1
|
Luger SM, Wang VX, Rowe JM, Litzow MR, Paietta E, Ketterling RP, Lazarus H, Rybka WB, Craig MD, Karp J, Cooper BW, Makary AZ, Kaminer LS, Appelbaum FR, Larson RA, Tallman MS. Tipifarnib as maintenance therapy did not improve disease-free survival in patients with acute myelogenous leukemia at high risk of relapse: Results of the phase III randomized E2902 trial. Leuk Res 2021; 111:106736. [PMID: 34773794 DOI: 10.1016/j.leukres.2021.106736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Despite the achievement of complete remission with chemotherapy in patients with acute myeloid leukemia (AML), relapse is common and the majority of patients will die of their disease. Patients who achieve a remission after refractory or relapsed disease as well as elderly patients have a very high rate of relapse even if they achieve a complete remission. A phase 3 randomized ECOG-ACRIN-led intergroup study was conducted to determine whether post-remission therapy with the farnesyl transferase inhibitor, tipifarnib (R115777), improved the disease-free survival (DFS) of adult patients with AML in complete remission (CR), at high risk for relapse. PATIENTS AND METHODS Adult patients with AML in remission after salvage therapy and/or over age 60 in first remission were enrolled in this study. They were randomly assigned to treatment with tipifarnib or observation (control). The primary objective was to compare the disease-free survival (DFS) between the two arms based on intention to treat, which includes all randomized patients. RESULTS One hundred and forty-four patients were enrolled on the study. Median DFS was 8.9 vs 5.3 months, for tipifarnib vs observation (one-sided p = 0.026) and did not cross the pre-specified boundary to call the study positive. For the 134 eligible patients, median DFS was 10.8 vs 5.3 months for those randomized to tipifarnib vs observation (one-sided p = 0.008). Moreover in an ad hoc evaluation of all women (n = 71) median DFS was 12.1 vs 3.9 months for tipifarnib vs observation (one-sided p = 0.0004) while median OS was 26.5 vs 8.4 months respectively (one-sided p = 0.001). CONCLUSION This study was not able to demonstrate a benefit to tipifarnib as maintenance therapy in patients with AML in remission. While subsets of patients may indeed benefit, additional studies would be needed to elucidate that benefit which is unlikely given that other seemingly better options have since become available.
Collapse
Affiliation(s)
- Selina M Luger
- Abramson Cancer Center, University of Pennsylvania, Perelman Center for Advanced Medicine, South Tower, 12th Floor, Philadelphia, PA, PA 19104, United States.
| | - Victoria X Wang
- Dana Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, AMA, United States
| | | | | | | | | | - Hillard Lazarus
- Case Western Reserve University, Cleveland, OH, United States
| | - Witold B Rybka
- Penn State Hershey Cancer Institute, Hershey, PA, United States
| | - Michael D Craig
- West Virginia University Healthcare, Morgantown, WB, United States
| | - Judith Karp
- Johns Hopkins University, Baltimore, MD, United States
| | - Brenda W Cooper
- Case Western Reserve University, Cleveland, OH, United States
| | - Adel Z Makary
- Geisinger Medical Center, Danville, PA, United States
| | - Lynne S Kaminer
- North Shore Health System-Evanston Hospital, Evanston, IL, United States
| | | | | | - Martin S Tallman
- Northwestern University, Chicago, IL, United States(1); Memorial Sloan Kettering Cancer Center, NY, NY, United States(2)
| |
Collapse
|
2
|
Evolving Therapeutic Approaches for Older Patients with Acute Myeloid Leukemia in 2021. Cancers (Basel) 2021; 13:cancers13205075. [PMID: 34680226 PMCID: PMC8534216 DOI: 10.3390/cancers13205075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The better understanding of disease biology, the availability of new effective drugs and the increased awareness of patients’ heterogeneity in terms of fitness and personal expectations has made the current treatment paradigm of AML in the elderly very challenging. Here, we discuss the evolving criteria used to define eligibility for induction chemotherapy and transplantation, the introduction of new agents in the treatment of patients with very different clinical conditions, the implications of precision medicine and the importance of quality of life and supportive care, proposing a simplified algorithm that we follow in 2021. Abstract Acute myeloid leukemia (AML) in older patients is characterized by unfavorable prognosis due to adverse disease features and a high rate of treatment-related complications. Classical therapeutic options range from intensive chemotherapy in fit patients, potentially followed by allogeneic hematopoietic cell transplantation (allo-HCT), to hypomethylating agents or palliative care alone for unfit/frail ones. In the era of precision medicine, the treatment paradigm of AML is rapidly changing. On the one hand, a plethora of new targeted drugs with good tolerability profiles are becoming available, offering the possibility to achieve a prolonged remission to many patients not otherwise eligible for more intensive therapies. On the other hand, better tools to assess patients’ fitness and improvements in the selection and management of those undergoing allo-HCT will hopefully reduce treatment-related mortality and complications. Importantly, a detailed genetic characterization of AML has become of paramount importance to choose the best therapeutic option in both intensively treated and unfit patients. Finally, improving supportive care and quality of life is of major importance in this age group, especially for the minority of patients that are still candidates for palliative care because of very poor clinical conditions or unwillingness to receive active treatments. In the present review, we discuss the evolving approaches in the treatment of older AML patients, which is becoming increasingly challenging following the advent of new effective drugs for a very heterogeneous and complex population.
Collapse
|
3
|
Retinoids in hematology: a timely revival? Blood 2021; 137:2429-2437. [PMID: 33651885 DOI: 10.1182/blood.2020010100] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/17/2021] [Indexed: 12/27/2022] Open
Abstract
The retinoic acid receptors (RARA, RARB, and RARG) are ligand-regulated nuclear receptors that act as transcriptional switches. These master genes drew significant interest in the 1990s because of their key roles in embryogenesis and involvement in a rare malignancy, acute promyelocytic leukemia (APL), in which the RARA (and very rarely, RARG or RARB) genes are rearranged, underscoring the central role of deregulated retinoid signaling in leukemogenesis. Several recent provocative observations have revived interest in the roles of retinoids in non-APL acute myeloid leukemia (AML), as well as in normal hematopoietic differentiation. We review the role of retinoids in hematopoiesis, as well as in the treatment of non-APL AMLs. From this perspective, broader uses of retinoids in the management of hematopoietic tumors are discussed.
Collapse
|
4
|
Abou Dalle I, Kantarjian HM, Ravandi F, Daver N, Wang X, Jabbour E, Estrov Z, DiNardo CD, Pemmaraju N, Ferrajoli A, Jain N, Wang SA, Jammal N, Borthakur G, Naqvi K, Pelletier S, Pierce S, Andreeff M, Garcia-Manero G, Cortes JE, Kadia TM. Phase 2 study of lenalidomide maintenance for patients with high-risk acute myeloid leukemia in remission. Cancer 2021; 127:1894-1900. [PMID: 33449377 DOI: 10.1002/cncr.33409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/12/2020] [Accepted: 11/27/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUND New drug combinations have led to significant improvements in remission rates for patients with acute myeloid leukemia (AML). However, many patients with high-risk AML who respond to their initial treatment and are not candidates for allogeneic stem cell transplantation (ASCT) will eventually relapse with poor outcomes. METHODS In this phase 2 trial, the efficacy of lenalidomide maintenance was evaluated in patients with high-risk AML who had achieved their first or second remission after induction chemotherapy and at least 1 consolidation cycle and who were not candidates for immediate ASCT. Lenalidomide was given orally at 10 to 20 mg daily on days 1 to 28 of a 28-day cycle for up to 24 cycles. RESULTS A total of 28 patients were enrolled in this study with a median age of 61 years (range, 24-87 years). The median number of cycles was 8 (range, 1-24 cycles). Ten patients (36%) completed 24 months of maintenance treatment. With a median follow-up of 22.5 months (range, 2.6-55 months), 12 patients (43%) relapsed after a median of 3 months (range, 0.7-23 months). The median duration of remission for all patients was 18.7 months (range, 0.7-55.1 months). The 2-year overall survival and relapse-free survival rates from the time of enrollment were 63% and 50%, respectively. Overall, lenalidomide was well tolerated; serious adverse events of grade 3 or 4, including rash (n = 5), thrombocytopenia (n = 4), neutropenia (n = 4), and fatigue (n = 2), were observed in 13 patients (46%). CONCLUSIONS Lenalidomide is a safe and feasible maintenance strategy in patients with high-risk AML who are not candidates for ASCT, and it has beneficial effects for patients with negative measurable residual disease.
Collapse
Affiliation(s)
- Iman Abou Dalle
- Hematology-Oncology Division, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zeev Estrov
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nadya Jammal
- Department of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kiran Naqvi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarah Pelletier
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
5
|
Levin M, Stark M, Ofran Y, Assaraf YG. Deciphering molecular mechanisms underlying chemoresistance in relapsed AML patients: towards precision medicine overcoming drug resistance. Cancer Cell Int 2021; 21:53. [PMID: 33446189 PMCID: PMC7809753 DOI: 10.1186/s12935-021-01746-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/02/2021] [Indexed: 12/13/2022] Open
Abstract
Background Acute myeloid leukemia (AML) remains a devastating disease with a 5-year survival rate of less than 30%. AML treatment has undergone significant changes in recent years, incorporating novel targeted therapies along with improvements in allogeneic bone marrow transplantation techniques. However, the standard of care remains cytarabine and anthracyclines, and the primary hindrance towards curative treatment is the frequent emergence of intrinsic and acquired anticancer drug resistance. In this respect, patients presenting with chemoresistant AML face dismal prognosis even with most advanced therapies. Herein, we aimed to explore the potential implementation of the characterization of chemoresistance mechanisms in individual AML patients towards efficacious personalized medicine. Methods Towards the identification of tailored treatments for individual patients, we herein present the cases of relapsed AML patients, and compare them to patients displaying durable remissions following the same chemotherapeutic induction treatment. We quantified the expression levels of specific genes mediating drug transport and metabolism, nucleotide biosynthesis, and apoptosis, in order to decipher the molecular mechanisms underlying intrinsic and/or acquired chemoresistance modalities in relapsed patients. This was achieved by real-time PCR using patient cDNA, and could be readily implemented in the clinical setting. Results This analysis revealed pre-existing differences in gene expression levels between the relapsed patients and patients with lasting remissions, as well as drug-induced alterations at different relapse stages compared to diagnosis. Each of the relapsed patients displayed unique chemoresistance mechanisms following similar treatment protocols, which could have been missed in a large study aimed at identifying common drug resistance determinants. Conclusions Our findings emphasize the need for standardized evaluation of key drug transport and metabolism genes as an integral component of routine AML management, thereby allowing for the selection of treatments of choice for individual patients. This approach could facilitate the design of efficacious personalized treatment regimens, thereby reducing relapse rates of therapy refractory disease.
Collapse
Affiliation(s)
- May Levin
- The Fred Wyszkowski Cancer Research Laboratory, Dept. of Biology, Technion-Israel Institute of Technology, 3200003, Haifa, Israel
| | - Michal Stark
- The Fred Wyszkowski Cancer Research Laboratory, Dept. of Biology, Technion-Israel Institute of Technology, 3200003, Haifa, Israel
| | - Yishai Ofran
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Dept. of Biology, Technion-Israel Institute of Technology, 3200003, Haifa, Israel.
| |
Collapse
|
6
|
Xuan L, Liu Q. Maintenance therapy in acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. J Hematol Oncol 2021; 14:4. [PMID: 33407700 PMCID: PMC7786934 DOI: 10.1186/s13045-020-01017-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Relapse remains the main cause of treatment failure in acute myeloid leukemia (AML) undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). Emerging evidence has demonstrated that AML patients might benefit from maintenance therapy post-transplantation, especially for high-risk AML patients. In this mini-review, we will summarize targeted drugs, such as hypomethylating agents, FLT3 inhibitors and isocitrate dehydrogenase inhibitors, as maintenance therapy post-transplantation in AML patients undergoing allo-HSCT.
Collapse
Affiliation(s)
- Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Zhao X, Liu HQ, Wang LN, Yang L, Liu XL. Current and emerging molecular and epigenetic disease entities in acute myeloid leukemia and a critical assessment of their therapeutic modalities. Semin Cancer Biol 2020; 83:121-135. [PMID: 33242577 DOI: 10.1016/j.semcancer.2020.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
Acute myeloid leukemia (AML) is the most frequently diagnosed acute leukemia, and its incidence increases with age. Although the etiology of AML remains unknown, exposure to genotoxic agents or some prior hematologic disorders could lead to the development of this condition. The pathogenesis of AML involves the development of malignant transformation of hematopoietic stem cells that undergo successive genomic alterations, ultimately giving rise to a full-blown disease. From the disease biology perspective, AML is considered to be extremely complex with significant genetic, epigenetic, and phenotypic variations. Molecular and cytogenetic alterations in AML include mutations in those subsets of genes that are involved in normal cell proliferation, maturation and survival, thus posing significant challenge to targeting these pathways without attendant toxicity. In addition, multiple malignant cells co-exist in the majority of AML patients. Individual subclones are characterized by unique genetic and epigenetic abnormalities, which contribute to the differences in their response to treatment. As a result, despite a dramatic progress in our understanding of the pathobiology of AML, not much has changed in therapeutic approaches to treat AML in the past four decades. Dose and regimen modifications with improved supportive care have contributed to improved outcomes by reducing toxicity-related side effects. Several drug candidates are currently being developed, including targeted small-molecule inhibitors, cytotoxic chemotherapies, monoclonal antibodies and epigenetic drugs. This review summarizes the current state of affairs in the pathobiological and therapeutic aspects of AML.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Paediatrics, The First Hospital of Jilin University, Changchun, China
| | - Huan-Qiu Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Li-Na Wang
- Department of Paediatrics, The First Hospital of Jilin University, Changchun, China
| | - Le Yang
- Department of Endocrinology, The People's Hospital of Jilin Province, Changchun, China.
| | - Xiao-Liang Liu
- Department of Hematology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Demichelis-Gómez R, Zapata-Canto N, Leyto-Cruz F, Terreros-Muñoz E, Carrillo A, Montaño-Figueroa E, Solís-Poblano JC, Colunga-Pedraza P, Díaz-Vargas G, Amador-Medina LF, Martínez-Hernández R, Turrubiates F, Cabrera-García A, Zaragoza A, Espinoza R, Gutiérrez-Serdán R, Apodaca E, Moreira C, García-Castillo C, García-Stivalet L, Limón A, Gómez-Almaguer D, Rozen-Fuller E, Espinosa-Bautista KA, Crespo-Solís E, Meillón L. Acute Myeloid Leukemia in Mexico: The Specific Challenges of a Developing Country. Results From a Multicenter National Registry. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 20:e295-e303. [PMID: 32192977 DOI: 10.1016/j.clml.2019.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/13/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND In the past decades, long-term survival outcomes for younger patients with acute myeloid leukemia (AML) have improved. Nonetheless, developing nations might be lagging behind, highlighting the need to assess real-world outcomes in such regions. METHODS We performed a multicenter retrospective study, which included patients with AML diagnosed between January 2013 and December 2017 from 13 centers in Mexico. RESULTS A total of 525 patients with AML met the inclusion criteria and were included in the study. Median age for the entire cohort was 47 years. The patients were classified according to cytogenetic risk: favorable 16.0%, intermediate 55.6%, and unfavorable 28.4%. Most patients received intensive chemotherapy (80.2%), and among these 74.1% underwent a 7 + 3 induction regimen. A complete remission was achieved in 71.3% of patients. Induction-related mortality occurred in 17.8% and we identify the following as independent risk factors: >60 years (odds ratio [OR] 2.09 [1.09-4.02]), Eastern Cooperative Oncology Group >2 (OR 4.82 [2.46-9.43]), prior solid tumor (OR 3.8 [1.24-11.59]) and active infection (OR 1.82 [1.06-3.12]). Further, allogeneic hematopoietic stem-cell transplantation (AlloHSCT) was performed in 8.2% in CR1. The 3-year overall survival (OS) was 34.8%. In a multivariate analysis, several factors were independently associated with a worse OS, including secondary AML (hazard ratio [HR] 2.14 [1.15-4.01]) and unfavorable cytogenetic risk (HR 1.81 [1.16-2.82]), whereas maintenance therapy (HR 0.53 [0.32-0.86]) and AlloHSCT (HR 0.40 [0.17-0.94]) were associated with better OS. CONCLUSIONS This is the first multicenter report analyzing AML survival in Mexico. Challenges in this setting include a high induction-related mortality and low AlloHSCT rate, which should be addressed to improve outcomes.
Collapse
Affiliation(s)
- Roberta Demichelis-Gómez
- Hematology and Oncology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Nidia Zapata-Canto
- Hematology Department, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | | | | | | | | | - Perla Colunga-Pedraza
- Hematology Department, Hospital Universitario "Dr. JoséEleuterio González", Monterrey, Mexico
| | | | | | - Ramón Martínez-Hernández
- Hematology Department, Hospital Regional de Alta Especialidad de Ciudad Victoria, Ciudad Victoria, Mexico
| | - Francisco Turrubiates
- Hematology Department, Hospital Regional de Alta Especialidad de Ciudad Victoria, Ciudad Victoria, Mexico
| | - Alvaro Cabrera-García
- Hematology Department, Hospital Regional de Alta Especialidad de Ixtapaluca, Estado de México, Mexico
| | - Alva Zaragoza
- Hospital General Regional no. 1, IMSS, Chihuahua, Mexico
| | - Ramiro Espinoza
- Hematology Department, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Elia Apodaca
- Hematology and Oncology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carolina Moreira
- Hematology Department, Hospital Juárez de México, Mexico City, Mexico
| | | | | | - Alejandro Limón
- Hospital de Especialidades CMN "Manuel Ávila Camacho", IMSS, Puebla, Mexico
| | - David Gómez-Almaguer
- Hematology Department, Hospital Universitario "Dr. JoséEleuterio González", Monterrey, Mexico
| | - Etta Rozen-Fuller
- Hematology Department, Hospital General de México, Mexico City, Mexico
| | | | - Erick Crespo-Solís
- Hematology Department, Hospital Regional de Alta Especialidad de Ciudad Victoria, Ciudad Victoria, Mexico
| | - Luis Meillón
- Hematology Department, Centro Médico ABC, Mexico City, Mexico
| |
Collapse
|
9
|
Molica M, Breccia M, Foa R, Jabbour E, Kadia TM. Maintenance therapy in AML: The past, the present and the future. Am J Hematol 2019; 94:1254-1265. [PMID: 31429099 DOI: 10.1002/ajh.25620] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/29/2022]
Abstract
Curative treatment in acute myeloid leukemia (AML) depends on successful induction therapy to achieve a complete remission (CR), and subsequent post-remission therapy to prevent relapse. High relapse rates after consolidation therapy and after allogeneic stem cell transplant contribute to suboptimal outcomes in AML patients, and continue to represent a difficult challenge. Effective maintenance therapy could play an important role in prolonging the remission interval in the post-consolidation setting, especially in high risk AML patients. Maintenance treatment approaches based on conventional chemotherapy, immunotherapy, hypomethylating agents, and targeted small molecules have been explored in this setting, but no data so far have been convincing enough to establish this approach as the standard of care. However, ongoing and future studies including novel targeted therapies may demonstrate promising efficacy that could facilitate incorporation of maintenance therapy into clinical practice. In this review we summarize previous and ongoing approaches of maintenance therapy in AML and discuss the most promising strategies.
Collapse
Affiliation(s)
- Matteo Molica
- Hematology, Department of Translational and Precision MedicineUniversity Sapienza Rome Roma Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision MedicineUniversity Sapienza Rome Roma Italy
| | - Roberto Foa
- Hematology, Department of Translational and Precision MedicineUniversity Sapienza Rome Roma Italy
| | - Elias Jabbour
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Tapan M. Kadia
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| |
Collapse
|
10
|
Cerrano M, Candoni A, Crisà E, Dubbini MV, D'Ardia S, Zannier ME, Boccadoro M, Audisio E, Bruno B, Ferrero D. FLAI induction regimen in elderly patients with acute myeloid leukemia. Leuk Lymphoma 2019; 60:3339-3340. [PMID: 31159609 DOI: 10.1080/10428194.2019.1620943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Marco Cerrano
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,Department of Oncology, Division of Hematology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Anna Candoni
- Division of Hematology and SCT, University of Udine, Udine, Italy
| | - Elena Crisà
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,Department of Oncology, Division of Hematology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Stefano D'Ardia
- Department of Oncology, Division of Hematology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Mario Boccadoro
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,Department of Oncology, Division of Hematology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Ernesta Audisio
- Department of Oncology, Division of Hematology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,Department of Oncology, SSD Trapianto Allogenico, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Dario Ferrero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,Department of Oncology, Division of Hematology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| |
Collapse
|
11
|
Liu P, Li P, Lei T, Qu L, Huang H, Mu Q. Acute lymphoblastic leukemia following temozolomide treatment in a patient with glioblastoma: A case report and review of the literature. Oncol Lett 2018; 15:8663-8668. [PMID: 29805603 DOI: 10.3892/ol.2018.8422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/22/2016] [Indexed: 01/24/2023] Open
Abstract
Temozolomide (TMZ) is a second-generation oral alkylating agent that functions against a number of central nervous system neoplasms, and is generally used to treat high-grade gliomas, including anaplastic astrocytoma and glioblastoma multiforme. Therapy-related secondary myelodysplastic syndrome and acute myeloid leukemia have been reported in patients following prolonged exposure to TMZ. However, TMZ-related acute lymphoblastic leukemia (ALL) is extremely rare. The present study describes the case of an 11-year-old boy with a 3-day history of generalized tonic-clonic seizures and a contrast-enhanced lesion in the left temporooccipital region with focal cystic degeneration, as detected by magnetic resonance imaging. The patient underwent craniotomy and gross-total resection andpathological analysis confirmed the diagnosis of giant cell glioblastoma. Postoperatively, the patient received TMZ-based concurrent chemoradiation during radiotherapy, and developed B-cell ALL 6 months following TMZ treatment. A thorough literature search identified only six published cases of TMZ-related ALL. The chemotherapeutic efficacy of TMZ has been identified, however, its leukemogenic potential should be emphasized among practitioners and patients. Further studies are required to determine the specific pathogenic mechanism of TMZ-related ALL. Close hematological monitoring of patients following TMZ treatment is vital and a high index of suspicion is necessary.
Collapse
Affiliation(s)
- Pengfei Liu
- Department of Neurosurgery The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China.,Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China.,Department of Neurosurgery, Guangdong 999 Brain Hospital, Guangzhou, Guangdong 510510, P.R. China
| | - Peiwen Li
- Department of Neurosurgery The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Ting Lei
- Department of Neurosurgery The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Limei Qu
- Department of Pathology, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Haiyan Huang
- Department of Neurosurgery The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Qingchun Mu
- Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
12
|
Dong W, Lin Y, Cao Y, Liu Y, Xie X, Gu W. Luteolin induces myelodysplastic syndrome‑derived cell apoptosis via the p53‑dependent mitochondrial signaling pathway mediated by reactive oxygen species. Int J Mol Med 2018; 42:1106-1115. [PMID: 29786746 DOI: 10.3892/ijmm.2018.3696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/08/2018] [Indexed: 11/06/2022] Open
Abstract
Luteolin, a common dietary flavonoid, induces the apoptosis of cells in several types of cancer. However, its role in myelodysplastic syndrome (MDS) and the potential underlying mechanisms remain to be elucidated. To evaluate the potential benefit and underlying mechanisms of luteolin in MDS cells, the viability of SKM‑1 cells and primary bone marrow (PBM) mononuclear cells from patients with intermediate‑ or high‑risk MDS were assessed using a Cell Counting Kit‑8 assay. The apoptotic features of cell morphology were assessed using Wright‑Giemsa staining, DNA fragmentation was analyzed by agarose gel electrophoresis, and the extent of apoptosis was quantified by flow cytometry (FCM). Reactive oxygen species (ROS) were measured by FCM with 2,7‑dichlorodihydrofluorescein diacetate staining and mitochondrial membrane potential (ΔΨm) was determined using 5,5',6,6'‑tetrachloro‑1,1',3,3'‑tetraethylbenzimidazolylcarbocyanine iodide staining. Caspase activity was detected using a fluorometric protease assay. Furthermore, the effects of luteolin on the expression of apoptosis‑related proteins were analyzed using western blot analysis. The resulting data revealed that luteolin significantly inhibited the proliferation of SKM‑1 cells in vitro, and its half maximal inhibitory concentration was 139.41 µM at 24 h and 23.95 µM at 72 h. Luteolin also markedly inhibited the proliferation of mononuclear cells from patients with intermediate‑ or high‑risk MDS. Luteolin suppressed cell proliferation, mainly as a result of the induction of apoptosis, as demonstrated by typical apoptotic morphological features, the ladder pattern of genomic DNA fragmentation, and the results of FCM using Annexin V‑FITC/PI double staining. It was also found that short‑term exposure of SKM‑1 cells to luteolin led to a marked increase in the accumulation of ROS. The increased intracellular level of ROS appeared to induce the activation of p53 and elevate the B‑cell lymphoma 2 (Bcl‑2)‑associated X protein/Bcl‑2 ratio, which modulates ΔΨm and triggers the release of cytochrome c, and may increase the activities of apoptotic protease activating factor 1, caspase‑3, ‑8 and ‑9 to further trigger the destruction of structural and specific proteins and thereby cell apoptosis. Notably, the inhibition of ROS generation by the antioxidant N‑acetyl‑L‑cysteine significantly attenuated the luteolin‑induced loss of ΔΨm and activities of caspase‑3, ‑8 and ‑9. These data suggested that luteolin exerts its pro‑apoptotic action partly through the p53‑dependent mitochondrial signaling pathway mediated by intracellular ROS, which provides a promising therapeutic candidate for patients with MDS.
Collapse
Affiliation(s)
- Weimin Dong
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213003, P.R. China
| | - Yan Lin
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213003, P.R. China
| | - Yang Cao
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213003, P.R. China
| | - Yue Liu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213003, P.R. China
| | - Xiaobao Xie
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213003, P.R. China
| | - Weiying Gu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
13
|
The role of VDR and BIM in potentiation of cytarabine-induced cell death in human AML blasts. Oncotarget 2017; 7:36447-36460. [PMID: 27144333 PMCID: PMC5095012 DOI: 10.18632/oncotarget.8998] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/08/2016] [Indexed: 12/16/2022] Open
Abstract
Acute Myeloid Leukemia (AML) has grave prognosis due to aggressive nature of the disease, the toxicity of standard treatment, and overall low cure rates. We recently showed that AML cells in established culture treated with cytarabine (AraC) and a differentiation agent combination show enhancement of AraC cytotoxicity. Here we elucidate molecular changes which underlie this observation with focus on AML blasts in primary culture. The cells were treated with AraC at concentrations achievable in clinical settings, and followed by the addition of Doxercalciferol, a vitamin D2 derivative (D2), together with Carnosic acid (CA), a plant-derived antioxidant. Importantly, although AraC is also toxic to normal bone marrow cell population, the enhanced cell kill by D2/CA was limited to malignant blasts. This enhancement of cell death was associated with activation of the monocytic differentiation program as shown by molecular markers, and the increased expression of vitamin D receptor (VDR). Apoptosis elicited by this treatment is caspase-dependent, and the optimal blast killing required the increased expression of the apoptosis regulator Bim. These data suggest that testing of this regimen in the clinic is warranted.
Collapse
|
14
|
Wang X, Harrison JS, Studzinski GP. BRAF signals to pro-apoptotic BIM to enhance AraC cytotoxicity induced in AML cells by Vitamin D-based differentiation agents. J Steroid Biochem Mol Biol 2017; 173:139-147. [PMID: 27637326 PMCID: PMC5348281 DOI: 10.1016/j.jsbmb.2016.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 01/29/2023]
Abstract
Vitamin D has so far not fulfilled its early promise as an antineoplastic agent, in spite of compelling in vitro data. With the aim of bringing vitamin D or its derivatives (VDDs) effectively to the clinic, we developed a two-pronged approach. First, by adding the plant-derived Carnosic Acid (CA) to a vitamin D2 derivative Doxercalciferol we increased its differentiation potency without increasing it hypercalcemic properties. Second, we added these two agents together to AML cells already treated with Cytarabine (AraC), the standard drug for the treatment of patients with AML. We now report that BRAF, a part of the MAPK signaling pathway, is required for the optimally increased cell death in this system and acts upstream of BIM, the regulator of the caspase cascade that leads to cell death by apoptosis. It is proposed that this therapeutic regimen should be tested in a clinical trial.
Collapse
Affiliation(s)
- Xuening Wang
- Department of Pathology & Laboratory Medicine, NJ Medical School, Rutgers, 185 South Orange Ave., Newark, NJ 07103, United States
| | - Jonathan S Harrison
- Department of Medicine, University of Connecticut, Farmington, CT 06030, United States
| | - George P Studzinski
- Department of Pathology & Laboratory Medicine, NJ Medical School, Rutgers, 185 South Orange Ave., Newark, NJ 07103, United States.
| |
Collapse
|
15
|
Pigneux A, Béné MC, Guardiola P, Recher C, Hamel JF, Sauvezie M, Harousseau JL, Tournilhac O, Witz F, Berthou C, Escoffre-Barbe M, Guyotat D, Fegueux N, Himberlin C, Hunault M, Delain M, Lioure B, Jourdan E, Bauduer F, Dreyfus F, Cahn JY, Sotto JJ, Ifrah N. Addition of Androgens Improves Survival in Elderly Patients With Acute Myeloid Leukemia: A GOELAMS Study. J Clin Oncol 2017; 35:387-393. [DOI: 10.1200/jco.2016.67.6213] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Elderly patients with acute myeloid leukemia (AML) have a poor prognosis, and innovative maintenance therapy could improve their outcomes. Androgens, used in the treatment of aplastic anemia, have been reported to block proliferation of and initiate differentiation in AML cells. We report the results of a multicenter, phase III, randomized open-label trial exploring the benefit of adding androgens to maintenance therapy in patients 60 years of age or older. Patients and Methods A total of 330 patients with AML de novo or secondary to chemotherapy or radiotherapy were enrolled in the study. Induction therapy included idarubicin 8 mg/m2 on days 1 to 5, cytarabine 100 mg/m2 on days 1 to 7, and lomustine 200 mg/m2 on day 1. Patients in complete remission or partial remission received six reinduction courses, alternating idarubicin 8 mg/m2 on day 1, cytarabine 100 mg/m2 on days 1 to 5, and a regimen of methotrexate and mercaptopurine. Patients were randomly assigned to receive norethandrolone 10 or 20 mg/day, according to body weight, or no norethandrolone for a 2-year maintenance therapy regimen. The primary end point was disease-free survival by intention to treat. Secondary end points were event-free survival, overall survival, and safety. This trial was registered at www.ClinicalTrials.gov identifier NCT00700544. Results Random assignment allotted 165 patients to each arm; arm A received norethandrolone, and arm B did not receive norethandrolone. Complete remission or partial remission was achieved in 247 patients (76%). The Schoenfeld time-dependent model showed that norethandrolone significantly improved survival for patients still in remission at 1 year after induction. In arms A and B, respectively, 5-year disease-free survival was 31.2% and 16.2%, event-free survival was 21.5% and 12.9%, and overall survival was 26.3% and 17.2%. Norethandrolone improved outcomes irrelevant to all prognosis factors. Only patients with baseline leukocytes > 30 × 109/L did not benefit from norethandrolone. Conclusion This study demonstrates that maintenance therapy with norethandrolone significantly improves survival in elderly patients with AML without increasing toxicity.
Collapse
Affiliation(s)
- Arnaud Pigneux
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Marie C. Béné
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Philippe Guardiola
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Christian Recher
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Jean-Francois Hamel
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Mathieu Sauvezie
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Jean-Luc Harousseau
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Olivier Tournilhac
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Francis Witz
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Christian Berthou
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Martine Escoffre-Barbe
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Denis Guyotat
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Nathalie Fegueux
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Chantal Himberlin
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Mathilde Hunault
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Martine Delain
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Bruno Lioure
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Eric Jourdan
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Frederic Bauduer
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Francois Dreyfus
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Jean-Yves Cahn
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Jean-Jacques Sotto
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| | - Norbert Ifrah
- Arnaud Pigneux and Mathieu Sauvezie, Centre Hospitalier Universitaire (CHU) Bordeaux, Université Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM) 1035, Bordeaux; Marie C. Béné and Jean-Luc Harousseau, Hôpital Hôtel Dieu, Nantes; Philippe Guardiola, Mathilde Hunault, and Norbert Ifrah, CHU Angers, UMR INSERM U892/CNRS 6299; Jean-Francois Hamel, Université Nantes Angers Le Mans, CHU Angers, Angers; Christian Recher, CHU de Toulouse, Université de Toulouse III, Toulouse; Olivier
| |
Collapse
|
16
|
Wang X, Harrison JS, Studzinski GP. Enhancement of arabinocytosine (AraC) toxicity to AML cells by a differentiation agent combination. J Steroid Biochem Mol Biol 2016; 164:72-78. [PMID: 26319201 PMCID: PMC4769988 DOI: 10.1016/j.jsbmb.2015.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/28/2015] [Accepted: 08/20/2015] [Indexed: 12/11/2022]
Abstract
Arabinocytosine (AraC, also known as cytarabine) is one of the mainstays of AML therapy, but like other DNA damaging therapeutic agents it is rarely curative by itself. There is an emerging realization that the therapeutic outcomes may be improved by combining AraC with other compounds. Here we report that the addition of a differentiating agent combination immediately following AraC damage to AML blasts, selectively increases the cell kill. The experiments were performed using cultured cells from established cell lines of AML (HL60 and U937). The cells were exposed to 100nM AraC, a concentration which produced approximately 25-50% cell kill, followed by a combination of 100nM 1alpha-hydroxyvitamin D2 (1-D2) and 10μM carnosic acid (CA), which together can serve as a powerful differentiating agent combination for AML cells, but are not toxic alone. AraC-induced cell death, measured by annexin V/propidium iodide, was significantly (p<0.01) increased by the 1-D2/CA combination in both cell lines, but not by 1-D2 or CA alone. The enhancement of cell death occurred by both apoptosis and necrosis, was associated with increased DNA damage and with higher levels of DNA damage response (DDR) activated marker Chk1, but the expression of p27, a cell cycle inhibitor protein, was not enhanced by 1-D2/CA. The principal finding is that a vitamin D analog 1-D2 combined with a plant-derived antioxidant CA can markedly augment the cytotoxic action of AraC, an anti-leukemia therapeutic agent.
Collapse
Affiliation(s)
- Xuening Wang
- Department of Pathology & Laboratory Medicine, NJ Medical School, Rutgers, 185 South Orange Ave, Newark, NJ 07103, United States
| | - Jonathan S Harrison
- Department of Medicine, University of Missouri Medical School, Columbia, MO 65212, United States
| | - George P Studzinski
- Department of Pathology & Laboratory Medicine, NJ Medical School, Rutgers, 185 South Orange Ave, Newark, NJ 07103, United States.
| |
Collapse
|
17
|
Nachliely M, Sharony E, Kutner A, Danilenko M. Novel analogs of 1,25-dihydroxyvitamin D 2 combined with a plant polyphenol as highly efficient inducers of differentiation in human acute myeloid leukemia cells. J Steroid Biochem Mol Biol 2016; 164:59-65. [PMID: 26365556 DOI: 10.1016/j.jsbmb.2015.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 12/19/2022]
Abstract
1α,25-Dihydroxyvitamin D3 [1,25(OH)2D3] is known to act as a powerful differentiation inducer in various types of cancer cells, including acute myeloid leukemia (AML) cells. However, supraphysiological concentrations of 1,25(OH)2D3 required to induce terminal maturation of AML cells can cause lethal hypercalcemia in vivo. Here we characterized the differentiation-inducing effects of novel double-point modified analogs of 1,25-dihydroxyvitamin D2 [1,25(OH)2D2], PRI-5201 and PRI-5202 [Pietraszek et al. (2013) Steroids, 78:1003-1014], on HL60, U937 and MOLM-13 human AML cells in comparison with their direct precursors (PRI-1906 and PRI-1907, respectively) and 1,25(OH)2D3. The results demonstrated the following order of potency for the tested compounds: PRI-5202>PRI-1907>PRI-5201>PRI-1906≥1,25(OH)2D3, as determined by measuring the expression of cell surface markers of myeloid differentiation. Particularly, the sensitivity of different AML cell lines to PRI-5201 and PRI-5202 was 3-15-fold and 13-50 fold higher, respectively, compared to that of 1,25(OH)2D3. Importantly, all the analogs tested at 0.25-1nM concentrations retained the ability of 1,25(OH)2D3 to cooperate with the rosemary polyphenol carnosic acid, which strongly potentiated their prodifferentiation activity in a cell type-dependent manner. These synergistic effects were associated with increased induction of the vitamin D receptor (VDR) protein expression. However, surprisingly, carnosic acid was able to significantly enhance only 1,25(OH)2D3-induced transactivation of the direct repeat 3 (DR3)-type vitamin D response element (VDRE), whereas no such cooperation was seen with 1,25(OH)2D2 analogs. Furthermore, dose-response analysis revealed that 1,25(OH)2D3 was more efficacious than the analogs in inducing VDRE activation. This suggests that the superior prodifferentiation activity of the analogs, as compared to 1,25(OH)2D3, may be due to their potential for enhanced activation of the differentiation-related VDRE(s) that differ from the DR3-type element tested in this study. Collectively, the results demonstrate that the new double-point modified 1,25(OH)2D2 analogs are much stronger inducers of myeloid differentiation than 1,25(OH)2D3 and that their efficacy can be further enhanced by combination with plant polyphenols. These combinations warrant their further mechanistic and translational exploration in AML and other types of cancer.
Collapse
Affiliation(s)
- Matan Nachliely
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ehud Sharony
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Andrzej Kutner
- Department of Pharmacology, Pharmaceutical Research Institute, Warsaw 01-793, Poland
| | - Michael Danilenko
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
18
|
Studzinski GP, Harrison JS, Wang X, Sarkar S, Kalia V, Danilenko M. Vitamin D Control of Hematopoietic Cell Differentiation and Leukemia. J Cell Biochem 2016; 116:1500-12. [PMID: 25694395 DOI: 10.1002/jcb.25104] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/23/2015] [Indexed: 12/20/2022]
Abstract
It is now well known that in the mammalian body vitamin D is converted by successive hydroxylations to 1,25-dihydroxyvitamin D (1,25D), a steroid-like hormone with pleiotropic properties. These include important contributions to the control of cell proliferation, survival and differentiation, as well as the regulation of immune responses in disease. Here, we present recent advances in current understanding of the role of 1,25D in myelopoiesis and lymphopoiesis, and the potential of 1,25D and analogs (vitamin D derivatives; VDDs) for the control of hematopoietic malignancies. The reasons for the unimpressive results of most clinical studies of the therapeutic effects of VDDs in leukemia and related diseases may include the lack of a precise rationale for the conduct of these studies. Further, clinical trials to date have generally used extremely heterogeneous patient populations and, in many cases, small numbers of patients, generally without controls. Although low calcemic VDDs have been used and combined with agents that can increase the leukemia cell killing or differentiation effects in acute leukemias, the sequencing of agents used for combination therapy should to be more clearly delineated. Most importantly, it is recommended that in future clinical trials the rationale for the basis of the enhancing action of drug combinations should be clearly articulated and the effects on anticancer immunity should also be evaluated.
Collapse
Affiliation(s)
- George P Studzinski
- Department of Pathology & Laboratory Medicine, Rutgers, NJ Medical School, 185 South Orange Ave, Newark, New Jersey 07103
| | - Jonathan S Harrison
- Department of Medicine, University of Missouri Medical School, One Hospital Drive, Columbia, Missouri 65212
| | - Xuening Wang
- Department of Pathology & Laboratory Medicine, Rutgers, NJ Medical School, 185 South Orange Ave, Newark, New Jersey 07103
| | - Surojit Sarkar
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Vandana Kalia
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Michael Danilenko
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, 84105, Beer-Sheva, Israel
| |
Collapse
|
19
|
Ocio EM, Herrera P, Olave MT, Castro N, Pérez-Simón JA, Brunet S, Oriol A, Mateo M, Sanz MÁ, López J, Montesinos P, Chillón MC, Prieto-Conde MI, Díez-Campelo M, González M, Vidriales MB, Mateos MV, San Miguel JF. Panobinostat as part of induction and maintenance for elderly patients with newly diagnosed acute myeloid leukemia: phase Ib/II panobidara study. Haematologica 2015; 100:1294-300. [PMID: 26160880 DOI: 10.3324/haematol.2015.129577] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/25/2015] [Indexed: 01/08/2023] Open
Abstract
This phase Ib/II trial combined the pan-deacetylase inhibitor panobinostat with chemotherapy followed by panobinostat maintenance in elderly patients with newly diagnosed acute myeloid leukemia. Patients with prior history of myelodysplastic syndrome were excluded and 38 evaluable patients were included in the study (median age: 71 years; range: 65-83). Study patients received an induction with idarubicin (8 mg/m(2) iv days 1-3) plus cytarabine (100 mg/m(2) iv days 1-7) plus panobinostat po at escalating doses (days 8, 10, 12, 15, 17 and 19) that could be repeated in non-responding patients. Patients achieving complete remission received a consolidation cycle with the same schema, followed by panobinostat maintenance (40 mg po 3 days/week) every other week until progression. Thirty-one patients were treated at the maximum tolerated dose of panobinostat in the combination (10 mg) with good tolerability. Complete remission rate was 64% with a time to relapse of 17.0 months (12.8-21.1). Median overall survival for the whole series was 17 months (5.5-28.4). Moreover, in 4 of 5 patients with persistent minimal residual disease before maintenance, panobinostat monotherapy reduced its levels, with complete negativization in two of them. Maintenance phase was well tolerated. The most frequent adverse events were thrombocytopenia (25% grades 3/4), and gastrointestinal toxicity, asthenia and anorexia (mainly grades 1/2). Five patients required dose reduction during this phase, but only one discontinued therapy due to toxicity. These results suggest that panobinostat is one of the first novel agents with activity in elderly acute myeloid leukemia patients, and suggest further investigation is warranted, particularly in the context of maintenance therapy. This trial is registered at clinicaltrials.gov identifier: 00840346.
Collapse
Affiliation(s)
- Enrique M Ocio
- Complejo Asistencial Universitario de Salamanca (IBSAL), Centro Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca
| | | | | | | | - José A Pérez-Simón
- Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Seville
| | | | | | | | | | | | | | - María-Carmen Chillón
- Complejo Asistencial Universitario de Salamanca (IBSAL), Centro Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca
| | - María-Isabel Prieto-Conde
- Complejo Asistencial Universitario de Salamanca (IBSAL), Centro Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca
| | - María Díez-Campelo
- Complejo Asistencial Universitario de Salamanca (IBSAL), Centro Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca
| | - Marcos González
- Complejo Asistencial Universitario de Salamanca (IBSAL), Centro Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca
| | - María-Belén Vidriales
- Complejo Asistencial Universitario de Salamanca (IBSAL), Centro Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca
| | - María-Victoria Mateos
- Complejo Asistencial Universitario de Salamanca (IBSAL), Centro Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca
| | - Jesús F San Miguel
- Clínica Universidad de Navarra, Centro de Investigaciones Médicas Aplicadas (CIMA), IDISNA, Pamplona, Spain
| | | |
Collapse
|
20
|
Kong X, Chen Y, Wang LI, Zhou Y, He Y, Nie W, Zhang X, Yin X. Effect of the microtransplantation of allogeneic hematopoietic stem cells as maintenance therapy for elderly patients with acute leukemia. Oncol Lett 2015; 9:2331-2334. [PMID: 26137066 DOI: 10.3892/ol.2015.2995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 02/10/2015] [Indexed: 12/26/2022] Open
Abstract
The incidence of acute myeloid leukemia (AML) increases with age. Elderly patients with AML are less tolerant to high-intensity consolidation therapy than younger patients, with significantly worse prognoses. Induction and consolidation therapy combined with allogeneic hematopoietic stem cell microtransplantation may improve the prognosis of elderly patients with AML. The present study reports the effect of maintenance therapy with low-dose chemotherapy treatment combined with microtransplantation in elderly patients with AML following consolidation. Between January 2011 and April 2014, three elderly patients (>55 years old), including one 58-year-old patient with acute mixed lineage leukemia (AMLL) and two patients with AML aged 59 years and 62 years, underwent microtransplantation maintenance therapy. Following a complete response to induction chemotherapy and consolidation chemotherapy with two to four cycles of medium dose Ara-c (auto transplantation was performed in the patient with AMLL), maintenance therapy was performed using low-dose Ara-c combined with human leukocyte antigen haploidentical allogeneic hematopoietic stem cell microtransplantation, which was repeated every 3 to 6 months. The patients were followed up for 27, 20 and 16 months, respectively, and all three patients achieved disease-free survival. The bone marrow Wilms' tumor suppression gene (WT1) level of the case with AMLL was dynamically monitored. The results showed that the WT1 level was abnormally high prior to microtransplantation and gradually declined to normal levels subsequent to the process. None of the patients suffered from graft versus host disease during the microtransplantation process. In conclusion, microtransplantation maintenance therapy following consolidation therapy is feasible in elderly patients with AML, and is expected to be able to further remove residual lesions and improve treatment efficacy. A large-scale clinical trial is required to confirm the effect of maintenance therapy in elderly patients with AML.
Collapse
Affiliation(s)
- Xiangjing Kong
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| | - Yongsheng Chen
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| | - L I Wang
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| | - Yali Zhou
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| | - Yuanyuan He
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| | - Weiye Nie
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| | - Xinhua Zhang
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| | - Xiaolin Yin
- Department of Hematology, 303rd Hospital of The People's Liberation Army, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
21
|
Gocek E, Studzinski GP. The Potential of Vitamin D-Regulated Intracellular Signaling Pathways as Targets for Myeloid Leukemia Therapy. J Clin Med 2015; 4:504-34. [PMID: 26239344 PMCID: PMC4470153 DOI: 10.3390/jcm4040504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/06/2015] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
The current standard regimens for the treatment of acute myeloid leukemia (AML) are curative in less than half of patients; therefore, there is a great need for innovative new approaches to this problem. One approach is to target new treatments to the pathways that are instrumental to cell growth and survival with drugs that are less harmful to normal cells than to neoplastic cells. In this review, we focus on the MAPK family of signaling pathways and those that are known to, or potentially can, interact with MAPKs, such as PI3K/AKT/FOXO and JAK/STAT. We exemplify the recent studies in this field with specific relevance to vitamin D and its derivatives, since they have featured prominently in recent scientific literature as having anti-cancer properties. Since microRNAs also are known to be regulated by activated vitamin D, this is also briefly discussed here, as are the implications of the emerging acquisition of transcriptosome data and potentiation of the biological effects of vitamin D by other compounds. While there are ongoing clinical trials of various compounds that affect signaling pathways, more studies are needed to establish the clinical utility of vitamin D in the treatment of cancer.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| | - George P Studzinski
- Department of Pathology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 17101, USA.
| |
Collapse
|
22
|
Recent developments in the treatment of older individuals with acute myeloid leukemia. Curr Opin Hematol 2015; 22:108-15. [DOI: 10.1097/moh.0000000000000120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
23
|
Stopping higher-risk myelodysplastic syndrome in its tracks. Curr Hematol Malig Rep 2014; 9:421-31. [PMID: 25208927 DOI: 10.1007/s11899-014-0234-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Higher-risk myelodysplastic syndromes (MDS) are a collection of diseases associated with poor outcomes from complications related to bone marrow failure and evolution to acute myeloid leukemia. While most patients receive epigenetic therapies, intensive chemotherapy or allogeneic stem cell transplantation, more tolerable and effective treatments are necessary to realize the goal of stopping this disease in its tracks. Recent efforts, building on decades of research exploring the pathogenesis of this disease, have revealed exciting clues that elucidate critical biological features that drive or contribute to MDS, and may serve as targets for selective and well-tolerated future therapies. Here, we review the current diagnostic, prognostic, and therapeutic approaches to higher-risk MDS.
Collapse
|