; Susan P. Whitman; John Curfman; Shujun Liu; Larry J. Schaaf; Jon Mickle; Cheryl Kefauver; Steven M. Devine; Michael R. Grever; Guido Marcucci; John C. Byrd

PURPOSE Lenalidomide is effective in myeloma and low-risk myelodysplastic syndromes with deletion 5q. We report results of a phase I dose-escalation trial of lenalidomide in relapsed or refractory acute leukemia. PATIENTS AND METHODS Thirty-one adults with acute myeloid leukemia (AML) and four adults with acute lymphoblastic leukemia (ALL) were enrolled. Lenalidomide was given orally at escalating doses of 25 to 75 mg daily on days 1 through 21 of 28-day cycles to determine the dose-limiting toxicity (DLT) and maximum-tolerated dose (MTD), as well as to provide pharmacokinetic and preliminary efficacy data. RESULTS Patients had a median age of 63 years (range, 22 to 79 years) and a median of two prior therapies (range, one to four therapies). The DLT was fatigue; 50 mg/d was the MTD. Infectious complications were frequent. Plasma lenalidomide concentration increased proportionally with dose. In AML, five (16%) of 31 patients achieved complete remission (CR); three of three patients with cytogenetic abnormalities achieved cytogenetic CR (none with deletion 5q). Response duration ranged from 5.6 to 14 months. All responses occurred in AML with low presenting WBC count. No patient with ALL responded. Two of four patients who received lenalidomide as initial therapy for AML relapse after allogeneic transplantation achieved durable CR after development of cutaneous graft-versus-host disease, without donor leukocyte infusion. CONCLUSION Lenalidomide was safely escalated to 50 mg daily for 21 days, every 4 weeks, and was active with relatively low toxicity in patients with relapsed/refractory AML. Remissions achieved after transplantation suggest a possible immunomodulatory effect of lenalidomide, and results provide enthusiasm for further studies in AML, either alone or in combination with conventional agents or other immunotherapies.


Blood | 2013

Lenalidomide-mediated enhanced translation of C/EBPα-p30 protein up-regulates expression of the antileukemic microRNA-181a in acute myeloid leukemia

Christopher Hickey; Sebastian Schwind; Hanna S. Radomska; Adrienne M. Dorrance; Ramasamy Santhanam; Anjali Mishra; Yue-Zhong Wu; Houda Alachkar; K. Maharry; Nicolet D; Krzysztof Mrózek; Alison Walker; Anna M. Eiring; Susan P. Whitman; Heiko Becker; Danilo Perrotti; Lai-Chu Wu; Xiaobin Zhao; Todd A. Fehniger; Ravi Vij; John C. Byrd; William Blum; Ly James Lee; Michael A. Caligiuri; Clara D. Bloomfield; Ramiro Garzon; Guido Marcucci

Recently, we showed that increased miR-181a expression was associated with improved outcomes in cytogenetically normal acute myeloid leukemia (CN-AML). Interestingly, miR-181a expression was increased in CN-AML patients harboring CEBPA mutations, which are usually biallelic and associate with better prognosis. CEBPA encodes the C/EBPα transcription factor. We demonstrate here that the presence of N-terminal CEBPA mutations and miR-181a expression are linked. Indeed, the truncated C/EBPα-p30 isoform, which is produced from the N-terminal mutant CEBPA gene or from the differential translation of wild-type CEBPA mRNA and is commonly believed to have no transactivation activity, binds to the miR-181a-1 promoter and up-regulates the microRNA expression. Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPα-p30 isoform, resulting in higher miR-181a levels. In xenograft mouse models, ectopic miR-181a expression inhibits tumor growth. Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. This regulatory pathway may explain an increased sensitivity to apoptosis-inducing chemotherapy in subsets of AML patients. Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPα-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy.


Leukemia | 2013

Increased anti-leukemic activity of decitabine via AR-42-induced upregulation of miR-29b: a novel epigenetic-targeting approach in acute myeloid leukemia.

Alice S. Mims; Alison Walker; Xiaomeng Huang; Jin Sun; Hongyan Wang; Ramasamy Santhanam; Adrienne M. Dorrance; Christopher J. Walker; Pia Hoellerbauer; Somayeh S. Tarighat; Kenneth K. Chan; Rebecca B. Klisovic; Danilo Perrotti; Michael A. Caligiuri; John C. Byrd; Ching-Shih Chen; L. James Lee; Samson T. Jacob; Krzysztof Mrózek; Clara D. Bloomfield; William Blum; Ramiro Garzon; Sebastian Schwind; Guido Marcucci

Histone deacetylase (HDAC) inhibitors either alone or in combination with hypomethylating agents have limited clinical effect in acute myeloid leukemia (AML). Previously, we demonstrated that AML patients with higher miR (microRNA)-29b expression had better response to the hypomethylating agent decitabine. Therefore, an increase in miR-29b expression preceding decitabine treatment may provide a therapeutic advantage. We previously showed that miR-29b expression is suppressed by a repressor complex that includes HDACs. Thus, HDAC inhibition may increase miR-29b expression. We hypothesized that priming AML cells with the novel HDAC inhibitor (HDACI) AR-42 would result in increased response to decitabine treatment via upregulation of miR-29b. Here, we show that AR-42 is a potent HDACI in AML, increasing miR-29b levels and leading to downregulation of known miR-29b targets (that is, SP1, DNMT1, DNMT3A and DNMT3B). We then demonstrated that the sequential administration of AR-42 followed by decitabine resulted in a stronger anti-leukemic activity in vitro and in vivo than decitabine followed by AR-42 or either drug alone. These preclinical results with AR-42 priming before decitabine administration represent a promising, novel treatment approach and a paradigm shift with regard to the combination of epigenetic-targeting compounds in AML, where decitabine has been traditionally given before HDACIs.


Leukemia | 2016

The dual epigenetic role of PRMT5 in acute myeloid leukemia: gene activation and repression via histone arginine methylation.

Somayeh S. Tarighat; Ramasamy Santhanam; David Frankhouser; Radomska Hs; H Lai; Mirela Anghelina; Hengbin Wang; Xiaomeng Huang; Lapo Alinari; Alison Walker; Michael A. Caligiuri; Carlo M. Croce; Linsen Li; Ramiro Garzon; Chenglong Li; Robert A. Baiocchi; Guido Marcucci

Changes in the enzymatic activity of protein arginine methyltransferase (PRMT) 5 have been associated with cancer; however, the protein’s role in acute myeloid leukemia (AML) has not been fully evaluated. Here, we show that increased PRMT5 activity enhanced AML growth in vitro and in vivo while PRMT5 downregulation reduced it. In AML cells, PRMT5 interacted with Sp1 in a transcription repressor complex and silenced miR-29b preferentially via dimethylation of histone 4 arginine residue H4R3. As Sp1 is also a bona fide target of miR-29b, the miR silencing resulted in increased Sp1. This event in turn led to transcription activation of FLT3, a gene that encodes a receptor tyrosine kinase. Inhibition of PRMT5 via sh/siRNA or a first-in-class small-molecule inhibitor (HLCL-61) resulted in significantly increased expression of miR-29b and consequent suppression of Sp1 and FLT3 in AML cells. As a result, significant antileukemic activity was achieved. Collectively, our data support a novel leukemogenic mechanism in AML where PRMT5 mediates both silencing and transcription of genes that participate in a ‘yin-yang’ functional network supporting leukemia growth. As FLT3 is often mutated in AML and pharmacologic inhibition of PRMT5 appears feasible, the PRMT5–miR-29b–FLT3 network should be further explored as a novel therapeutic target for AML.


Cancer Investigation | 2008

Profound Hypogammaglobulinemia 7 Years after Treatment for Indolent Lymphoma

Alison Walker; Anatole Kleiner; Lynn Rich; Charlene Conners; Richard I. Fisher; Jennifer H. Anolik; Jonathan W. Friedberg

We report a case of a previously healthy 31 year old woman diagnosed with Stage IIIA follicular lymphoma treated with fludarabine in combination with cyclophosphamide and rituximab who presented seven years after the completion of therapy with CD4 count depression, panhypogammaglobulinema and a history of recurrent sinus infections. We performed comprehensive immunophenotypic analysis and found her to have only 1.2% switched (normal 21 ± 8%) and 4.7% non-switched (11 ± 4%) memory B cells with 92% of B cells belonging to the naïve compartment. We have previously evaluated reconstitution of the B cell compartment during the 6 to 12 month recovery period after treatment with rituximab and found a similar immunophenotypic pattern. In our patient, this defect was observed seven years after the administration of rituximab in combination with an alkylating agent and purine analog. The patient was started on monthly intravenous immunoglobulin treatments with complete resolution of her symptoms.

Collaboration


Dive into the Alison Walker's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge