151
|
Knapper S. The clinical development of FLT3 inhibitors in acute myeloid leukemia. Expert Opin Investig Drugs 2011; 20:1377-95. [DOI: 10.1517/13543784.2011.611802] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
152
|
Walz C, Erben P, Ritter M, Bloor A, Metzgeroth G, Telford N, Haferlach C, Haferlach T, Gesk S, Score J, Hofmann WK, Hochhaus A, Cross NCP, Reiter A. Response of ETV6-FLT3-positive myeloid/lymphoid neoplasm with eosinophilia to inhibitors of FMS-like tyrosine kinase 3. Blood 2011; 118:2239-42. [PMID: 21705501 DOI: 10.1182/blood-2011-03-343426] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Imatinib-resistant tyrosine kinase (TK) fusions involving FGFR1, JAK2, or FLT3 are rare but recurrent in patients with eosinophilia-associated neoplasms. We report here 2 male patients with ETV6-FLT3(+) myeloid/lymphoid neoplasms with eosinophilia who were treated with the multitargeted TK inhibitors sunitinib and sorafenib. Patient 1 achieved rapid complete hematologic response and complete cytogenetic response after 3 months of taking sunitinib. A secondary blast phase caused by clonal evolution was diagnosed after 6 months. He achieved a second complete hematologic response after taking sorafenib but relapsed 2 months later. An N841K point mutation within the TK domain of FLT3, previously reported in acute myeloid leukemia and potentially conferring resistance to sorafenib, was subsequently identified. Patient 2 was heavily pretreated according to the initial diagnosis of T-lymphoblastic lymphoma and died in sunitinib-induced pancytopenia. This report highlights the importance of a careful diagnostic workup for eosinophilia-associated neoplasms to evaluate the possibility of TK inhibitor therapy.
Collapse
Affiliation(s)
- Christoph Walz
- Pathologisches Institut, Ludwig-Maximilians-Universität, München, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Sharma M, Ravandi F, Bayraktar UD, Chiattone A, Bashir Q, Giralt S, Chen J, Qazilbash M, Kebriaei P, Konopleva M, Andreeff M, Cortes J, McCue D, Kantarjian H, Champlin RE, de Lima M. Treatment of FLT3-ITD-positive acute myeloid leukemia relapsing after allogeneic stem cell transplantation with sorafenib. Biol Blood Marrow Transplant 2011; 17:1874-7. [PMID: 21767516 DOI: 10.1016/j.bbmt.2011.07.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 07/10/2011] [Indexed: 10/17/2022]
Abstract
Patients with acute myeloid leukemia (AML) and internal tandem duplication of FMS-like tyrosine kinase receptor-3 gene (FLT3-ITD) mutation have poor prognoses and are often treated with allogeneic hematopoietic stem cell transplantation (HSCT). Sorafenib, an inhibitor of multiple kinases including FLT3, has shown promising activity in FLT3-ITD-positive AML. We treated 16 patients with FLT3-ITD-positive AML who relapsed after HSCT with sorafenib alone (n = 8) or in combination with cytotoxic chemotherapy (n = 8). The number of circulating blasts decreased in 80% of cases, but none of the patients achieved complete remission (CR); 3 achieved partial remission. Two patients were bridged to a second transplantation but both relapsed within 3 months of the transplantation. Median overall survival (OS) was 83 days, with none surviving more than a year. Sorafenib is not effective in the treatment of FLT3-ITD-positive AML relapsing after HSCT. Preventive strategies after HSCT may be more suitable for these high-risk patients.
Collapse
Affiliation(s)
- Manish Sharma
- Department of Stem Cell Transplantation, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030-4009, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Fathi AT, Chabner BA. FLT3 inhibition as therapy in acute myeloid leukemia: a record of trials and tribulations. Oncologist 2011; 16:1162-74. [PMID: 21765192 DOI: 10.1634/theoncologist.2011-0084] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy with a poor prognosis. Approximately one quarter of the patients with AML also carry an internal tandem duplication (ITD) mutation in the gene encoding FMS-like tyrosine kinase 3 (FLT3), which has a significantly deleterious impact on prognosis. The ITD mutation renders FLT3 constitutively active and leads to uncontrolled proliferation of the leukemic blast. Over the course of the last decade, a variety of compounds have been developed in preclinical and clinical studies as potent inhibitors of FLT3. Many of the earlier agents under investigation, such as lestaurtinib, midostaurin, and sunitinib, were initially developed as inhibitors of other tyrosine kinases and as targeted therapies in a variety of malignancies. These compounds have been demonstrated to have some efficacy in clinical trials of AML, mainly manifesting as transient decreases in circulating blasts correlating with effective in vivo suppression of the FLT3 target. Nevertheless, the cumbersome pharmacokinetics of some compounds and the suboptimal specificity and potency of others have limited their therapeutic efficacy. In the last few years, newer, more potent and specific agents have been under investigation, with the leading example being AC220. This agent has shown significant promise in early phases of clinical investigation, and is currently in more advanced clinical trials. Hope remains that FLT3 inhibition will be become an effective therapeutic adjunct to our current treatment approach to AML.
Collapse
Affiliation(s)
- Amir T Fathi
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | |
Collapse
|
155
|
Wilhelm C, Neubauer A, Burchert A. Poor-risk cytogenetics may be associated with inferior outcome after fludarabine, cytarabine, and amsacrine reduced intensity conditioning in patients with high-risk acute myeloid leukemia. Leuk Lymphoma 2011; 52:2031-5. [PMID: 21702642 DOI: 10.3109/10428194.2011.588760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
156
|
Abstract
PURPOSE OF REVIEW Induction followed by postremission therapy is recognized as the main road toward cure in acute myeloid leukemia (AML). Induction approach has not changed substantially for many years. Despite identifying numerous heterogeneous factors in AML, bench-derived insights are sluggishly translated into clinical protocols. In the current review, advances in intensified dose induction protocols, risk stratification, and disease monitoring are presented. Some promising as well as disappointing agents used for tailoring and targeting therapies in AML are also discussed. RECENT FINDINGS Intensifying anthracycline dose in young AML patients was recently shown to improve induction results. Importantly, maximal doses in induction have been shown to be safe also in older adults, negating a common perception that too often led to attenuation of induction therapy in this population. Novel targeted agents and the explosive number of newly identified prognostic factors or disease-specific biomarkers are promoting AML therapy toward a more personalized future. SUMMARY Survival of AML patients is constantly improving. High doses of 'old drugs' for chemotherapy induction, followed by a postremission combination of chemotherapy and novel targeted agents may hold the key for cure. Disease-specific molecular abnormalities may play a role in monitoring and guiding therapy.
Collapse
|
157
|
Abstract
PURPOSE OF REVIEW Ever since the recognition that FMS-like tyrosine kinase 3 (FLT3) mutations exert a profound negative prognostic impact on the clinical outcome of patients with acute myeloid leukemia (AML), researchers have sought to find effective small-molecule inhibitors of this receptor tyrosine kinase. This review will attempt to provide a survey of the FLT3 inhibitors currently under investigation and provide a discussion on their current status in clinical trials. RECENT FINDINGS Over the past 10 years, a number of different compounds have been studied in vitro and clinically as FLT3 inhibitors. The first inhibitors studied were hampered by cumbersome pharmacokinetics and a general lack of potency. However, some agents have shown promise in clinical trials with transient responses in AML. Newer compounds, such as AC220, have demonstrated profound selectivity and potency against the FLT3 target, and are currently being investigated in clinical trials. SUMMARY Clinical trials have so far demonstrated that inhibitors of FLT3 do have clinical activity in patients with FLT3-mutant AML, although this activity is often transient and correlates with effective in-vivo suppression of the FLT3 target. As newer, more potent agents are now entering advanced clinical trials, opportunities will emerge for real progress against this grim disease.
Collapse
|
158
|
Abstract
Acute myeloid leukemia with a FLT3 internal tandem duplication (FLT3/ITD) mutation is an aggressive hematologic malignancy with a generally poor prognosis. It can be successfully treated into remission with intensive chemotherapy, but it routinely relapses. At relapse, the blasts tend to have higher mutant allelic ratios and, in vitro, are more addicted to the aberrant signaling from the FLT3/ITD oncoprotein. They remain highly responsive to FLT3 ligand, the levels of which rise several-fold during the course of chemotherapy. The question now arises as to whether these high levels of FLT3 ligand are actually promoting relapse, and, if so, how we can use this information to adjust our therapeutic approach and improve the cure rate for acute myeloid leukemia with FLT3/ITD.
Collapse
|
159
|
Zauli G, Bosco R, Secchiero P. Molecular targets for selective killing of TRAIL-resistant leukemic cells. Expert Opin Ther Targets 2011; 15:931-42. [DOI: 10.1517/14728222.2011.580278] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
160
|
Parmar A, Marz S, Rushton S, Holzwarth C, Lind K, Kayser S, Döhner K, Peschel C, Oostendorp RA, Götze KS. Stromal Niche Cells Protect Early Leukemic FLT3-ITD+ Progenitor Cells against First-Generation FLT3 Tyrosine Kinase Inhibitors. Cancer Res 2011; 71:4696-706. [DOI: 10.1158/0008-5472.can-10-4136] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
161
|
Fullmer A, Cortes J, Kantarjian H, Jabbour E. ASH 2010 meeting report-Top 10 clinically oriented abstracts in acute leukemia. Am J Hematol 2011. [DOI: 10.1002/ajh.21998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
162
|
Abstract
PURPOSE OF REVIEW Relapse has become the leading cause of death following allogeneic hematopoietic stem cell transplantation (HSCT). Despite improved understanding of the biology that underlies the graft-versus-leukemia/tumor effect the relapse rate did not decrease over the past 20 years. In general, prognosis is poor for patients who relapsed to an allograft since effective treatment options are limited. Here, we review the available and upcoming treatment approaches for relapse. RECENT FINDINGS Treatment of relapse after allogeneic HSCT has been rarely investigated systematically and results differ substantially from diseases. Withdrawal of immunosuppressive medication, donor lymphocyte infusions with or without chemotherapy and/or second allogeneic HSCT are the most used options. New specific cellular approaches such as disease-specific T-cells, alloreactive natural killer cells or vaccination strategies are under investigation. Novel agents such as tyrosine-kinase inhibitors, hypomethylating agents, monoclonal antibodies, immunomodulating drugs, or proteasome-inhibitors either alone or in combination with adoptive immunotherapy are upcoming promising options, but valid data are lacking so far. SUMMARY With some exceptions (chronic myeloid leukemia), treatment options for patients who relapse are limited. The results are poor and the majority of patients ultimately die of their disease. More effort and research is needed to prevent and treat relapse after allogeneic HSCT.
Collapse
|
163
|
Margolin JF. Molecular diagnosis and risk-adjusted therapy in pediatric hematologic malignancies: a primer for pediatricians. Eur J Pediatr 2011; 170:419-25. [PMID: 21350806 DOI: 10.1007/s00431-011-1424-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 02/08/2011] [Indexed: 12/17/2022]
Abstract
UNLABELLED Progress in the care of the hematologic malignancies of childhoond has been one of the proudest success stories in modern pediatrics. The cure rates of these diseases have improved from essentially zero in the 1950's and early 1960's to cure rates that range from 65%-90% in modern centers. While the largest improvements have been made in the most common (and the lower risk subtypes) of Acute Lymphoblastic Leukemia (ALL), there has also been significant progress in both the higher risk forms of ALL (i.e. Philadelphia chromosome positive, Ph+ ALL) and in Acute Myeloid Leukemia (AML). This progress has been achieved by the careful and stepwise identification of clinical, cytogenetic, molecular, and most recently response-based prognostic criteria, that now allow oncologists to focus the intensity of the therapy more closely to what is required to cure individual subgroups of patients. CONCLUSION Pediatricians need to be familiar with the changes in diagnostic and therapeutic approaches, because these changes have impact on: the laboratory tests that should be ordered at the time of specialist referral; counseling of patients and their families; and with the advent of "shared care models" pediatricians will need to be more involved in the general, supportive, and long-term care of these patients.
Collapse
|
164
|
Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood 2011; 117:3294-301. [PMID: 21270442 PMCID: PMC3069671 DOI: 10.1182/blood-2010-08-301796] [Citation(s) in RCA: 321] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In a randomized trial of therapy for FMS-like tyrosine kinase-3 (FLT3) mutant acute myeloid leukemia in first relapse, 224 patients received chemotherapy alone or followed by 80 mg of the FLT3 inhibitor lestaurtinib twice daily. Endpoints included complete remission or complete remission with incomplete platelet recovery (CR/CRp), overall survival, safety, and tolerability. Correlative studies included pharmacokinetics and analysis of in vivo FLT3 inhibition. There were 29 patients with CR/CRp in the lestaurtinib arm and 23 in the control arm (26% vs 21%; P = .35), and no difference in overall survival between the 2 arms. There was evidence of toxicity in the lestaurtinib-treated patients, particularly those with plasma levels in excess of 20 μM. In the lestaurtinib arm, FLT3 inhibition was highly correlated with remission rate, but target inhibition on day 15 was achieved in only 58% of patients receiving lestaurtinib. Given that such a small proportion of patients on this trial achieved sustained FLT3 inhibition in vivo, any conclusions regarding the efficacy of combining FLT3 inhibition with chemotherapy are limited. Overall, lestaurtinib treatment after chemotherapy did not increase response rates or prolong survival of patients with FLT3 mutant acute myeloid leukemia in first relapse. This study is registered at www.clinicaltrials.gov as #NCT00079482.
Collapse
|
165
|
Prescott H, Kantarjian H, Cortes J, Ravandi F. Emerging FMS-like tyrosine kinase 3 inhibitors for the treatment of acute myelogenous leukemia. Expert Opin Emerg Drugs 2011; 16:407-23. [PMID: 21417961 DOI: 10.1517/14728214.2011.568938] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The FMS-like tyrosine kinase 3 (FLT3) is highly expressed in acute leukemias. Mutations involving FLT3 are among the most common molecular abnormalities in acute myelogenous leukemia (AML). Available evidence suggests that these molecular lesions confer a shorter disease-free survival and overall survival in patients with intermediate-risk cytogenetics. Therefore, substantial interest in FLT3 as a therapeutic target has led to the development of several promising inhibitors that target this tyrosine kinase. AREAS COVERED This review covers the molecular pathways associated with FLT3 activation in patients with AML, the biological rationale for inhibiting FLT3 and recent clinical progress with FLT3 inhibitors for the treatment of AML. Six FLT3 inhibitors undergoing clinical evaluation are discussed. A review of selected published manuscripts on the subject of FLT3 inhibition in AML and a search of the English language manuscripts in PubMed using the index words FLT3 and AML were conducted and articles of interest selected. EXPERT OPINION Mutated forms of FLT3, specifically FLT3-internal tandem duplication, have a significant impact on the prognosis of AML patients, particularly those with a normal karyotype. Inhibiting FLT3 may lead to clinical benefit for patients with AML. Newly developed FLT3 inhibitors have shown encouraging activity as monotherapy and in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Hillary Prescott
- The University of Texas, M.D. Anderson Cancer Center, Department of Leukemia, Houston, USA
| | | | | | | |
Collapse
|
166
|
Krüger WH, Hirt C, Kiefer T, Neumann T, Busemann C, Dölken G. Molecular remission of FLT3-ITD(+) positive AML relapse after allo-SCT by acute GVHD in addition to sorafenib. Bone Marrow Transplant 2011; 47:137-8. [PMID: 21423122 DOI: 10.1038/bmt.2011.7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
167
|
Fesler MJ. Marked bone marrow hypoplasia associated with sorafenib-induced marrow blast clearance in two patients with FLT3-ITD acute myeloid leukemia. Leuk Res 2011; 35:e21-2. [DOI: 10.1016/j.leukres.2010.10.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 10/19/2010] [Accepted: 10/23/2010] [Indexed: 11/28/2022]
|
168
|
Chevallier P, Labopin M, Turlure P, Prebet T, Pigneux A, Hunault M, Filanovsky K, Cornillet-Lefebvre P, Luquet I, Lode L, Richebourg S, Blanchet O, Gachard N, Vey N, Ifrah N, Milpied N, Harousseau JL, Bene MC, Mohty M, Delaunay J. A new Leukemia Prognostic Scoring System for refractory/relapsed adult acute myelogeneous leukaemia patients: a GOELAMS study. Leukemia 2011; 25:939-44. [PMID: 21331073 DOI: 10.1038/leu.2011.25] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A simplified prognostic score is presented based on the multivariate analysis of 138 refractory/relapsed acute myeloid leukaemia (AML) patients (median age 55 years, range: 19-70) receiving a combination of intensive chemotherapy+Gemtuzumab as salvage regimen. Overall, 2-year event-free survival (EFS) and overall survival (OS) were 29±4% and 36±4%, respectively. Disease status (relapse <12 months, including refractory patients), FLT3-ITD-positive status and high-risk cytogenetics were the three strongest independent adverse prognostic factors for OS and EFS in this series. We then defined three subgroups with striking different outcomes at 2 years: no adverse factor (favourable, N=36): OS 58%, EFS 45%; one adverse factor (intermediate, N=54): OS 37%, EFS 31%; two or three adverse factors (poor, N=43): OS 12%, EFS 12% (P<10(-4), P=0.001). This new simplified Leukemia Prognostic Scoring System was then validated on an independent cohort of 111 refractory/relapsed AML patients. This new simplified prognostic score, using three clinical and biological parameters routinely applied, allow to discriminate around two third of the patients who should benefit from a salvage intensive regimen in the setting of refractory/relapsed AML patients. The other one third of the patients should receive investigational therapy.
Collapse
Affiliation(s)
- P Chevallier
- Service d'Hématologie Clinique, CHU and Université de Nantes, INSERM U892, Nantes, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Pemmaraju N, Kantarjian H, Ravandi F, Cortes J. FLT3 inhibitors in the treatment of acute myeloid leukemia: the start of an era? Cancer 2011; 117:3293-304. [PMID: 21319142 DOI: 10.1002/cncr.25908] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/09/2010] [Accepted: 12/03/2010] [Indexed: 12/19/2022]
Abstract
Despite recent modest improvements in the chemotherapy regimens used to treat acute myeloid leukemia (AML), many patients diagnosed with AML ultimately die of the disease. Commonly occurring genetic alterations have been identified that strongly affect the prognosis for patients with AML. These alterations represent possible targets for investigational therapies that could act to specifically halt the aberrant growth of AML cells while limiting damage to normal cells. One such gene is the Fms-like tyrosine kinase 3 (FLT3) gene, which is mutated in approximately 30% of adult patients with AML and has a significant impact on prognosis. In particular, internal tandem duplications in FLT3 confer a poor prognosis to this large subgroup of patients with AML. Agents that target FLT3 are in development for the treatment of patients who have AML and offer a potential paradigm change in the current standard treatment of AML. For this report, the authors reviewed the prognostic significance of genetic alterations observed in AML with a focus on the therapeutic implications of targeting FLT3. The introduction of such agents may be the next major step toward the era of personalized therapy in AML.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
170
|
Borthakur G, Kantarjian H, Ravandi F, Zhang W, Konopleva M, Wright JJ, Faderl S, Verstovsek S, Mathews S, Andreeff M, Cortes JE. Phase I study of sorafenib in patients with refractory or relapsed acute leukemias. Haematologica 2011; 96:62-8. [PMID: 20952518 PMCID: PMC3012766 DOI: 10.3324/haematol.2010.030452] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED Background Sorafenib is a multi-kinase inhibitor with activity against fms-like tyrosine kinase 3 with internal tandem duplication mutation and Raf kinase among others. A phase I dose escalation study of sorafenib was conducted in patients with advanced myelodysplastic syndrome and relapsed or refractory acute leukemias. DESIGN AND METHODS Fifty patients received one of two different schedules; Schedule "A": once or twice daily, five days per week, every week for a 21 day cycle, and Schedule "B": once or twice daily, for 14 days every 21 days. Dose limiting toxicities were grade 3/4 hypertension, hyperbilirubinemia, and amylase elevation. The recommended phase II dose in hematologic malignancies is 400 mg twice daily for both schedules. RESULTS Complete remissions or complete remissions with incomplete recovery of platelets were achieved in 5 (10%) patients (all with fms-like tyrosine kinase 3-internal tandem duplication). Significant reduction in bone marrow and/or peripheral blood blasts was seen in an additional 17 (34%) patients (all with fms-like tyrosine kinase 3-internal tandem duplication). Eleven of these responses (including 3 complete remissions/complete remissions with incomplete recovery) lasted for 2 cycles or beyond. In conclusion, sorafenib is active and well tolerated in acute myelogenous leukemia with fms-like tyrosine kinase 3 internal tandem duplication mutation. Conclusions Additional studies of sorafenib in patients with acute myelogenous leukemia, particularly those with fms-like tyrosine kinase 3 internal tandem duplication, are warranted, including sorafenib-based combinations. (ClinicalTrials.gov Identifier: NCT00217646).
Collapse
Affiliation(s)
| | | | | | - Weiguo Zhang
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M. D. Anderson Cancer Center
| | - Marina Konopleva
- Leukemia Department and, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M. D. Anderson Cancer Center
| | - John J. Wright
- Cancer Therapy Evaluation Program, National Cancer Institute
| | | | | | | | - Michael Andreeff
- Leukemia Department and, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M. D. Anderson Cancer Center
| | - Jorge E. Cortes
- Leukemia Department and,Correspondence: Jorge E. Cortes, M.D., Leukemia Department 1515 Holcombe Blvd., Unit 428 Houston, TX 77030, USA. Phone: +1.713.7945783. Fax: +1.713.7944297. E-mail:
| |
Collapse
|
171
|
Roboz GJ. Novel approaches to the treatment of acute myeloid leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2011; 2011:43-50. [PMID: 22160011 DOI: 10.1182/asheducation-2011.1.43] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Approximately 12 000 adults are diagnosed with acute myeloid leukemia (AML) in the United States annually, the majority of whom die from their disease. The mainstay of initial treatment, cytosine arabinoside (ara-C) combined with an anthracycline, was developed nearly 40 years ago and remains the worldwide standard of care. Advances in genomics technologies have identified AML as a genetically heterogeneous disease, and many patients can now be categorized into clinicopathologic subgroups on the basis of their underlying molecular genetic defects. It is hoped that enhanced specificity of diagnostic classification will result in more effective application of targeted agents and the ability to create individualized treatment strategies. This review describes the current treatment standards for induction, consolidation, and stem cell transplantation; special considerations in the management of older AML patients; novel agents; emerging data on the detection and management of minimal residual disease (MRD); and strategies to improve the design and implementation of AML clinical trials.
Collapse
Affiliation(s)
- Gail J Roboz
- Weill Medical College of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
172
|
Lange T, Hubmann M, Burkhardt R, Franke GN, Cross M, Scholz M, Leiblein S, Al-Ali HK, Edelmann J, Thiery J, Niederwieser D. Monitoring of WT1 expression in PB and CD34(+) donor chimerism of BM predicts early relapse in AML and MDS patients after hematopoietic cell transplantation with reduced-intensity conditioning. Leukemia 2010; 25:498-505. [PMID: 21135860 DOI: 10.1038/leu.2010.283] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Relapse of malignant disease remains the major complication in patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) after hematopoietic cell transplantation (HCT) with reduced-intensity conditioning (RIC). In this study, we investigated the predictive value of disease-specific markers (DSMs), donor chimerism (DC) analysis of unsorted (UDC) or CD34(+) sorted cells and Wilms' tumor gene 1 (WT1) expression. Eighty-eight patients with AML or MDS were monitored after allogenic HCT following 2 Gy total-body irradiation with (n=84) or without (n=4) fludarabine 3 × 30 mg/m(2), followed by cyclosporin A and mycophenolate mofetil. DSMs were determined by fluorescence in situ hybridization (FISH) and WT1 expression by real-time polymerase chain reaction. Chimerism analysis was performed on unsorted or CD34(+) sorted cells, by FISH or short tandem repeat polymerase chain reaction. Twenty-one (24%) patients relapsed within 4 months after HCT. UDC, CD34(+) DC and WT1 expression were each significant predictors of relapse with sensitivities ranging from 53 to 79% and specificities of 82-91%. Relapse within 28 days was excluded almost entirely on the basis of WT1 expression combined with CD34(+) DC kinetics. Monitoring of WT1 expression and CD34(+) DC predict relapse of AML and MDS after RIC-HCT.
Collapse
Affiliation(s)
- T Lange
- Department of Hematology, Oncology and Hemostaseology, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Keir ST, Maris JM, Lock R, Kolb EA, Gorlick R, Carol H, Morton CL, Reynolds CP, Kang MH, Watkins A, Houghton PJ, Smith MA. Initial testing (stage 1) of the multi-targeted kinase inhibitor sorafenib by the pediatric preclinical testing program. Pediatr Blood Cancer 2010; 55:1126-33. [PMID: 20672370 PMCID: PMC3823056 DOI: 10.1002/pbc.22712] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Sorafenib is an inhibitor of multiple kinases (e.g., VEGF receptors, PDGFR, FLT3, RET, BRAF, KIT) and is approved by FDA for treatment of two adult cancers. The activity of sorafenib was evaluated against the PPTP's in vitro and in vivo panels. PROCEDURES Sorafenib was evaluated against the PPTP in vitro panel using 96-hr exposure at concentrations ranging from 1.0 nM to 10.0 µM. It was tested against the PPTP in vivo panels at a dose of 60 mg/kg administered by oral gavage daily for 5 days per week, repeated for 6 weeks. RESULTS In vitro sorafenib demonstrated cytotoxic activity, with a median IC(50) value of 4.3 µM. Twenty of 23 cell lines had IC(50) values between 1.0 and 10.0 µM. A single cell line (Kasumi-1) with an activating KIT mutation had an IC(50) value < 1.0 µM (IC(50) = 0.02 µM). In vivo sorafenib induced significant differences in event-free survival (EFS) distribution compared to control in 27 of 36 (75%) of the evaluable solid tumor xenografts and in 1 of 8 (12.5%) of the evaluable ALL xenografts. Sorafenib induced tumor growth inhibition meeting criteria for intermediate activity (EFS T/C) in 15 of 34 (44%) evaluable solid tumor xenografts. No xenografts achieved an objective response. CONCLUSIONS The primary in vitro activity of sorafenib was noted at concentrations above 1 µM, with the exception of a more sensitive cell line with an activating KIT mutation. The primary in vivo effect for sorafenib was tumor growth inhibition, which was observed across multiple histotypes.
Collapse
Affiliation(s)
| | - John M. Maris
- Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine and Abramson Family Cancer Research Institute, Philadelphia, PA
| | - Richard Lock
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | | | | | - Hernan Carol
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | | | | | - Min H. Kang
- Texas Tech University Health Sciences Center, Lubbock, TX
| | - Amy Watkins
- St. Jude Children's Research Hospital, Memphis, TN
| | | | | |
Collapse
|
174
|
Sorà F, Chiusolo P, Metafuni E, Bellesi S, Giammarco S, Laurenti L, Ausoni G, Zini G, Bayer AJ, Mario B, Leone G, Sica S. Sorafenib for refractory FMS-like tyrosine kinase receptor-3 (FLT3/ITD+) acute myeloid leukemia after allogenic stem cell transplantation. Leuk Res 2010; 35:422-3. [PMID: 21093053 DOI: 10.1016/j.leukres.2010.10.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 09/21/2010] [Accepted: 10/23/2010] [Indexed: 10/18/2022]
Affiliation(s)
- Federica Sorà
- Institute of Haematology, Università Cattolica Sacro Cuore, Roma, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Toxic effects of sorafenib when given early after allogeneic hematopoietic stem cell transplantation. Blood 2010; 116:2858-9. [PMID: 20947691 DOI: 10.1182/blood-2010-06-291104] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
176
|
Abstract
FLT3 inhibition has been a goal of acute myeloid leukemia (AML) therapy since FLT3 mutations were discovered to have a role in AML. Several FLT3 inhibitors have been developed in the last several years, beginning with less potent, less selective agents. The newer FLT3 inhibitors appear to be more potent in vivo and have shown more promise than the older agents in monotherapy trials.
Collapse
Affiliation(s)
- Mark J Levis
- Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA.
| |
Collapse
|
177
|
Porter DL, Alyea EP, Antin JH, DeLima M, Estey E, Falkenburg JHF, Hardy N, Kroeger N, Leis J, Levine J, Maloney DG, Peggs K, Rowe JM, Wayne AS, Giralt S, Bishop MR, van Besien K. NCI First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: Report from the Committee on Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2010; 16:1467-503. [PMID: 20699125 PMCID: PMC2955517 DOI: 10.1016/j.bbmt.2010.08.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 08/03/2010] [Indexed: 12/31/2022]
Abstract
Relapse is a major cause of treatment failure after allogeneic hematopoietic stem cell transplantation (alloHSCT). Treatment options for relapse have been inadequate, and the majority of patients ultimately die of their disease. There is no standard approach to treating relapse after alloHSCT. Withdrawal of immune suppression and donor lymphocyte infusions are commonly used for all diseases; although these interventions are remarkably effective for relapsed chronic myelogenous leukemia, they have limited efficacy in other hematologic malignancies. Conventional and novel chemotherapy, monoclonal antibody therapy, targeted therapies, and second transplants have been utilized in a variety of relapsed diseases, but reports on these therapies are generally anecdotal and retrospective. As such, there is an immediate need for well-designed, disease-specific trials for treatment of relapse after alloHSCT. This report summarizes current treatment options under investigation for relapse after alloHSCT in a disease-specific manner. In addition, recommendations are provided for specific areas of research necessary in the treatment of relapse after alloHSCT.
Collapse
MESH Headings
- Hematologic Neoplasms/therapy
- Hematopoietic Stem Cell Transplantation
- Hodgkin Disease/therapy
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myeloid, Acute/therapy
- Lymphocyte Transfusion
- Lymphoma, Non-Hodgkin
- Multiple Myeloma/therapy
- Neoplasm Recurrence, Local/therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Recurrence
- Transplantation, Homologous
- Treatment Failure
Collapse
Affiliation(s)
- David L Porter
- University of Pennsylvania Medical Center, Philadelphia, 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
VEGF targeted therapy in acute myeloid leukemia. Crit Rev Oncol Hematol 2010; 80:241-56. [PMID: 21035354 DOI: 10.1016/j.critrevonc.2010.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 09/24/2010] [Accepted: 09/28/2010] [Indexed: 01/07/2023] Open
Abstract
The cooperation of two classes of mutations in hematopoietic cells is hypothesized in a multistep pathogenesis model of acute myeloid leukemia (AML). Class I mutations confer a proliferative and/or survival advantage, whereas Class II mutations block hematopoietic differentiation and impair apoptosis in AML cells. In addition to these two classes of mutations, a relevant role for angiogenesis in the pathophysiology of AML has been recently proposed. The recognition that the vascular endothelial growth factor (VEGF) pathway is a key regulator of angiogenesis has led to the development of several VEGF-targeted approaches. These include neutralizing antibodies, VEGF traps or selective tyrosine kinase inhibitors for VEGFRs. Other drugs that indirectly affect VEGF pathway, such as statins or arsenic trioxide, also have been shown to possess antiangiogenic activity in leukemias. The benefits of these VEGF targeted agents and their current stage of development as novel anti-antiangiogenic therapies in AML are discussed in this review.
Collapse
|
179
|
The multikinase inhibitor Sorafenib displays significant antiproliferative effects and induces apoptosis via caspase 3, 7 and PARP in B- and T-lymphoblastic cells. BMC Cancer 2010; 10:560. [PMID: 20950443 PMCID: PMC2972283 DOI: 10.1186/1471-2407-10-560] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 10/15/2010] [Indexed: 12/16/2022] Open
Abstract
Background Targeted therapy approaches have been successfully introduced into the treatment of several cancers. The multikinase inhibitor Sorafenib has antitumor activity in solid tumors and its effects on acute lymphoblastic leukemia (ALL) cells are still unclear. Methods ALL cell lines (SEM, RS4;11 and Jurkat) were treated with Sorafenib alone or in combination with cytarabine, doxorubicin or RAD001. Cell count, apoptosis and necrosis rates, cell cycle distribution, protein phosphorylation and metabolic activity were determined. Results Sorafenib inhibited the proliferation of ALL cells by cell cycle arrest accompanied by down-regulation of CyclinD3 and CDK4. Furthermore, Sorafenib initiated apoptosis by cleavage of caspases 3, 7 and PARP. Apoptosis and necrosis rates increased significantly with most pronounced effects after 96 h. Antiproliferative effects of Sorafenib were associated with a decreased phosphorylation of Akt (Ser473 and Thr308), FoxO3A (Thr32) and 4EBP-1 (Ser65 and Thr70) as early as 0.5 h after treatment. Synergistic effects were seen when Sorafenib was combined with other cytotoxic drugs or a mTOR inhibitor emphasizing the Sorafenib effect. Conclusion Sorafenib displays significant antileukemic activity in vitro by inducing cell cycle arrest and apoptosis. Furthermore, it influences PI3K/Akt/mTOR signaling in ALL cells.
Collapse
|
180
|
Lal D, Park JA, Demock K, Marinaro J, Perez AM, Lin MH, Tian L, Mashtare TJ, Murphy M, Prey J, Wetzler M, Fetterly GJ, Wang ES. Aflibercept Exerts Antivascular Effects and Enhances Levels of Anthracycline Chemotherapy In vivo in Human Acute Myeloid Leukemia Models. Mol Cancer Ther 2010; 9:2737-51. [DOI: 10.1158/1535-7163.mct-10-0334] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
181
|
A case of blast clearance on sorafenib in relapsed FLT3-ITD acute myeloid leukemia: Evidence of efficacy continues to mount. Leuk Res 2010; 34:e268-9. [DOI: 10.1016/j.leukres.2010.03.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2010] [Revised: 03/28/2010] [Accepted: 03/29/2010] [Indexed: 11/21/2022]
|
182
|
Pratz KW, Levis MJ. Bench to bedside targeting of FLT3 in acute leukemia. Curr Drug Targets 2010; 11:781-9. [PMID: 20370649 DOI: 10.2174/138945010791320782] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 01/01/2010] [Indexed: 02/03/2023]
Abstract
FMS-Like-Tyrosine kinase-3 (FLT3) mutations are found in about 30% of cases of acute myeloid leukemia and confer an increased relapse rate and reduced overall survival. Targeting this tyrosine kinase by direction inhibition is the focus of both preclinical and clinical research in AML. Several molecules are in clinical development inhibit FLT3, but thus far clinical responses have been limited. Correlative studies from monotherapy trials have established that responses require sustained, effective FLT3 inhibition in vivo. Studies combining FLT3 inhibitors with chemotherapy have demonstrated increased remission rates to date but have yet to produce a survival advantage. Currently the only approved FLT3 inhibitor available for off-label use is sorafenib, which clearly has clinical activity but does not commonly lead to a complete response. Several FLT3 inhibitors are currently being tested as single agents and in combination with chemotherapy, and it seems likely that a clinically useful drug will eventually emerge.
Collapse
Affiliation(s)
- Keith W Pratz
- Department of Oncology, Division of Hematologic Malignancies, Sidney Kimmel Cancer Center at Johns Hopkins, 1650 Orleans Street, Baltimore, MD 21231, USA
| | | |
Collapse
|
183
|
Abstract
Mutations within the FMS-like tyrosine kinase 3 (FLT3) gene on chromosome 13q12 have been detected in up to 35% of acute myeloid leukemia (AML) patients and represent one of the most frequently identified genetic alterations in AML. Over the last years, FLT3 has emerged as a promising molecular target in therapy of AML. Here, we review results of clinical trials and of correlative laboratory studies using small molecule FLT3 tyrosine kinase inhibitors (TKIs) in AML patients. We also review mechanisms of primary and secondary drug resistance to FLT3-TKI, and from the data currently available we summarize lessons learned from FLT3-TKI monotherapy. Finally, for using FLT3 as a molecular target, we discuss novel strategies to overcome treatment failure and to improve FLT3 inhibitor therapy.
Collapse
|
184
|
Motyckova G, Stone RM. The role of molecular tests in acute myelogenous leukemia treatment decisions. Curr Hematol Malig Rep 2010; 5:109-17. [PMID: 20425404 DOI: 10.1007/s11899-010-0049-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The prognosis for patients with acute myelogenous leukemia (AML) is dependent on age, karyotype, and the genetics of the neoplastic cell. The molecular markers with prognostic impact include mutations in FLT3, NPM1, MLL, WT1, c-KIT, and expression levels of BAALC, NM1, ERG, and CXCR4. Gene expression profiles and microRNA expression patterns in AML may prove highly useful in defining the prognosis of AML. Cytogenetic and, increasingly, molecular findings are used in determining the best therapy for AML patients, especially the choice of whether to perform allogeneic stem cell transplantation.
Collapse
|
185
|
Secondary resistance to sorafenib in two patients with acute myeloid leukemia (AML) harboring FLT3-ITD mutations. Ann Hematol 2010; 90:473-5. [PMID: 20652568 DOI: 10.1007/s00277-010-1027-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 06/30/2010] [Indexed: 01/27/2023]
|
186
|
Weisberg E, Sattler M, Ray A, Griffin JD. Drug resistance in mutant FLT3-positive AML. Oncogene 2010; 29:5120-34. [DOI: 10.1038/onc.2010.273] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
187
|
Abstract
We report the results of a Phase I dose escalation trial of the multikinase inhibitor sorafenib in relapsed/refractory acute leukemias using an intermittent dosing regimen. Fifteen patients with advanced leukemia (Acute myeloid leukemia(AML), 2=Acute lymphoblastic leukemia(ALL), 1 Biphenotypic) and a median age of 63 (range 37–85) years were enrolled and treated on a dose escalation trial. Toxicities ≥grade 3 were present in 55% of cycles and the maximum tolerated dose (MTD) was determined to be 400mg BID × 21days in a 28 day cycle. Plasma inhibitory assays of kinase targets ERK and FLT3-ITD demonstrated excellent target inhibition, with FLT3-ITD silencing occurring below the MTD. The N-oxide metabolite of sorafenib appeared to be a more potent inhibitor of FLT3-ITD than the parent compound. Despite marked ex vivo FLT-3 ITD inhibition, no patients met criteria for complete or partial response in this monotherapy study. Eleven of fifteen patients experienced stable disease as best response. Although sorafenib demonstrated only modest clinical activity as a single agent in this heavily treated population, robust inhibition of FLT3 and ERK suggest there may be a potential important role in combination therapies.
Collapse
|
188
|
Abstract
Drugs introduced over the past 25 years have benefitted many patients with acute myeloid leukemia (AML) and provided cure for some. Still, AML remains difficult to treat, and most patients will eventually die from their disease. Therefore, novel drugs and drug combinations are under intense investigation, and promising results eagerly awaited and embraced. However, drug development is lengthy and costs are staggering. While the phase 1-phase 2-phase 3 sequence of clinical drug testing has remained inviolate for decades, it appears intrinsically inefficient, and scientific flaws have been noted by many authors. Of major concern is the high frequency of false-positive results obtained in phase 2 studies. Here, we review features of phase 2 trials in AML that may contribute to this problem, particularly lack of control groups, patient heterogeneity, selection bias, and choice of end points. Recognizing these problems and challenges should provide us with opportunities to make drug development more efficient and less costly. We also suggest strategies for trial design improvement. Although our focus is on the treatment of AML, the principles that we highlight should be broadly applicable to the evaluation of new treatments for a variety of diseases.
Collapse
|
189
|
von Bubnoff N, Rummelt C, Menzel H, Sigl M, Peschel C, Duyster J. Identification of a secondary FLT3/A848P mutation in a patient with FLT3-ITD-positive blast phase CMML and response to sunitinib and sorafenib. Leukemia 2010; 24:1523-5. [DOI: 10.1038/leu.2010.122] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
190
|
Winkler J, Rech D, Kallert S, Rech J, Meidenbauer N, Roesler W, Mackensen A. Sorafenib induces sustained molecular remission in FLT3-ITD positive AML with relapse after second allogeneic stem cell transplantation without exacerbation of acute GVHD: a case report. Leuk Res 2010; 34:e270-2. [PMID: 20627386 DOI: 10.1016/j.leukres.2010.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Revised: 03/31/2010] [Accepted: 04/13/2010] [Indexed: 12/01/2022]
|
191
|
Novel Approaches to Prevent Leukemia Relapse Following Allogeneic Hematopoietic Cell Transplantation. Curr Hematol Malig Rep 2010; 5:157-62. [DOI: 10.1007/s11899-010-0051-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
192
|
Affiliation(s)
- Tamar Tadmor
- Hematology-Oncology Unit, Bnai-Zion Medical Center, Haifa, Israel.
| | | | | |
Collapse
|
193
|
Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY, O'Brien S, Estrov Z, Borthakur G, Thomas D, Pierce SR, Brandt M, Byrd A, Bekele BN, Pratz K, Luthra R, Levis M, Andreeff M, Kantarjian HM. Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol 2010; 28:1856-62. [PMID: 20212254 PMCID: PMC2930809 DOI: 10.1200/jco.2009.25.4888] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To determine the efficacy and toxicity of the combination of sorafenib, cytarabine, and idarubicin in patients with acute myeloid leukemia (AML) younger than age 65 years. PATIENTS AND METHODS In the phase I part of the study, 10 patients with relapsed AML were treated with escalating doses of sorafenib with chemotherapy to establish the feasibility of the combination. We then treated 51 patients (median age, 53 years; range, 18 to 65 years) who had previously untreated AML with cytarabine at 1.5 g/m(2) by continuous intravenous (IV) infusion daily for 4 days (3 days if > 60 years of age), idarubicin at 12 mg/m(2) IV daily for 3 days, and sorafenib at 400 mg orally twice daily for 7 days. RESULTS Overall, 38 (75%) patients have achieved a complete remission (CR), including 14 (93%) of 15 patients with mutated FMS-like tyrosine kinase-3 (FLT3; the 15th patient had complete remission with incomplete platelet recovery [CRp]) and 24 (66%) of 36 patients with FLT3 wild-type (WT) disease (three additional FLT3-WT patients had CRp). FLT3-mutated patients were more likely to achieve a CR than FLT3-WT patients (P = .033). With a median follow-up of 54 weeks (range, 8 to 87 weeks), the probability of survival at 1 year is 74%. Among the FLT3-mutated patients, 10 have relapsed and five remain in CR with a median follow-up of 62 weeks (range, 10 to 76 weeks). Plasma inhibitory assay demonstrated an on-target effect on FLT3 kinase activity. CONCLUSION Sorafenib can be safely combined with chemotherapy, produces a high CR rate in FLT3-mutated patients, and inhibits FLT3 signaling.
Collapse
Affiliation(s)
- Farhad Ravandi
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD.,Corresponding author: Farhad Ravandi, MD, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX 77030; e-mail:
| | - Jorge E. Cortes
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Daniel Jones
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Stefan Faderl
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Guillermo Garcia-Manero
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Marina Y. Konopleva
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Susan O'Brien
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Zeev Estrov
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Gautam Borthakur
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Deborah Thomas
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Sherry R. Pierce
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Mark Brandt
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Anna Byrd
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - B. Nebiyou Bekele
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Keith Pratz
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Rajyalakshmi Luthra
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Mark Levis
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Michael Andreeff
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Hagop M. Kantarjian
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| |
Collapse
|
194
|
Jin Y, Lu Z, Ding K, Li J, Du X, Chen C, Sun X, Wu Y, Zhou J, Pan J. Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species. Cancer Res 2010; 70:2516-27. [PMID: 20215516 DOI: 10.1158/0008-5472.can-09-3950] [Citation(s) in RCA: 252] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
NF-kappaB may be a potential therapeutic target for acute myelogenous leukemia (AML) because NF-kappaB activation is found in primitive human AML blast cells. In this report, we initially discovered that the potent antineoplastic effect of niclosamide, a Food and Drug Administration-approved antihelminthic agent, was through inhibition of the NF-kappaB pathway in AML cells. Niclosamide inhibited the transcription and DNA binding of NF-kappaB. It blocked tumor necrosis factor-induced IkappaBalpha phosphorylation, translocation of p65, and expression of NF-kappaB-regulated genes. Niclosamide inhibited the steps TAK1-->IkappaB kinase (IKK) and IKK-->IkappaBalpha. Niclosamide also increased the levels of reactive oxygen species (ROS) in AML cells. Quenching ROS by the glutathione precursor N-acetylcysteine attenuated niclosamide-induced apoptosis. Our results together suggest that niclosamide inhibited the NF-kappaB pathway and increased ROS levels to induce apoptosis in AML cells. On translational study of the efficacy of niclosamide against AML, niclosamide killed progenitor/stem cells from AML patients but spared those from normal bone marrow. Niclosamide was synergistic with the frontline chemotherapeutic agents cytarabine, etoposide, and daunorubicin. It potently inhibited the growth of AML cells in vitro and in nude mice. Our results support further investigation of niclosamide in clinical trials of AML patients.
Collapse
Affiliation(s)
- Yanli Jin
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Fathi AT, Grant S, Karp JE. Exploiting cellular pathways to develop new treatment strategies for AML. Cancer Treat Rev 2010; 36:142-50. [PMID: 20056334 DOI: 10.1016/j.ctrv.2009.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 12/01/2009] [Accepted: 12/05/2009] [Indexed: 12/14/2022]
Abstract
The standard approaches to the treatment of acute myeloid leukemia (AML) have been predominantly based on cytarabine and anthracyclines. Yet, the outcomes associated with AML continue to be poor, especially for those patients who are older or carry higher-risk disease. In recent years, extensive research has led to the development and study of novel agents which target AML by diverse and varied mechanisms. Among these are targeted therapeutics such as kinase inhibitors and oligonucleotide constructs. These aim to suppress the production or activity of proteins, such as FLT3 and BCL2, among others, and thus disrupt related signaling cascades essential for leukemogenesis and proliferation. In addition, other agents like flavopiridol appear to target the myeloid blast by various mechanisms including suppression of cyclin-dependent kinases and interference with nucleotide synthesis. Another class of novel therapies includes inhibitors of histone deacetylase, which cause growth arrest and apoptosis through histone acetylation and resultant conformational changes. Clinical trials are now studying these and other agents alone and in combination with traditional cytotoxic therapies, with some encouraging results. In this review, we aim to provide a summary of the preclinical and clinical investigations of selected promising agents currently under study.
Collapse
Affiliation(s)
- Amir T Fathi
- Division of Hematologic Malignancies, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, 1650 Orleans Street, Baltimore, MD 21231, USA.
| | | | | |
Collapse
|
196
|
|
197
|
Ravandi F, Kantarjian H, Faderl S, Garcia-Manero G, O'Brien S, Koller C, Pierce S, Brandt M, Kennedy D, Cortes J, Beran M. Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. Leuk Res 2009; 34:752-6. [PMID: 19878996 DOI: 10.1016/j.leukres.2009.10.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 10/01/2009] [Accepted: 10/01/2009] [Indexed: 11/19/2022]
Abstract
Mutations of Fms-like tyrosine kinase-3 (FLT3) have been described in about 30% of patients with acute myeloid leukemia (AML) and are associated with a shorter disease-free and overall survival after initial therapy. We sought to examine whether the presence of these mutations in relapsed disease was also associated with a poor response to salvage chemotherapy by comparing the outcome of 34 patients with diploid cytogenetics and mutated FLT3 (internal tandem duplication mutation, ITD) to 69 patients with normal karyotype and wild-type FLT3 (FLT3-WT) in first relapse. On univariate analysis, patients with mutated FLT3 were less likely to achieve a CR to first salvage compared to FLT3-WT patients (24% vs. 41%; P=0.09). Furthermore, survival was longer for the FLT3-WT patients achieving a second CR after salvage compared to FLT3-mutated patients (P=0.017). Overall, patients with mutated FLT3 had a shorter survival from the time of relapse compared to those with FLT3-WT (P<0.001). The adverse prognostic impact of FLT3 mutations appears to persist beyond the initial treatment.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cohort Studies
- Follow-Up Studies
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Middle Aged
- Mutation/physiology
- Prognosis
- Recurrence
- Salvage Therapy
- Survival Analysis
- Treatment Outcome
- Young Adult
- fms-Like Tyrosine Kinase 3/genetics
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
van den Brink MRM, Porter DL, Giralt S, Lu SX, Jenq RR, Hanash A, Bishop MR. Relapse after allogeneic hematopoietic cell therapy. Biol Blood Marrow Transplant 2009; 16:S138-45. [PMID: 19857588 DOI: 10.1016/j.bbmt.2009.10.023] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Disease relapse remains a major cause of mortality following allogeneic hematopoietic cell transplantation (HCT). Over the past decade, our understanding of the biology underlying the graft-versus-tumor/leukemia (GVT) effect has increased greatly; however, several other factors affect the occurrence and outcome of relapse, including conditioning regimen, type of allograft, and the histology, status, and sensitivity to chemotherapy of the disease being treated. The mainstay of relapse treatment is donor lymphocyte infusion (DLI), but the efficacy of DLI is quite variable depending on disease histology and state. As such, there is a significant need for novel therapies and strategies for relapse following allogeneic HCT, particularly in patients for whom DLI is not an option. The National Cancer Institute is sponsoring an international workshop to address issues and research questions relative to the biology, natural history, prevention, and treatment of relapse following allogeneic HCT.
Collapse
|
199
|
Influence of NPM1 and FLT3-ITD status on outcome in relapsed/refractory AML patients receiving salvage therapy including gemtuzumab ozogamicin. Leukemia 2009; 24:467-9. [PMID: 19847200 DOI: 10.1038/leu.2009.214] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
200
|
Lee SH, Paietta E, Racevskis J, Wiernik PH. Complete resolution of leukemia cutis with sorafenib in an acute myeloid leukemia patient with FLT3-ITD mutation. Am J Hematol 2009; 84:701-2. [PMID: 19714594 DOI: 10.1002/ajh.21511] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|