1
|
Xing T, Hu LJ, Zhao HY, Li CY, Wang ZK, Shen MZ, Lyu ZS, Wang J, Wang Y, Jiang H, Jiang Q, Chang YJ, Zhang XH, Kong Y, Huang XJ. Bone Marrow Endothelial Progenitor Cells remodelling facilitates normal hematopoiesis during Acute Myeloid Leukemia Complete Remission. Nat Commun 2024; 15:10832. [PMID: 39737962 DOI: 10.1038/s41467-024-55051-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Although acute myeloid leukemia (AML) affects hematopoietic stem cell (HSC)-supportive microenvironment, it is largely unknown whether leukemia-modified bone marrow (BM) microenvironment can be remodeled to support normal hematopoiesis after complete remission (CR). As a key element of BM microenvironment, endothelial progenitor cells (EPCs) provide a feasible way to investigate BM microenvironment remodeling. Here, we find reduced and dysfunctional BM EPCs in AML patients, characterized by impaired angiogenesis and high ROS levels, could be partially remodeled after CR and improved by N-acetyl-L-cysteine (NAC). Importantly, HSC-supporting ability of BM EPCs is partially recovered, whereas leukemia-supporting ability is decreased in CR patients. Mechanistically, the transcriptome characteristics of leukemia-modified BM EPCs return to near-normal after CR. In a classic AML mouse and chemotherapy model, BM vasculature and normal hematopoiesis are reversed after CR. In summary, we provide further insights into how leukemia-modified BM microenvironment can be remodeled to support normal hematopoiesis after CR, which can be further improved by NAC.
Collapse
Affiliation(s)
- Tong Xing
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Li-Juan Hu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Hong-Yan Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Chen-Yuan Li
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Zhen-Kun Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Meng-Zhu Shen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Zhong-Shi Lyu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Jing Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Hao Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
2
|
Signoretti C, Matsumura S, Fatehi S, D'Silva M, Mathew R, Cendali F, D'Alessandro A, Alam SMS, Garcia V, Miano JM, Gupte SA. G6pdN126D Variant Increases the Risk of Developing VEGFR (Vascular Endothelial Growth Factor Receptor) Blocker-Induced Pulmonary Vascular Disease. J Am Heart Assoc 2024; 13:e035174. [PMID: 39291493 DOI: 10.1161/jaha.123.035174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND G6PD (glucose-6-phosphate-dehydrogenase) is a key enzyme in the glycolytic pathway and has been implicated in the pathogenesis of cancer and pulmonary hypertension-associated vascular remodeling. Here, we investigated the role of an X-linked G6pd mutation (N126D polymorphism), which is known to increase the risk of cardiovascular disease in individuals from sub-Saharan Africa and many others with African ancestry, in the pathogenesis of pulmonary hypertension induced by a vascular endothelial cell growth factor receptor blocker used for treating cancer. METHODS AND RESULTS CRISPR-Cas9 genome editing was used to generate the G6pd variant (N126D; G6pdN126D) in rats. A single dose of the vascular endothelial cell growth factor receptor blocker sugen-5416 (SU; 20 mg/kg in DMSO), which is currently in a Phase 2/3 clinical trial for cancer treatment, was subcutaneously injected into G6pdN126D rats and their wild-type littermates. After 8 weeks of normoxic conditions, right ventricular pressure and hypertrophy, pulmonary artery remodeling, the metabolic profile, and cytokine expression were assessed. Right ventricular pressure and pulmonary arterial wall thickness were increased in G6PDN126D+SU/normoxic rats. Simultaneously, levels of oxidized glutathione, inositol triphosphate, and intracellular Ca2+ were increased in the lungs of G6PDN126D+SU/normoxic rats, whereas nitric oxide was decreased. Also increased in G6PDN126D+SU/normoxic rats were pulmonary levels of plasminogen activator inhibitor-1, thrombin-antithrombin complex, and expression of proinflammatory cytokines CCL3 (chemokine [C-C motif] ligand), CCL5, and CCL7. CONCLUSIONS Our results suggest G6PDN126D increases inositol triphosphate-Ca2+ signaling, inflammation, thrombosis, and hypertrophic pulmonary artery remodeling in SU-treated rats. This suggests an increased risk of vascular endothelial cell growth factor receptor blocker-induced pulmonary hypertension in those carrying this G6PD variant.
Collapse
MESH Headings
- Animals
- Glucosephosphate Dehydrogenase/genetics
- Glucosephosphate Dehydrogenase/metabolism
- Receptors, Vascular Endothelial Growth Factor/genetics
- Rats
- Male
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/physiopathology
- Disease Models, Animal
- Vascular Remodeling/drug effects
- Rats, Sprague-Dawley
- Indoles/pharmacology
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/physiopathology
- Pyrroles
Collapse
Affiliation(s)
| | - Shun Matsumura
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Samuel Fatehi
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Melinee D'Silva
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Rajamma Mathew
- Department of Medicine, Division of Pediatric Cardiology, Physiology New York Medical College Valhalla NY USA
| | - Francesca Cendali
- Department of Biochemistry and Molecular Genetics University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics University of Colorado Anschutz Medical Campus Aurora CO USA
| | - S M Shafiqul Alam
- Department of Pathology, Microbiology, and Immunology (PMI) New York Medical College Valhalla NY USA
| | - Victor Garcia
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Joseph M Miano
- Department of Medicine Vascular Biology Center, Medical College of Georgia at Augusta University Augusta GA USA
| | - Sachin A Gupte
- Department of Pharmacology New York Medical College Valhalla NY USA
| |
Collapse
|
3
|
Lap CJ, Abrahim MS, Nassereddine S. Perspectives and challenges of small molecule inhibitor therapy for FLT3-mutated acute myeloid leukemia. Ann Hematol 2024; 103:2215-2229. [PMID: 37975931 DOI: 10.1007/s00277-023-05545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disease characterized overall by an aggressive clinical course. The underlying genetic abnormalities present in leukemic cells contribute significantly to the AML phenotype. Mutations in FMS-like tyrosine kinase 3 (FLT3) are one of the most common genetic abnormalities identified in AML, and the presence of these mutations strongly influences disease presentation and negatively impacts prognosis. Since mutations in FLT3 were identified in AML, they have been recognized as a valid therapeutic target resulting in decades of research to develop effective small molecule inhibitor treatment that could improve outcome for these patients. Despite the approval of several FLT3 inhibitors over the last couple of years, the treatment of patients with FLT3-mutated AML remains challenging and many questions still need to be addressed. This review will provide an up-to-date overview of our current understanding of FLT3-mutated AML and discuss what the current status is of the available FLT3 inhibitors for the day-to-day management of this aggressive disease.
Collapse
Affiliation(s)
- Coen J Lap
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Marwa Sh Abrahim
- The George Washington Cancer Center, George Washington University, Washington, DC, USA
| | - Samah Nassereddine
- The George Washington Cancer Center, George Washington University, Washington, DC, USA.
| |
Collapse
|
4
|
Garcia-Hernandez V, Raya-Sandino A, Azcutia V, Miranda J, Kelm M, Flemming S, Birkl D, Quiros M, Brazil JC, Parkos CA, Nusrat A. Inhibition of Soluble Stem Cell Factor Promotes Intestinal Mucosal Repair. Inflamm Bowel Dis 2023; 29:1133-1144. [PMID: 36688460 PMCID: PMC10320368 DOI: 10.1093/ibd/izad003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Incidences of inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, are escalating worldwide and can be considered a global public health problem. Given that the gold standard approach to IBD therapeutics focuses on reducing the severity of symptoms, there is an urgent unmet need to develop alternative therapies that halt not only inflammatory processes but also promote mucosal repair. Previous studies have identified increased stem cell factor (SCF) expression in inflamed intestinal mucosal tissues. However, the role that SCF plays in mediating intestinal inflammation and repair has not been explored. METHODS Changes in the expression of SCF were evaluated in the colonic tissue of healthy mice and during dextran sodium sulfate (DSS)-induced colitis. Furthermore, mucosal wound healing and colitis severity were analyzed in mice subjected to either mechanical biopsy or DSS treatment, respectively, following intestinal epithelial cell-specific deletion of SCF or anti-SCF antibody administration. RESULTS We report robust expression of SCF by intestinal epithelial cells during intestinal homeostasis with a switch to immune cell-produced SCF during colitis. Data from mice with intestinal epithelial cell-specific deletion of SCF highlight the importance of immune cell-produced SCF in driving the pathogenesis of colitis. Importantly, antibody-mediated neutralization of total SCF or the specific SCF248 isoform decreased immune cell infiltration and enhanced mucosal wound repair following biopsy-induced colonic injury or DSS-induced colitis. CONCLUSIONS These data demonstrate that SCF functions as a pro-inflammatory mediator in mucosal tissues and that specific neutralization of SCF248 could be a viable therapeutic option to reduce intestinal inflammation and promote mucosal wound repair in individuals with IBD.
Collapse
Affiliation(s)
| | - Arturo Raya-Sandino
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jael Miranda
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthias Kelm
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sven Flemming
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dorothee Birkl
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Miguel Quiros
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Swaminathan M, Aly MM, Khan AM, Share BA, Dhillon V, Lalo E, Ramos H, Akers KG, Kim S, Balasubramanian S. Efficacy analysis of different FLT3 inhibitors in patients with relapsed/refractory acute myeloid leukemia and high-risk myelodysplastic syndrome. EJHAEM 2023; 4:165-173. [PMID: 36819163 PMCID: PMC9928788 DOI: 10.1002/jha2.616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022]
Abstract
Several FLT3 inhibitors(i) are available to treat relapsed/refractory (R/R) FLT3-internal tandem duplicated acute myeloid leukemia (AML). This study analyzes the efficacies of various FLT3i (types 1 and 2) tested in clinical trials in treating R/R AML and high-risk myelodysplastic syndromes (HR-MDS). PubMed and EMBASE databases were searched for single/double-arm phase I/II/III R/R AML or HR-MDS clinical trials published between 1/1/2000 and 6/1/2021. The outcomes studied were composite response rate (CRc) and overall response rate (ORR). Toxicities were compared based on the organ system. The 28 studies analyzed had 1927 patients. The pooled ORR and (CRc) for all FLT3i were 53% (95% CI, 43%-63%) and 34% (95% CI, 26%-44%). Pooled ORR and CRc were 37% (95% CI, 25%-51%) and 35% (95% CI, 21%-52%) for type 1 and 58% (95% CI, 43%-71%) and 38% (95% CI, 27%-50%) for type 2, respectively. Gastrointestinal (GI) and hematological toxicity occurred in 22% (95% CI, 19%-25.4%) and 74.6% (95% CI, 70%-79%) with type 1 and 13.9% (95% CI, 12%-16%) and 57.7% (95% CI, 54.6%-60.8%) with type 2 FLT3i. QTc prolongation occurred in 2.06% (95% CI, 1.03%-3.65%) with type 1 and 7% (95% CI, 5.3%-9%) with type 2 FLT3i. Type 2 FLT3i had less GI toxicity but more QTc prolongation. Prospective studies are needed to compare the efficacy of type 1 and 2 FLT3i.
Collapse
Affiliation(s)
- Mahesh Swaminathan
- Department of MedicineRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Mai M. Aly
- Clinical Hematology UnitInternal Medicine DepartmentAssiut University HospitalAssiutEgypt
| | - Abdul Moiz Khan
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
| | - Bayan Al Share
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
| | - Vikram Dhillon
- Department of Internal MedicineWayne State University School of MedicineDetroitMichiganUSA
| | - Enxhi Lalo
- Wayne State University School of MedicineDetroitMichiganUSA
| | - Harry Ramos
- Wayne State University School of MedicineDetroitMichiganUSA
| | | | - Seongho Kim
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
- Biostatistics and Bioinformatics CoreKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
| | - Suresh Balasubramanian
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
- Translational Hematology and Oncology ResearchTaussig Cancer Institute, Cleveland ClinicClevelandOhioUSA
| |
Collapse
|
6
|
Seo W, Silwal P, Song IC, Jo EK. The dual role of autophagy in acute myeloid leukemia. J Hematol Oncol 2022; 15:51. [PMID: 35526025 PMCID: PMC9077970 DOI: 10.1186/s13045-022-01262-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/14/2022] [Indexed: 01/18/2023] Open
Abstract
Acute myeloid leukemia (AML) is a severe hematologic malignancy prevalent in older patients, and the identification of potential therapeutic targets for AML is problematic. Autophagy is a lysosome-dependent catabolic pathway involved in the tumorigenesis and/or treatment of various cancers. Mounting evidence has suggested that autophagy plays a critical role in the initiation and progression of AML and anticancer responses. In this review, we describe recent updates on the multifaceted functions of autophagy linking to genetic alterations of AML. We also summarize the latest evidence for autophagy-related genes as potential prognostic predictors and drivers of AML tumorigenesis. We then discuss the crosstalk between autophagy and tumor cell metabolism into the impact on both AML progression and anti-leukemic treatment. Moreover, a series of autophagy regulators, i.e., the inhibitors and activators, are described as potential therapeutics for AML. Finally, we describe the translation of autophagy-modulating therapeutics into clinical practice. Autophagy in AML is a double-edged sword, necessitating a deeper understanding of how autophagy influences dual functions in AML tumorigenesis and anti-leukemic responses.
Collapse
Affiliation(s)
- Wonhyoung Seo
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Korea.,Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Korea.,Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Korea
| | - Prashanta Silwal
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Korea.,Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Korea
| | - Ik-Chan Song
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, 35015, Korea
| | - Eun-Kyeong Jo
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Korea. .,Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Korea. .,Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Korea.
| |
Collapse
|
7
|
Dausinas Ni P, Basile C, Junge C, Hartman M, O’Leary HA. Hypoxia and Hematopoiesis. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-021-00203-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
8
|
Zhong J, Wu H, Bu X, Li W, Cai S, Du M, Gao Y, Ping B. Establishment of Prognosis Model in Acute Myeloid Leukemia Based on Hypoxia Microenvironment, and Exploration of Hypoxia-Related Mechanisms. Front Genet 2021; 12:727392. [PMID: 34777463 PMCID: PMC8578022 DOI: 10.3389/fgene.2021.727392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/22/2021] [Indexed: 01/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematologic neoplasm with poor survival outcomes. However, the routine clinical features are not sufficient to accurately predict the prognosis of AML. The expression of hypoxia-related genes was associated with survival outcomes of a variety of hematologic and lymphoid neoplasms. We established an 18-gene signature-based hypoxia-related prognosis model (HPM) and a complex model that consisted of the HPM and clinical risk factors using machine learning methods. Both two models were able to effectively predict the survival of AML patients, which might contribute to improving risk classification. Differentially expressed genes analysis, Gene Ontology (GO) categories, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed to reveal the underlying functions and pathways implicated in AML development. To explore hypoxia-related changes in the bone marrow immune microenvironment, we used CIBERSORT to calculate and compare the proportion of 22 immune cells between the two groups with high and low hypoxia-risk scores. Enrichment analysis and immune cell composition analysis indicated that the biological processes and molecular functions of drug metabolism, angiogenesis, and immune cell infiltration of bone marrow play a role in the occurrence and development of AML, which might help us to evaluate several hypoxia-related metabolic and immune targets for AML therapy.
Collapse
Affiliation(s)
- Jinman Zhong
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hang Wu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyin Bu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiru Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengchun Cai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meixue Du
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ya Gao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Baohong Ping
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Huiqiao, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Golla U, Ehudin MA, Annageldiyev C, Zeng Z, Bastihalli Tukaramrao D, Tarren A, Date AA, Elcheva I, Berg A, Amin S, Loughran TP, Kester M, Desai D, Dovat S, Claxton D, Sharma A. DJ4 Targets the Rho-Associated Protein Kinase Pathway and Attenuates Disease Progression in Preclinical Murine Models of Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:4889. [PMID: 34638385 PMCID: PMC8508452 DOI: 10.3390/cancers13194889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 01/22/2023] Open
Abstract
The poor prognosis of acute myeloid leukemia (AML) and the highly heterogenous nature of the disease motivates targeted gene therapeutic investigations. Rho-associated protein kinases (ROCKs) are crucial for various actin cytoskeletal changes, which have established malignant consequences in various cancers, yet are still not being successfully utilized clinically towards cancer treatment. This work establishes the therapeutic activity of ROCK inhibitor (5Z)-2-5-(1H-pyrrolo[2,3-b]pyridine-3-ylmethylene)-1,3-thiazol-4(5H)-one (DJ4) in both in vitro and in vivo preclinical models of AML to highlight the potential of this class of inhibitors. Herein, DJ4 induced cytotoxic and proapoptotic effects in a dose-dependent manner in human AML cell lines (IC50: 0.05-1.68 μM) and primary patient cells (IC50: 0.264-13.43 μM); however, normal hematopoietic cells were largely spared. ROCK inhibition by DJ4 disrupts the phosphorylation of downstream targets, myosin light chain (MLC2) and myosin-binding subunit of MLC phosphatase (MYPT), yielding a potent yet selective treatment response at micromolar concentrations, from 0.02 to 1 μM. Murine models injected with luciferase-expressing leukemia cell lines subcutaneously or intravenously and treated with DJ4 exhibited an increase in overall survival and reduction in disease progression relative to the vehicle-treated control mice. Overall, DJ4 is a promising candidate to utilize in future investigations to advance the current AML therapy.
Collapse
Affiliation(s)
- Upendarrao Golla
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (C.A.); (A.T.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
| | - Melanie A. Ehudin
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.A.E.); (D.B.T.); (I.E.)
| | - Charyguly Annageldiyev
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (C.A.); (A.T.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
| | - Zheng Zeng
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (Z.Z.); (M.K.)
| | - Diwakar Bastihalli Tukaramrao
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.A.E.); (D.B.T.); (I.E.)
| | - Anna Tarren
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (C.A.); (A.T.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
| | - Abhijit A. Date
- The Daniel K. Inouye College of Pharmacy, University of Hawaii, Hilo, HI 96720, USA;
| | - Irina Elcheva
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.A.E.); (D.B.T.); (I.E.)
| | - Arthur Berg
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
| | - Shantu Amin
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (Z.Z.); (M.K.)
| | - Thomas P. Loughran
- Department of Medicine, Division of Hematology and Oncology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA;
- Department of Medicine, Division of Hematology and Oncology, University of Virginia Cancer Center, Charlottesville, VA 22903, USA
| | - Mark Kester
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (Z.Z.); (M.K.)
- Department of Medicine, Division of Hematology and Oncology, University of Virginia Cancer Center, Charlottesville, VA 22903, USA
| | - Dhimant Desai
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (Z.Z.); (M.K.)
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.A.E.); (D.B.T.); (I.E.)
| | - David Claxton
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (C.A.); (A.T.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
| | - Arati Sharma
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (C.A.); (A.T.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.B.); (S.A.); (D.D.)
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (Z.Z.); (M.K.)
| |
Collapse
|
10
|
Xu C, Liu Y, Zhao G. The development of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy. Curr Med Chem 2021; 29:1891-1919. [PMID: 34465277 DOI: 10.2174/0929867328666210831142311] [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: 02/25/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 11/22/2022]
Abstract
Kinases are pivotal regulators in tumorigenesis and metastasis by modulating the expression of oncogenes and the transcription of antioncogenes directly or indirectly. Correspondingly, multifarious 3-substituted indolin-2-one derivatives as selective kinase inhibitors for cancer therapy exhibited a low nanomolar activity with prominent efficacy, superior response rate and admirable tolerability. Particularly, certain 3-substituted indolin-2-one derivatives have met the requirements for clinical trials or the pharmaceutical market. Herein, we focus on the traits of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy, overview recent progress of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy, analyze the selectivity for tyrosine kinases inhibitors and serine/threonine kinases inhibitors from the molecular aspects based on the molecular docking studies, summarize the structure-activity relationships (SARs) as selective kinase inhibitors and provide our perspectives for the development of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Changqing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong. China
| | - Yang Liu
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas. United States
| | - Guisen Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong. China
| |
Collapse
|
11
|
Soyfer EM, Fleischman AG. Inflammation in Myeloid Malignancies: From Bench to Bedside. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:160-167. [PMID: 35663100 PMCID: PMC9138438 DOI: 10.36401/jipo-21-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/21/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022]
Abstract
Myeloid malignancies, stemming from a somatically mutated hematopoietic clone, can cause a wide variety of clinical consequences, including pancytopenia in myelodysplastic syndrome, overproduction of three myeloid lineages in myeloproliferative neoplasm, and the rapid growth of immature hematopoietic cells in acute myeloid leukemia (AML). It is becoming clear that inflammation is a hallmark feature of clonal myeloid conditions, ranging from clonal hematopoiesis of indeterminate potential to AML. Fundamental findings from laboratory research on inflammation in myeloid malignancies has potential implications for diagnosis, prognostication, and treatment in these diseases. In this review, we highlighted some pertinent basic science findings regarding the role of inflammation in myeloid malignancies and speculated how these findings could impact the clinical care of patients.
Collapse
Affiliation(s)
- Eli M Soyfer
- School of Medicine, University of California, Irvine, CA, USA
| | - Angela G Fleischman
- Division of Hematology/Oncology, UC Irvine Health, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, USA
| |
Collapse
|
12
|
Mosteo L, Storer J, Batta K, Searle EJ, Duarte D, Wiseman DH. The Dynamic Interface Between the Bone Marrow Vascular Niche and Hematopoietic Stem Cells in Myeloid Malignancy. Front Cell Dev Biol 2021; 9:635189. [PMID: 33777944 PMCID: PMC7991089 DOI: 10.3389/fcell.2021.635189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells interact with bone marrow niches, including highly specialized blood vessels. Recent studies have revealed the phenotypic and functional heterogeneity of bone marrow endothelial cells. This has facilitated the analysis of the vascular microenvironment in steady state and malignant hematopoiesis. In this review, we provide an overview of the bone marrow microenvironment, focusing on refined analyses of the marrow vascular compartment performed in mouse studies. We also discuss the emerging role of the vascular niche in “inflamm-aging” and clonal hematopoiesis, and how the endothelial microenvironment influences, supports and interacts with hematopoietic cells in acute myeloid leukemia and myelodysplastic syndromes, as exemplar states of malignant myelopoiesis. Finally, we provide an overview of strategies for modulating these bidirectional interactions to therapeutic effect in myeloid malignancies.
Collapse
Affiliation(s)
- Laura Mosteo
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Joanna Storer
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Kiran Batta
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Emma J Searle
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Delfim Duarte
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.,Department of Biomedicine, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal.,Department of Onco-Hematology, Instituto Português de Oncologia (IPO)-Porto, Porto, Portugal
| | - Daniel H Wiseman
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
13
|
Hu X, Wang Y, Gao X, Xu S, Zang L, Xiao Y, Li Z, Hua H, Xu J, Li D. Recent Progress of Oridonin and Its Derivatives for the Treatment of Acute Myelogenous Leukemia. Mini Rev Med Chem 2020; 20:483-497. [PMID: 31660811 DOI: 10.2174/1389557519666191029121809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/13/2019] [Accepted: 09/06/2019] [Indexed: 01/03/2023]
Abstract
First stage human clinical trial (CTR20150246) for HAO472, the L-alanine-(14-oridonin) ester trifluoroacetate, was conducted by a Chinese company, Hengrui Medicine Co. Ltd, to develop a new treatment for acute myelogenous leukemia. Two patents, WO2015180549A1 and CN201410047904.X, covered the development of the I-type crystal, stability experiment, conversion rate research, bioavailability experiment, safety assessment, and solubility study. HAO472 hewed out new avenues to explore the therapeutic properties of oridonin derivatives and develop promising treatment of cancer originated from naturally derived drug candidates. Herein, we sought to overview recent progress of the synthetic, physiological, and pharmacological investigations of oridonin and its derivatives, aiming to disclose the therapeutic potentials and broaden the platform for the discovery of new anticancer drugs.
Collapse
Affiliation(s)
- Xu Hu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yan Wang
- Valiant Co. Ltd., 11 Wuzhishan Road, YEDA Yantai, Shandong 264006, China
| | - Xiang Gao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Shengtao Xu
- Department of Medicinal Chemistry and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Linghe Zang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yan Xiao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Zhanlin Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Jinyi Xu
- Department of Medicinal Chemistry and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
14
|
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: 0.8] [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
|
15
|
Wang H, Bai J, Pei Z, Zhang B, Wang J, Lian X, Song Q. Venetoclax + hypomethylating agents combined with dose-adjusted HAG for relapsed/refractory acute myeloid leukemia: Two case reports. Medicine (Baltimore) 2020; 99:e23265. [PMID: 33217852 PMCID: PMC7676608 DOI: 10.1097/md.0000000000023265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
RATIONALE Some acute myeloid leukemia (AML) patients are unresponsive to treatment or have remission followed by worsening of disease (known as relapsed/refractory AML [R/RAML]) after standardized treatment. The CAG/HAG regimen is not often used clinically because heterogenous patient responses, resistance, and hematopoietic bone marrow dysfunction have been reported with its use. We present 2 cases of R/RAML treated with a new combined therapy (venetoclax+ hypomethylating agents [HMAs]) in which the HAG dose was adjusted and effective in the first course of treatment. PATIENT CHARACTERISTICS Case 1 involved a 23-year-old man who had suffered from AML for >4 years, and his FLT3 mutation status was positive at the initial diagnosis. After the first course of treatment with the standard-dose "Da" plan, the patient experienced complete remission. During the subsequent courses of treatment, the patient experienced 6 recurrences and was treated with the "ID Ara-C + MIT + sidaaniline" and "CAG + sidaaniline" regimens. However, the disease did not respond. Case 2 involved a 26-year-old man who received chemotherapy with the "Da," "ID Ara-C," "decitabine + half-dose CAG," and "HAE" regimens. In this patients, remission could not be achieved. Reintroduction of the "ia" scheme also failed after treatment in our hospital. DIAGNOSIS Two patients were diagnosed with R/RAML. INTERVENTIONS The patient in case 2 received chemotherapy interventions, whereas the patient in case 1 refused to receive medical services at our hospital. OUTCOMES The patient in case 1 was discharged after complete response treatment due to economic reasons and relapsed 2 months later. The patient ultimately died of infection and heart failure. The patient in case 2 is receiving a second cycle of chemotherapy. LESSONS We recommend the "venetoclax + HMAs combined with dose-adjusted CAH/HAG" regimen as an effective treatment for adult R/RAML.
Collapse
Affiliation(s)
- Hongxia Wang
- Department of Hematology, Jiaozuo People's Hospital
| | - Junjun Bai
- Department of Hematology, Jiaozuo People's Hospital
| | - Zhixin Pei
- Department of Hematology, Jiaozuo People's Hospital
| | - Bei Zhang
- Department of Hematology, Jiaozuo People's Hospital
| | - Junjie Wang
- Department of Hematology, Jiaozuo People's Hospital
| | - Xingli Lian
- Pharmacy Intravennous Admixture Service, Second People's Hospital of Jiaozuo, Jiaozuo, China
| | - Qinglin Song
- Department of Hematology, Jiaozuo People's Hospital
| |
Collapse
|
16
|
Tyrosine kinase inhibitors for acute myeloid leukemia: A step toward disease control? Blood Rev 2020; 44:100675. [DOI: 10.1016/j.blre.2020.100675] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/17/2020] [Accepted: 02/11/2020] [Indexed: 12/24/2022]
|
17
|
Pievani A, Biondi M, Tomasoni C, Biondi A, Serafini M. Location First: Targeting Acute Myeloid Leukemia Within Its Niche. J Clin Med 2020; 9:E1513. [PMID: 32443460 PMCID: PMC7290711 DOI: 10.3390/jcm9051513] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Despite extensive research and development of new treatments, acute myeloid leukemia (AML)-backbone therapy has remained essentially unchanged over the last decades and is frequently associated with poor outcomes. Eradicating the leukemic stem cells (LSCs) is the ultimate challenge in the treatment of AML. Emerging evidence suggests that AML remodels the bone marrow (BM) niche into a leukemia-permissive microenvironment while suppressing normal hematopoiesis. The mechanism of stromal-mediated protection of leukemic cells in the BM is complex and involves many adhesion molecules, chemokines, and cytokines. Targeting these factors may represent a valuable approach to complement existing therapies and overcome microenvironment-mediated drug resistance. Some strategies for dislodging LSCs and leukemic blasts from their protective niche have already been tested in patients and are in different phases of the process of clinical development. Other strategies, such as targeting the stromal cells remodeling processes, remain at pre-clinical stages. Development of humanized xenograft mouse models, which overcome the mismatch between human leukemia cells and the mouse BM niche, is required to generate physiologically relevant, patient-specific human niches in mice that can be used to unravel the role of human AML microenvironment and to carry out preclinical studies for the development of new targeted therapies.
Collapse
Affiliation(s)
- Alice Pievani
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, 20900 Monza, Italy; (A.P.); (M.B.); (C.T.)
| | - Marta Biondi
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, 20900 Monza, Italy; (A.P.); (M.B.); (C.T.)
| | - Chiara Tomasoni
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, 20900 Monza, Italy; (A.P.); (M.B.); (C.T.)
| | - Andrea Biondi
- Department of Pediatrics, Pediatric Hematology-Oncology Unit, Fondazione MBBM/San Gerardo Hospital, 20900 Monza, Italy;
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, 20900 Monza, Italy; (A.P.); (M.B.); (C.T.)
| |
Collapse
|
18
|
Deng M, Zha J, Zhao H, Jia X, Shi Y, Li Z, Fu G, Yu L, Fang Z, Xu B. Apatinib exhibits cytotoxicity toward leukemia cells by targeting VEGFR2-mediated prosurvival signaling and angiogenesis. Exp Cell Res 2020; 390:111934. [PMID: 32126236 DOI: 10.1016/j.yexcr.2020.111934] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Vascular permeability contributes to disease progression and drug resistance in hematological malignancies, including AML. Thus, targeting angiogenic signaling is a promising treatment strategy, especially for relapsed and resistant AML. The aim of this study was to evaluate the efficacy of apatinib, a novel receptor tyrosine kinase inhibitor that selectively targets VEGFR2. METHODS Several AML cell lines were exposed to various concentrations of apatinib, and then CCK8 and Annexin V/PI assays were performed to determine IC50 values and apoptosis, respectively. The effect of apatinib against primary AML cells from 57 adult patients and 11 normal controls was also analyzed utilizing an apoptosis assay. Next, we tested the underlying mechanism of apatinib in AML using western blotting and mass cytometry (CyTOF). Finally, the activity of apatinib against tumor growth and angiogenesis was further evaluated in vivo in xenograft models. RESULTS We found apatinib significantly inhibited growth and promoted apoptosis in AML cell lines in vitro. Similarly, apatinib showed cytotoxicity against primary AML cells but didn't affect normal BMMCs. Its effect was highly correlated with several clinical features, such as NPM1 mutation, extramedullary infiltration, relapsed/refractory disease, and M2 and M5 FAB subtypes. In addition, apatinib suppressed AML growth and attenuated angiogenesis in xenograft models. Mechanistically, apatinib-induced cytotoxicity was closely associated with inhibition of the VEGFR2-mediated Src/STAT3 and AKT/mTOR pathways and induction of mitochondria-mediated apoptosis. CONCLUSION Apatinib exerts antileukemia effects by targeting VEGFR2-induced prosurvival signaling and angiogenesis, thus providing a rationale for the application of apatinib in AML.
Collapse
Affiliation(s)
- Manman Deng
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, PR China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, PR China
| | - Haijun Zhao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, PR China
| | - Xian Jia
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Yuanfei Shi
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, PR China
| | - Zhifeng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, PR China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Lian Yu
- Department of Hematology and Rheumatology, Longyan First Hospital, Affiliated to Fujian Medical University, Longyan, 364000, PR China
| | - Zhihong Fang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, PR China.
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, PR China.
| |
Collapse
|
19
|
Targeting Tyrosine Kinases in Acute Myeloid Leukemia: Why, Who and How? Int J Mol Sci 2019; 20:ijms20143429. [PMID: 31336846 PMCID: PMC6679203 DOI: 10.3390/ijms20143429] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 12/21/2022] Open
Abstract
Acute myeloid leukemia (AML) is a myeloid malignancy carrying a heterogeneous molecular panel of mutations participating in the blockade of differentiation and the increased proliferation of myeloid hematopoietic stem and progenitor cells. The historical "3 + 7" treatment (cytarabine and daunorubicin) is currently challenged by new therapeutic strategies, including drugs depending on the molecular landscape of AML. This panel of mutations makes it possible to combine some of these new treatments with conventional chemotherapy. For example, the FLT3 receptor is overexpressed or mutated in 80% or 30% of AML, respectively. Such anomalies have led to the development of targeted therapies using tyrosine kinase inhibitors (TKIs). In this review, we document the history of TKI targeting, FLT3 and several other tyrosine kinases involved in dysregulated signaling pathways.
Collapse
|
20
|
Kessler T, Koschmieder S, Schliemann C, Crysandt M, Mikesch JH, von Stillfried S, Stelljes M, Pohlen M, Lenz G, Kirsch A, Vehring K, Wardelmann E, Hartmann W, Bormann E, Gerss J, Brümmendorf TH, Müller-Tidow C, Berdel WE. Phase II clinical trial of pazopanib in patients with acute myeloid leukemia (AML), relapsed or refractory or at initial diagnosis without an intensive treatment option (PazoAML). Ann Hematol 2019; 98:1393-1401. [DOI: 10.1007/s00277-019-03651-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/02/2019] [Indexed: 12/01/2022]
|
21
|
Sansom GN, Kirk NS, Guise CP, Anderson RF, Smaill JB, Patterson AV, Kelso MJ. Prototyping kinase inhibitor-cytotoxin anticancer mutual prodrugs activated by tumour hypoxia: A chemical proof of concept study. Bioorg Med Chem Lett 2019; 29:1215-1219. [PMID: 30885680 DOI: 10.1016/j.bmcl.2019.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/07/2019] [Accepted: 03/10/2019] [Indexed: 01/16/2023]
Abstract
Amide- and ester-linked kinase inhibitor-cytotoxin conjugates were rationally designed and synthesised as prototype hypoxia-activated anticancer mutual prodrugs. Chemical reduction of an aryl nitro trigger moiety was shown to initiate a spontaneous cyclisation/fragmentation reaction that simultaneously released the kinase inhibitor semaxanib (SU5416) and the amine- or alcohol-linked cytotoxin from the prodrugs. Preliminary cell testing and reduction potential measurements support optimisation of the compounds towards tumour-selective mutual prodrugs.
Collapse
Affiliation(s)
- Geraud N Sansom
- Molecular Horizons and School of Chemistry & Molecular Bioscience, University of Wollongong, NSW 2522, Australia; Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Nicholas S Kirk
- Molecular Horizons and School of Chemistry & Molecular Bioscience, University of Wollongong, NSW 2522, Australia; Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Christopher P Guise
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; The Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Robert F Anderson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; The Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jeff B Smaill
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; The Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Adam V Patterson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; The Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Michael J Kelso
- Molecular Horizons and School of Chemistry & Molecular Bioscience, University of Wollongong, NSW 2522, Australia; Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia.
| |
Collapse
|
22
|
Behrmann L, Wellbrock J, Fiedler W. Acute Myeloid Leukemia and the Bone Marrow Niche-Take a Closer Look. Front Oncol 2018; 8:444. [PMID: 30370251 PMCID: PMC6195156 DOI: 10.3389/fonc.2018.00444] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
The bone marrow is the home of hematopoiesis and is therefore a hotspot for the development of hematopoietic diseases. Complex interactions between the bone marrow microenvironment and hematopoietic stem cells must find a balance between proliferation, differentiation and homeostasis of the stem cell compartment. Changes in this tightly regulated network can provoke malignant transformation, leading to hematopoietic diseases. Here we focus on acute myeloid leukemia (AML), since this is the most frequent acute leukemia in adulthood with very poor overall survival rates and where relapse after chemotherapy continues to be a major challenge, driving demand for new therapeutic strategies. Current research is focusing on the identification of specific interactions between leukemic blasts and their niche components, which may be exploited as novel treatment targets along with induction chemotherapy. Significant progress has been gained over the last few years in the field of high-resolution imaging. Confocal ex vivo and intravital microscopy have revealed a detailed map of bone marrow structures and components; as well as identifying numerous alterations in the stem cell niche that correspond to disease progression. However, the underlying mechanisms are still not completely understood and due to the complexity, their elucidation remains a challenging. This review discusses the constitution of the AML niche in the bone marrow, the improvement in visualization of the complex three-dimensional niche structures and points out new therapeutic strategies to increase the overall survival of AML patients.
Collapse
Affiliation(s)
- Lena Behrmann
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
23
|
Cheng H, Sun G, Cheng T. Hematopoiesis and microenvironment in hematological malignancies. CELL REGENERATION 2018; 7:22-26. [PMID: 30671226 PMCID: PMC6326248 DOI: 10.1016/j.cr.2018.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022]
Abstract
Adult hematopoietic stem cells (HSCs) and progenitors (HPCs) reside in the bone marrow, a highly orchestrated architecture. In the bone marrow, the process of how HSCs exert self-renewal and differentiation is tightly regulated by the surrounding microenvironment, or niche. Recent advances in imaging technologies and numerous knockout or knockin mouse models have greatly improved our understanding of the organization of the bone marrow niche. This niche compartment includes a complex network of mesenchymal stem cells (MSC), osteolineage cells, endothelial cells (arterioles and sinusoids), sympathetic nerves, nonmyelinating Schwann cells and megakaryocytes. In addition, different types of mediators, such as cytokines/chemokines, reactive oxygen species (ROS) and exosomes play a pivotal role in regulating the function of hematopoietic cells. Therefore, the niche components and the hematopoietic system make up an ecological environment that maintains the homeostasis and responds to stress, damage or disease conditions. On the other hand, the niche compartment can become a traitor that can do harm to normal hematopoietic cells under pathological conditions. Studies on the diseased bone marrow niche have only recently begun to appear in the extant literature. In this short review, we discuss the most recent advances regarding the behaviors of normal hematopoietic cells and their niche alterations in hematological malignancies.
Collapse
Affiliation(s)
- Hui Cheng
- State Key Laboratory of Experimental Hematology, China.,Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China
| | - Guohuan Sun
- State Key Laboratory of Experimental Hematology, China.,Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, China.,Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China
| |
Collapse
|
24
|
Yehya AHS, Asif M, Petersen SH, Subramaniam AV, Kono K, Majid AMSA, Oon CE. Angiogenesis: Managing the Culprits behind Tumorigenesis and Metastasis. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E8. [PMID: 30344239 PMCID: PMC6037250 DOI: 10.3390/medicina54010008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 12/14/2022]
Abstract
Deregulated angiogenesis has been identified as a key contributor in a number of pathological conditions including cancer. It is a complex process, which involves highly regulated interaction of multiple signalling molecules. The pro-angiogenic signalling molecule, vascular endothelial growth factor (VEGF) and its cognate receptor 2 (VEGFR-2), which is often highly expressed in majority of human cancers, plays a central role in tumour angiogenesis. Owing to the importance of tumour vasculature in carcinogenesis, tumour blood vessels have emerged as an excellent therapeutic target. The anti-angiogenic therapies have been shown to arrest growth of solid tumours through multiple mechanisms, halting the expansion of tumour vasculature and transient normalization of tumour vasculature which help in the improvement of blood flow resulting in more uniform delivery of cytotoxic agents to the core of tumour mass. This also helps in reduction of hypoxia and interstitial pressure leading to reduced chemotherapy resistance and more uniform delivery of cytotoxic agents at the targeted site. Thus, complimentary combination of different agents that target multiple molecules in the angiogenic cascade may optimize inhibition of angiogenesis and improve clinical benefit in the cancer patients. This review provides an update on the current trend in exploitation of angiogenesis pathways as a strategy in the treatment of cancer.
Collapse
Affiliation(s)
- Ashwaq Hamid Salem Yehya
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Muhammad Asif
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Sven Hans Petersen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117543, Singapore.
| | - Ayappa V Subramaniam
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Koji Kono
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117543, Singapore.
- Department of Surgery, National University of Singapore, Singapore 117543, Singapore.
- School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Amin Malik Shah Abdul Majid
- EMAN Testing and Research Laboratories, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia.
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Acton 0200, Australia.
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia.
| |
Collapse
|
25
|
Mourah S, Porcher R, Lescaille G, Rousselot P, Podgorniak MP, Labarchède G, Naïmi B, Medioni J, Dombret H, Calvo F. Quantification of VEGF Isoforms and VEGFR Transcripts by qRT-PCR and Their Significance in Acute Myeloid Leukemia. Int J Biol Markers 2018; 24:22-31. [DOI: 10.1177/172460080902400104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Vascular endothelial growth factor (VEGF) and its receptors are known to play an important role in normal and pathological hematopoiesis but the prognostic impact of VEGF isoform transcripts in acute myeloid leukemia (AML) has not been addressed. We conducted a single-institution prospective study to analyze the impact of these angiogenic factors and the expression of their receptors on the survival of adult patients newly diagnosed with AML. We investigated the levels of VEGF transcript isoforms VEGF121, -145, -165, -189 and -206 and their receptors, VEGFR-1 and VEGFR-2, using quantitative reverse transcriptase polymerase chain reaction assays in peripheral blood mononuclear cells (PBMCs) of 67 consecutive AML patients at diagnosis. VEGF total protein was measured for comparison with mRNA levels in PBMCs. The VEGF121 splice variant transcript in AML PBMCs was significantly higher than in the normal controls. VEGF transcripts were quantified in all samples while its protein was detected in 42/67 (63%) of AML samples. High levels of VEGF121, VEGF165 transcripts and VEGF protein in AML were significantly related to a worse prognosis when analyzing overall survival (p<0.0001, p=0.019 and p=0.012, respectively) or event-free survival (p<0.0001, p=0.010 and p=0.047) using univariate analysis. In multivariable analysis only VEGF121 expression remained an independent prognostic factor for either event-free survival or overall survival [aHR=8.83 (3.48–22.4), p<0.0001, and aHR=9.52 (3.41–26.6), p<0.0001]. No prognostic value was observed for the other isoforms and the two receptors. Our findings show that the level of VEGF121 mRNA in circulating cells from AML patients is a strong independent prognostic parameter, which could be useful in the management of unselected AML patients.
Collapse
Affiliation(s)
- Samia Mourah
- INSERM U716, Laboratoire de Pharmacologie AP-HP, Hôpital Saint-Louis, Paris
| | | | | | - Philippe Rousselot
- Service Clinique des Maladies du Sang, AP-HP, Hôpital Saint-Louis, Paris
| | | | | | - Benyoussef Naïmi
- INSERM U716, Laboratoire de Pharmacologie AP-HP, Hôpital Saint-Louis, Paris
| | - Jacques Medioni
- Centre d'investigation clinique (CIC 9504), INSERM/AP-HP, Hôpital Saint-Louis, Paris - France
| | - Hervé Dombret
- Service Clinique des Maladies du Sang, AP-HP, Hôpital Saint-Louis, Paris
| | - Fabien Calvo
- INSERM U716, Laboratoire de Pharmacologie AP-HP, Hôpital Saint-Louis, Paris
- Centre d'investigation clinique (CIC 9504), INSERM/AP-HP, Hôpital Saint-Louis, Paris - France
| |
Collapse
|
26
|
Duarte D, Hawkins ED, Akinduro O, Ang H, De Filippo K, Kong IY, Haltalli M, Ruivo N, Straszkowski L, Vervoort SJ, McLean C, Weber TS, Khorshed R, Pirillo C, Wei A, Ramasamy SK, Kusumbe AP, Duffy K, Adams RH, Purton LE, Carlin LM, Lo Celso C. Inhibition of Endosteal Vascular Niche Remodeling Rescues Hematopoietic Stem Cell Loss in AML. Cell Stem Cell 2018; 22:64-77.e6. [PMID: 29276143 PMCID: PMC5766835 DOI: 10.1016/j.stem.2017.11.006] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 09/15/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
Bone marrow vascular niches sustain hematopoietic stem cells (HSCs) and are drastically remodeled in leukemia to support pathological functions. Acute myeloid leukemia (AML) cells produce angiogenic factors, which likely contribute to this remodeling, but anti-angiogenic therapies do not improve AML patient outcomes. Using intravital microscopy, we found that AML progression leads to differential remodeling of vasculature in central and endosteal bone marrow regions. Endosteal AML cells produce pro-inflammatory and anti-angiogenic cytokines and gradually degrade endosteal endothelium, stromal cells, and osteoblastic cells, whereas central marrow remains vascularized and splenic vascular niches expand. Remodeled endosteal regions have reduced capacity to support non-leukemic HSCs, correlating with loss of normal hematopoiesis. Preserving endosteal endothelium with the small molecule deferoxamine or a genetic approach rescues HSCs loss, promotes chemotherapeutic efficacy, and enhances survival. These findings suggest that preventing degradation of the endosteal vasculature may improve current paradigms for treating AML.
Collapse
Affiliation(s)
- Delfim Duarte
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK; The Francis Crick Institute, WC2A 3LY London, UK.
| | - Edwin D Hawkins
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK; The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Olufolake Akinduro
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Heather Ang
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Katia De Filippo
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, SW7 2AZ London, UK
| | - Isabella Y Kong
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Myriam Haltalli
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Nicola Ruivo
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Lenny Straszkowski
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Stephin J Vervoort
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3052, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Catriona McLean
- Department of Haematology, Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Tom S Weber
- Hamilton Institute, Maynooth University, Maynooth, Ireland
| | - Reema Khorshed
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Chiara Pirillo
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Andrew Wei
- Department of Haematology, Alfred Hospital, Melbourne, VIC 3004, Australia
| | | | - Anjali P Kusumbe
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7FY, UK
| | - Ken Duffy
- Hamilton Institute, Maynooth University, Maynooth, Ireland
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, 48149 Munster, Germany; University of Münster, Faculty of Medicine, 48149 Munster, Germany
| | - Louise E Purton
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia; Department of Medicine, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Leo M Carlin
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, SW7 2AZ London, UK; Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Cristina Lo Celso
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK; The Francis Crick Institute, WC2A 3LY London, UK.
| |
Collapse
|
27
|
Passaro D, Di Tullio A, Abarrategi A, Rouault-Pierre K, Foster K, Ariza-McNaughton L, Montaner B, Chakravarty P, Bhaw L, Diana G, Lassailly F, Gribben J, Bonnet D. Increased Vascular Permeability in the Bone Marrow Microenvironment Contributes to Disease Progression and Drug Response in Acute Myeloid Leukemia. Cancer Cell 2017; 32:324-341.e6. [PMID: 28870739 PMCID: PMC5598545 DOI: 10.1016/j.ccell.2017.08.001] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 04/25/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022]
Abstract
The biological and clinical behaviors of hematological malignancies can be influenced by the active crosstalk with an altered bone marrow (BM) microenvironment. In the present study, we provide a detailed picture of the BM vasculature in acute myeloid leukemia using intravital two-photon microscopy. We found several abnormalities in the vascular architecture and function in patient-derived xenografts (PDX), such as vascular leakiness and increased hypoxia. Transcriptomic analysis in endothelial cells identified nitric oxide (NO) as major mediator of this phenotype in PDX and in patient-derived biopsies. Moreover, induction chemotherapy failing to restore normal vasculature was associated with a poor prognosis. Inhibition of NO production reduced vascular permeability, preserved normal hematopoietic stem cell function, and improved treatment response in PDX.
Collapse
Affiliation(s)
- Diana Passaro
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Alessandro Di Tullio
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ander Abarrategi
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kevin Rouault-Pierre
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Katie Foster
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Linda Ariza-McNaughton
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Beatriz Montaner
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Probir Chakravarty
- Bioinformatic Core Unit, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Leena Bhaw
- Advanced Sequencing Unit, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Giovanni Diana
- MRC Centre for Developmental Neurobiology, King's College London, London, UK
| | - François Lassailly
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - John Gribben
- Department of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
28
|
JAK1/2 and BCL2 inhibitors synergize to counteract bone marrow stromal cell-induced protection of AML. Blood 2017; 130:789-802. [PMID: 28619982 DOI: 10.1182/blood-2016-02-699363] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/31/2017] [Indexed: 02/07/2023] Open
Abstract
The bone marrow (BM) provides a protective microenvironment to support the survival of leukemic cells and influence their response to therapeutic agents. In acute myeloid leukemia (AML), the high rate of relapse may in part be a result of the inability of current treatment to effectively overcome the protective influence of the BM niche. To better understand the effect of the BM microenvironment on drug responses in AML, we conducted a comprehensive evaluation of 304 inhibitors, including approved and investigational agents, comparing ex vivo responses of primary AML cells in BM stroma-derived and standard culture conditions. In the stroma-based conditions, the AML patient cells exhibited significantly reduced sensitivity to 12% of the tested compounds, including topoisomerase II, B-cell chronic lymphocytic leukemia/lymphoma 2 (BCL2), and many tyrosine kinase inhibitors (TKIs). The loss of TKI sensitivity was most pronounced in patient samples harboring FLT3 or PDGFRB alterations. In contrast, the stroma-derived conditions enhanced sensitivity to Janus kinase (JAK) inhibitors. Increased cell viability and resistance to specific drug classes in the BM stroma-derived conditions was a result of activation of alternative signaling pathways mediated by factors secreted by BM stromal cells and involved a switch from BCL2 to BCLXL-dependent cell survival. Moreover, the JAK1/2 inhibitor ruxolitinib restored sensitivity to the BCL2 inhibitor venetoclax in AML patient cells ex vivo in different model systems and in vivo in an AML xenograft mouse model. These findings highlight the potential of JAK inhibitors to counteract stroma-induced resistance to BCL2 inhibitors in AML.
Collapse
|
29
|
|
30
|
Abstract
Research in the last few years has revealed a sophisticated interaction network between multiple bone marrow cells that regulate different hematopoietic stem cell (HSC) properties such as proliferation, differentiation, localization, and self-renewal during homeostasis. These mechanisms are essential to keep the physiological HSC numbers in check and interfere with malignant progression. In addition to the identification of multiple mutations and chromosomal aberrations driving the progression of myeloid malignancies, alterations in the niche compartment recently gained attention for contributing to disease progression. Leukemic cells can remodel the niche into a permissive environment favoring leukemic stem cell expansion over normal HSC maintenance, and evidence is accumulating that certain niche alterations can even induce leukemic transformation. Relapse after chemotherapy is still a major challenge during treatment of myeloid malignancies, and cure is only rarely achieved. Recent progress in understanding the niche-imposed chemoresistance mechanisms will likely contribute to the improvement of current therapeutic strategies. This article discusses the role of different niche cells and their stage- and disease-specific roles during progression of myeloid malignancies and in response to chemotherapy.
Collapse
|
31
|
Chen LT, Chen CT, Jiaang WT, Chen TY, Butterfield JH, Shih NY, Hsu JTA, Lin HY, Lin SF, Tsai HJ. BPR1J373, an Oral Multiple Tyrosine Kinase Inhibitor, Targets c-KIT for the Treatment of c-KIT–Driven Myeloid Leukemia. Mol Cancer Ther 2016; 15:2323-2333. [DOI: 10.1158/1535-7163.mct-15-1006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 07/26/2016] [Indexed: 11/16/2022]
|
32
|
Cogle CR, Bosse RC, Brewer T, Migdady Y, Shirzad R, Kampen KR, Saki N. Acute myeloid leukemia in the vascular niche. Cancer Lett 2015; 380:552-560. [PMID: 25963886 DOI: 10.1016/j.canlet.2015.05.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 04/26/2015] [Accepted: 05/04/2015] [Indexed: 12/12/2022]
Abstract
The greatest challenge in treating acute myeloid leukemia (AML) is refractory disease. With approximately 60-80% of AML patients dying of relapsed disease, there is an urgent need to define and target mechanisms of drug resistance. Unfortunately, targeting cell-intrinsic resistance has failed to improve clinical outcomes in AML. Emerging data show that cell-extrinsic factors in the bone marrow microenvironment protect and support AML cells. The vascular niche, in particular, regulates AML cell survival and cell cycling by both paracrine secretion and adhesive contact with endothelial cells. Moreover, AML cells can functionally integrate within vascular endothelia, undergo quiescence, and resist cytotoxic chemotherapy. Together, these findings support the notion of blood vessels as sanctuary sites for AML. Therefore, vascular targeting agents may serve to remit AML. Several early phase clinical trials have tested anti-angiogenic agents, leukemia mobilizing agents, and vascular disrupting agents in AML patients. In general, these agents can be safely administered to AML patients and cardiovascular side effects were reported. Response rates to vascular targeting agents in AML have been modest; however, a majority of vascular targeting trials in AML are monotherapy in design and indiscriminate in patient recruitment. When considering the chemosensitizing effects of targeting the microenvironment, there is a strong rationale to build upon these early phase clinical trials and initiate phase IB/II trials of combination therapy where vascular targeting agents are positioned as priming agents for cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Raphael C Bosse
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Takae Brewer
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yazan Migdady
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Reza Shirzad
- Health research institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kim Rosalie Kampen
- Department of Pediatric Oncology/Hematology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Najmaldin Saki
- Health research institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
33
|
Göring S, Bensinger D, Naumann EC, Schmidt B. Computer-Guided Design, Synthesis, and Biological Evaluation of Quinoxalinebisarylureas as FLT3 Inhibitors. ChemMedChem 2015; 10:511-22. [DOI: 10.1002/cmdc.201402477] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/02/2014] [Indexed: 11/06/2022]
|
34
|
Kimmelman J, London AJ. The Structure of Clinical Translation:Efficiency, Information, and Ethics. Hastings Cent Rep 2015; 45:27-39. [DOI: 10.1002/hast.433] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Engen CBN, Wergeland L, Skavland J, Gjertsen BT. Targeted Therapy of FLT3 in Treatment of AML-Current Status and Future Directions. J Clin Med 2014; 3:1466-89. [PMID: 26237612 PMCID: PMC4470194 DOI: 10.3390/jcm3041466] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 11/27/2014] [Accepted: 11/28/2014] [Indexed: 12/18/2022] Open
Abstract
Internal tandem duplications (ITDs) of the gene encoding the Fms-Like Tyrosine kinase-3 (FLT3) receptor are present in approximately 25% of patients with acute myeloid leukemia (AML). The mutation is associated with poor prognosis, and the aberrant protein product has been hypothesized as an attractive therapeutic target. Various tyrosine kinase inhibitors (TKIs) have been developed targeting FLT3, but in spite of initial optimism the first generation TKIs tested in clinical studies generally induce only partial and transient hematological responses. The limited treatment efficacy generally observed may be explained by numerous factors; extensively pretreated and high risk cohorts, suboptimal pharmacodynamic and pharmacokinetic properties of the compounds, acquired TKI resistance, or the possible fact that inhibition of mutated FLT3 alone is not sufficient to avoid disease progression. The second-generation agent quizartinb is showing promising outcomes and seems better tolerated and with less toxic effects than traditional chemotherapeutic agents. Therefore, new generations of TKIs might be feasible for use in combination therapy or in a salvage setting in selected patients. Here, we sum up experiences so far, and we discuss the future outlook of targeting dysregulated FLT3 signaling in the treatment of AML.
Collapse
Affiliation(s)
| | - Line Wergeland
- Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen N-5020, Norway.
| | - Jørn Skavland
- Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen N-5020, Norway.
| | - Bjørn Tore Gjertsen
- Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen N-5020, Norway.
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen N-5021, Norway.
| |
Collapse
|
36
|
Yoon JH, Kim HJ, Kim JW, Jeon YW, Shin SH, Lee SE, Cho BS, Eom KS, Kim YJ, Lee S, Min CK, Cho SG, Lee JW, Min WS, Park CW. Identification of molecular and cytogenetic risk factors for unfavorable core-binding factor-positive adult AML with post-remission treatment outcome analysis including transplantation. Bone Marrow Transplant 2014; 49:1466-1474. [DOI: 10.1038/bmt.2014.180] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/03/2014] [Accepted: 06/08/2014] [Indexed: 12/20/2022]
|
37
|
Buchy E, Valetti S, Mura S, Mougin J, Troufflard C, Couvreur P, Desmaële D. Synthesis and Cytotoxic Activity of Self-Assembling Squalene Conjugates of 3-[(Pyrrol-2-yl)methylidene]-2,3-dihydro-1H-indol-2-one Anticancer Agents. European J Org Chem 2014. [DOI: 10.1002/ejoc.201403088] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
Lee JY, Kim HJ. (Lymph)angiogenic influences on hematopoietic cells in acute myeloid leukemia. Exp Mol Med 2014; 46:e122. [PMID: 25412683 PMCID: PMC4262793 DOI: 10.1038/emm.2014.72] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/26/2014] [Accepted: 09/21/2014] [Indexed: 02/07/2023] Open
Abstract
The purpose of this review is to provide an overview of the effect of (lymph)angiogenic cytokines on hematopoietic cells involved in acute myeloid leukemia (AML). Like angiogenesis, lymphangiogenesis occurs in pathophysiological conditions but not in healthy adults. AML is closely associated with the vasculature system, and the interplay between lymphangiogenic cytokines maintains leukemic blast survival in the bone marrow (BM). Once AML is induced, proangiogenic cytokines function as angiogenic or lymphangiogenic factors and affect hematopoietic cells, including BM-derived immune cells. Simultaneously, the representative cytokines, VEGFs and their receptors are expressed on AML blasts in vascular and osteoblast niches in both the BM and the peripheral circulation. After exposure to (lymph)angiogenic cytokines in leukemogenesis and infiltration, immune cell phenotypes and functions are affected. These dynamic behaviors in the BM reflect the clinical features of AML. In this review, we note the importance of lymphangiogenic factors and their receptors in hematopoietic cells in AML. Understanding the functional characterization of (lymph)angiogenic factors in the BM niche in AML will also be helpful in interrupting the engraftment of leukemic stem cells and for enhancing immune cell function by modulating the tumor microenvironment.
Collapse
Affiliation(s)
- Ji Yoon Lee
- Cancer Research Institute, Department of Hematology, Catholic Blood and Marrow Transplantation Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Cancer Research Institute, Department of Hematology, Catholic Blood and Marrow Transplantation Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
39
|
Mattison R, Jumonville A, Flynn PJ, Moreno-Aspitia A, Erlichman C, LaPlant B, Juckett MB. A phase II study of AZD2171 (cediranib) in the treatment of patients with acute myeloid leukemia or high-risk myelodysplastic syndrome. Leuk Lymphoma 2014; 56:2061-6. [PMID: 25329007 DOI: 10.3109/10428194.2014.977886] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) not fit for intensive treatment need novel therapy options. Vascular endothelial growth factor (VEGF) receptor inhibition is one potential mechanism by which AML and MDS could be treated. The receptor tyrosine kinase inhibitor AZD2171 (cediranib) has activity against VEGF receptors KDR and FLT-1. This multicenter phase II study was designed to test cediranib's activity in patients with AML or high-risk MDS. The primary endpoint was confirmed disease response defined as a composite of complete remission, partial remission or hematologic improvement. The study enrolled 23 subjects in the AML cohort and 16 subjects in the MDS cohort. There were no confirmed responses in either group. Since the study met the stopping rule after the first stage of enrollment, the trial was closed to further accrual. Common adverse events in both cohorts included thrombocytopenia, neutropenia, anemia, fatigue, dyspnea, diarrhea, nausea and dehydration.
Collapse
Affiliation(s)
- Ryan Mattison
- Carbone Comprehensive Cancer Center, University of Wisconsin , Madison, WI , USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Crucke J, Huysseune A. Blocking VEGF signaling delays development of replacement teeth in zebrafish. J Dent Res 2014; 94:157-65. [PMID: 25391620 DOI: 10.1177/0022034514557156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The dentition in zebrafish is extremely and richly vascularized, but the function of the vasculature, in view of the continuous replacement of the teeth, remains elusive. Through application of SU5416, a vascular endothelial growth factor receptor inhibitor, we studied the role of the blood vessels in the dentition of the zebrafish. We were unable to show an effect on the development of first-generation teeth as well as first tooth replacement. However, in juvenile fish, a delay was observed in the developmental state of the replacement tooth compared with what was expected based on the maturation state of the functional tooth. Furthermore, we observed a difference between treated and nontreated fish in the distance between blood vessels and developing replacement teeth. In conclusion, our results provide support for a nutritive, rather than an inductive, function of the vasculature in the process of tooth development and replacement.
Collapse
Affiliation(s)
- J Crucke
- Evolutionary Developmental Biology, Ghent University, Belgium
| | - A Huysseune
- Evolutionary Developmental Biology, Ghent University, Belgium
| |
Collapse
|
41
|
Onishi C, Mori-Kimachi S, Hirade T, Abe M, Taketani T, Suzumiya J, Sugimoto T, Yamaguchi S, Kapur R, Fukuda S. Internal tandem duplication mutations in FLT3 gene augment chemotaxis to Cxcl12 protein by blocking the down-regulation of the Rho-associated kinase via the Cxcl12/Cxcr4 signaling axis. J Biol Chem 2014; 289:31053-31065. [PMID: 25237195 PMCID: PMC4223310 DOI: 10.1074/jbc.m114.568287] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 09/12/2014] [Indexed: 11/06/2022] Open
Abstract
Internal tandem duplication mutations in the Flt3 gene (ITD-FLT3) enhance cell migration toward the chemokine Cxcl12, which is highly expressed in the therapy-protective bone marrow niche, providing a potential mechanism underlying the poor prognosis of ITD-FLT3(+) acute myeloid leukemia. We aimed to investigate the mechanisms linking ITD-FLT3 to increased cell migration toward Cxcl12. Classification of the expression of Cxcl12-regulated genes in ITD-FLT3(+) cells demonstrated that the enhanced migration of ITD-FLT3(+) cells toward Cxcl12 was associated with the differential expression of genes downstream of Cxcl12/Cxcr4, which are functionally distinct from those expressed in ITD-FLT3(-) cells but are independent of the Cxcr4 expression levels. Among these differentially regulated genes, the expression of Rock1 in the ITD-FLT3(+) cells that migrated toward Cxcl12 was significantly higher than in ITD-FLT3(-) cells that migrated toward Cxcl12. In ITD-FLT3(-) cells, Rock1 expression and Mypt1 phosphorylation were transiently up-regulated but were subsequently down-regulated by Cxcl12. In contrast, the presence of ITD-FLT3 blocked the Cxcl12-induced down-regulation of Rock1 and early Mypt1 dephosphorylation. Likewise, the FLT3 ligand counteracted the Cxcl12-induced down-regulation of Rock1 in ITD-FLT3(-) cells, which coincided with enhanced cell migration toward Cxcl12. Rock1 antagonists or Rock1 shRNA abolished the enhanced migration of ITD-FLT3(+) cells toward Cxcl12. Our findings demonstrate that ITD-FLT3 increases cell migration toward Cxcl12 by antagonizing the down-regulation of Rock1 expression. These findings suggest that the aberrant modulation of Rock1 expression and activity induced by ITD-FLT3 may enhance acute myeloid leukemia cell chemotaxis to the therapy-protective bone marrow niche, where Cxcl12 is abundantly expressed.
Collapse
Affiliation(s)
- Chie Onishi
- From the Departments of Oncology/Hematology and
| | | | | | | | - Takeshi Taketani
- Pediatrics and the Division of Blood Transfusion, Shimane University Hospital, 693-8501 Izumo, Japan, and
| | | | - Toshitsugu Sugimoto
- First Department of Internal Medicine, Shimane University School of Medicine, Izumo 693-8501, Japan
| | | | - Reuben Kapur
- the Department of Pediatrics and the Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | | |
Collapse
|
42
|
Al-Hussaini M, DiPersio JF. Small molecule inhibitors in acute myeloid leukemia: from the bench to the clinic. Expert Rev Hematol 2014; 7:439-64. [PMID: 25025370 PMCID: PMC4283573 DOI: 10.1586/17474086.2014.932687] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many patients with acute myeloid leukemia will eventually develop refractory or relapsed disease. In the absence of standard therapy for this population, there is currently an urgent unmet need for novel therapeutic agents. Targeted therapy with small molecule inhibitors represents a new therapeutic intervention that has been successful for the treatment of multiple tumors (e.g., gastrointestinal stromal tumors, chronic myelogenous leukemia). Hence, there has been great interest in generating selective small molecule inhibitors targeting critical pathways of proliferation and survival in acute myeloid leukemia. This review highlights a selective group of intriguing therapeutic agents and their presumed targets in both preclinical models and in early human clinical trials.
Collapse
Affiliation(s)
- Muneera Al-Hussaini
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis Missouri
| | - John F. DiPersio
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis Missouri
- Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St Louis Missouri
| |
Collapse
|
43
|
Chung HJ, Kamli MR, Lee HJ, Ha JD, Cho SY, Lee J, Kong JY, Han SY. Discovery of quinolinone derivatives as potent FLT3 inhibitors. Biochem Biophys Res Commun 2014; 445:561-5. [PMID: 24530392 DOI: 10.1016/j.bbrc.2014.02.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/06/2014] [Indexed: 11/30/2022]
Abstract
Recently some fms-like tyrosine kinase 3 (FLT3) inhibitors have shown good efficacy in acute myeloid leukemia (AML) patients. In an effort to develop anti-leukemic drugs, we investigated quinolinone derivatives as novel FLT3 inhibitors. Two substituted quinolinones, KR65367 and KR65370 were subjected to FLT3 kinase activity assay and showed potent inhibition against FLT3 kinase activity in vitro, with IC50 of 2.7 and 0.57 nM, respectively. As a measure of selectivity, effects on the activity of other kinases were also tested. Both compounds have negligible activity against Met, Ron, epidermal growth factor receptor, Aurora A, Janus kinase 2, and insulin receptor; with IC50 greater than 10 μM. KR compounds showed strong growth inhibition in MV4;11 AML cells and increased the apoptotic cell death in flow cytometric analyses. A decrease in STAT5 phosphorylation by KR compounds was observed in MV4;11 cells. Furthermore, in vitro evaluation of compounds structurally related to KR65367 and KR65370 showed a good structure-activity relationship.
Collapse
Affiliation(s)
- Hye Jin Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Majid Rasool Kamli
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Hyo Jeong Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Jae Du Ha
- Bio-organic Science Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sung Yun Cho
- Bio-organic Science Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jongkook Lee
- College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Jae Yang Kong
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Sun-Young Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea.
| |
Collapse
|
44
|
Piazza F, Semenzato G. Molecular therapeutic approaches to acute myeloid leukemia: targeting aberrant chromatin dynamics and signal transduction. Expert Rev Anticancer Ther 2014; 4:387-400. [PMID: 15161438 DOI: 10.1586/14737140.4.3.387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute myeloid leukemia research and clinical management have greatly benefited from the achievements in molecular biology regarding the identification of the underlying pathogenetic mechanisms of transformation and resistance to therapy. In particular, two categories of alterations, the aberrant activity of transcription/chromatin-remodeling factors and the deregulated activation of signal transduction pathways, have been demonstrated to play a pivotal role in leukemic cell differentiation, proliferation and resistance to apoptosis. These molecular lesions have proven to be suitable therapeutic targets in acute promyelocytic leukemia and chronic myeloid leukemia and are now also seen as therapeutic targets for a wider group of leukemic disorders. The development of novel drugs such as histone deacetylase inhibitors, demethylating agents and inhibitors of receptor tyrosine kinases may potentially benefit acute myeloid leukemia patients.
Collapse
Affiliation(s)
- Francesco Piazza
- Padova University School of Medicine, Venetian Institute of Molecular Medicine, Unit of Hematological Malignancies, via Orus 2 35129 Padova, Italy.
| | | |
Collapse
|
45
|
Abstract
Since the initial hypotheses on the importance of angiogenesis in the pathogenesis of cancer approximately 30 years ago, there have been major advances in the understanding of the cellular and molecular mechanisms involved in the regulation of this complex process of new vessel formation. Among the multitude of factors, vascular endothelial growth factor (VEGF) has emerged as one of the most potent angiogenic factors, being implicated in the initiation of signal transduction responsible for cell proliferation, survival, migration and adhesion. Inhibition of VEGF and its signaling pathway offers a potential new molecular target in cancer therapy. This article reviews the role of angiogenesis and its mediators, particularly vascular endothelial growth factors, in hematological malignancies, as well as the potential use of anti-angiogenic therapies in the management of these conditions.
Collapse
Affiliation(s)
- Soon Thye Lim
- Keck School of Medicine/Norris, university of Southern california, Comprehensive Cancer center, Los Angeles, CA 90033, USA
| | | |
Collapse
|
46
|
Serve H, Krug U, Wagner R, Sauerland MC, Heinecke A, Brunnberg U, Schaich M, Ottmann O, Duyster J, Wandt H, Fischer T, Giagounidis A, Neubauer A, Reichle A, Aulitzky W, Noppeney R, Blau I, Kunzmann V, Stuhlmann R, Krämer A, Kreuzer KA, Brandts C, Steffen B, Thiede C, Müller-Tidow C, Ehninger G, Berdel WE. Sorafenib in Combination With Intensive Chemotherapy in Elderly Patients With Acute Myeloid Leukemia: Results From a Randomized, Placebo-Controlled Trial. J Clin Oncol 2013; 31:3110-8. [DOI: 10.1200/jco.2012.46.4990] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Purpose The prognosis of elderly patients with acute myeloid leukemia (AML) is still dismal even with intensive chemotherapy. In this trial, we compared the antileukemic activity of standard induction and consolidation therapy with or without the addition of the kinase inhibitor sorafenib in elderly patients with AML. Patients and Methods All patients received standard cytarabine and daunorubicin induction (7+3 regimen) and up to two cycles of intermediate-dose cytarabine consolidation. Two hundred one patients were equally randomly assigned to receive either sorafenib or placebo between the chemotherapy cycles and subsequently for up to 1 year after the beginning of therapy. The primary objective was to test for an improvement in event-free survival (EFS). Overall survival (OS), complete remission (CR) rate, tolerability, and several predefined subgroup analyses were among the secondary objectives. Results Age, sex, CR and early death (ED) probability, and prognostic factors were balanced between both study arms. Treatment in the sorafenib arm did not result in significant improvement in EFS or OS. This was also true for subgroup analyses, including the subgroup positive for FLT3 internal tandem duplications. Results of induction therapy were worse in the sorafenib arm, with higher treatment-related mortality and lower CR rates. More adverse effects occurred during induction therapy in the sorafenib arm, and patients in this arm received less consolidation chemotherapy as a result of higher induction toxicity. Conclusion In conclusion, combination of standard induction and consolidation therapy with sorafenib in the schedule investigated in our trial is not beneficial for elderly patients with AML.
Collapse
Affiliation(s)
- Hubert Serve
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Utz Krug
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Ruth Wagner
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - M. Cristina Sauerland
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Achim Heinecke
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Uta Brunnberg
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Markus Schaich
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Oliver Ottmann
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Justus Duyster
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Hannes Wandt
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Thomas Fischer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Aristoteles Giagounidis
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Andreas Neubauer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Albrecht Reichle
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Walter Aulitzky
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Richard Noppeney
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Igor Blau
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Volker Kunzmann
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Reingard Stuhlmann
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Alwin Krämer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Karl-Anton Kreuzer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Christian Brandts
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Björn Steffen
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Christian Thiede
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Carsten Müller-Tidow
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Gerhard Ehninger
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Wolfgang E. Berdel
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| |
Collapse
|
47
|
Peloquin GL, Chen YB, Fathi AT. The evolving landscape in the therapy of acute myeloid leukemia. Protein Cell 2013; 4:735-46. [PMID: 23982740 DOI: 10.1007/s13238-013-3057-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 07/25/2013] [Indexed: 11/26/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disorder of myeloid precursors arrested in their maturation, creating a diverse disease entity with a wide range of responses to historically standard treatment approaches. While significant progress has been made in characterizing and individualizing the disease at diagnosis to optimally inform those affected, progress in treatment to reduce relapse and induce remission has been limited thus far. In addition to a brief summary of the factors that shape prognostication at diagnosis, this review attempts to expand on the current therapies under investigation that have shown promise in treating AML, including hypomethylating agents, gemtuzumab ozogamicin, FLT3 tyrosine kinase inhibitors, antisense oligonucleotides, and other novel therapies, including aurora kinases, mTOR and PI3 kinase inhibitors, PIM kinase inhibitors, HDAC inhibitors, and IDH targeted therapies. With these, and undoubtedly many others in the future, it is the hope that by combining more accurate prognostication with more effective therapies, patients will begin to have a different, and more complete, outlook on their disease that allows for safer and more successful treatment strategies.
Collapse
Affiliation(s)
- Grace L Peloquin
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | | | | |
Collapse
|
48
|
Ribatti D. Angiogenesis as a treatment target in leukemia. Int J Hematol Oncol 2013. [DOI: 10.2217/ijh.13.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY The importance of angiogenesis in the growth and survival of leukemia has been well established and confirmed by several studies. In the last 20 years, several antiangiogenic agents have been used in preclinical and clinical studies of the treatment of leukemia. This review article summarizes the literature focusing on the relationship between angiogenesis and disease progression, and the advantages and limits of the antiangiogenic treatment of leukemia.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neuroscience, & Sensory Organs, University of Bari Medical School, Piazza Giulio Cesare, 11, 70124 Bari, Italy
| |
Collapse
|
49
|
Fetterly GJ, Aras U, Lal D, Murphy M, Meholick PD, Wang ES. Development of a preclinical PK/PD model to assess antitumor response of a sequential aflibercept and doxorubicin-dosing strategy in acute myeloid leukemia. AAPS JOURNAL 2013; 15:662-73. [PMID: 23550025 DOI: 10.1208/s12248-013-9480-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 03/21/2013] [Indexed: 12/27/2022]
Abstract
Timing of the anti-angiogenic agent with respect to the chemotherapeutic agent may be crucial in determining the success of combination therapy in cancer. We investigated the effects of sequential therapy with the potent VEGF inhibitor, aflibercept, and doxorubicin (DOX) in preclinical acute myeloid leukemia (AML) models. Mice were engrafted with human HL-60 and HEL-luciferase leukemia cells via S.C. and/or I.V. injection and treated with two to three doses of aflibercept (5-25 mg/kg) up to 3-7 days prior to doxorubicin (30 mg/kg) administration. Leukemia growth was determined by local tumor measurements (days 0-16) and systemic bioluminescent imaging (days 0-28) in animals receiving DOX (3 mg/kg) with or without aflibercept. A PK/PD model was developed to characterize how prior administration of aflibercept altered intratumoral DOX uptake. DOX concentration-time profiles were described using a four-compartment PK model with linear elimination. We determined that intratumoral DOX concentrations were 6-fold higher in the aflibercept plus DOX treatment group versus DOX alone in association with increased drug uptake rates (from 0.125 to 0.471 ml/h/kg) into tumor without affecting drug efflux. PD modeling demonstrated that the observed growth retardation was mainly due to the combination of DOX plus TRAP group; 0.00794 vs. 0.0043 h(-1). This PK/PD modeling approach in leukemia enabled us to predict the effects of dosing frequency and sequence for the combination of anti-VEGF and cytotoxic agents on AML growth in both xenograft and marrow, and may be useful in the design of future rational combinatorial dosing regimens in hematological malignancies.
Collapse
Affiliation(s)
- Gerald J Fetterly
- PK/PD Core Facility, Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Kampen KR, Ter Elst A, de Bont ESJM. Vascular endothelial growth factor signaling in acute myeloid leukemia. Cell Mol Life Sci 2013; 70:1307-17. [PMID: 22833169 PMCID: PMC11113417 DOI: 10.1007/s00018-012-1085-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 06/19/2012] [Accepted: 07/09/2012] [Indexed: 01/08/2023]
Abstract
This review is designed to provide an overview of the current literature concerning vascular endothelial growth factor signaling (VEGF) in acute myeloid leukemia (AML). Aberrant VEGF signaling operates in the bone marrow of AML patients and is related to a poor prognosis. The altered signaling pathway demonstrated to interfere in several autocrine and paracrine signaling pathways. VEGF signaling promotes autocrine AML blast cell proliferation, survival, and chemotherapy resistance. In addition, VEGF signaling can mediate paracrine vascular endothelial cell-controlled angiogenesis in AML. Both effects presumably explain the association of high VEGF levels and poor therapeutic outcome. More recently, researches focusing on bone marrow stem cell niches demonstrate a role for VEGF signaling in the preservation of several cell types within these niches. The bone marrow niches are proposed to be a protective microenvironment for AML cells that could be responsible for relapses in AML patients. This implies the need of sophisticated VEGF-targeted therapeutics in AML therapy strategies. This review highlights our current understanding of aberrant VEGF signaling in AML, appoints the interference of VEGF signaling in the AML-associated microenvironment, and reflects the novelty of current VEGF-targeted therapeutics used in clinical trails for the treatment of AML.
Collapse
Affiliation(s)
- Kim R Kampen
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands.
| | | | | |
Collapse
|