1
|
Synergistic Lethality Effects of Apatinib and Homoharringtonine in Acute Myeloid Leukemia. JOURNAL OF ONCOLOGY 2022; 2022:9005804. [PMID: 36081666 PMCID: PMC9448536 DOI: 10.1155/2022/9005804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022]
Abstract
Purpose The significance of vascular endothelial growth factor receptor (VEGFR)-2 in numerous solid tumors and acute myeloid leukemia (AML) has been demonstrated, but Apatinib remains largely unexplored. In this study, whether Apatinib combined with homoharringtonine (HHT) kills AML cell lines and its possible mechanisms have been explored. Methods AML cell lines were treated with Apatinib and HHT in different concentrations with control, Apatinib alone, HHT alone, and Apatinib combined with HHT. The changes of IC50 were measured by CCK8 assay, and apoptosis rate, cell cycle, and the mitochondrial membrane potential in each group were measured by flow cytometry. Finally, the possible cytotoxicity mechanism was analyzed by Western blotting. Results Our results noted that Apatinib combined with HHT remarkably inhibited cell proliferation, reduced the capacity of colony-forming, and induced apoptosis and cell cycle arrest in AML cells. Mechanistically, Apatinib and HHT play a role as a suppressor in the expression of VEGFR-2 and the downstream signaling cascades, such as the PI3K, MAPK, and STAT3 pathways. Conclusion Our preclinical data demonstrate that Apatinib combined with HHT exerts a better antileukemia effect than Apatinib alone by inhibiting the VEGFR-2 signaling pathway, suggesting the potential role of Apatinib and HHT in the treatment of AML. This study provides clinicians with innovative combination therapies and new therapeutic targets for the treatment of AML.
Collapse
|
2
|
Reikvam H, Hatfield KJ, Wendelbo Ø, Lindås R, Lassalle P, Bruserud Ø. Endocan in Acute Leukemia: Current Knowledge and Future Perspectives. Biomolecules 2022; 12:biom12040492. [PMID: 35454082 PMCID: PMC9027427 DOI: 10.3390/biom12040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Endocan is a soluble dermatan sulfate proteoglycan expressed by endothelial cells and detected in serum/plasma. Its expression is increased in tumors/tumor vessels in several human malignancies, and high expression (high serum/plasma levels or tumor levels) has an adverse prognostic impact in several malignancies. The p14 endocan degradation product can also be detected in serum/plasma, but previous clinical studies as well as previously unpublished results presented in this review suggest that endocan and p14 endocan fragment levels reflect different biological characteristics, and the endocan levels seem to reflect the disease heterogeneity in acute leukemia better than the p14 fragment levels. Furthermore, decreased systemic endocan levels in previously immunocompetent sepsis patients are associated with later severe respiratory complications, but it is not known whether this is true also for immunocompromised acute leukemia patients. Finally, endocan is associated with increased early nonrelapse mortality in (acute leukemia) patients receiving allogeneic stem cell transplantation, and this adverse prognostic impact seems to be independent of the adverse impact of excessive fluid overload. Systemic endocan levels may also become important to predict cytokine release syndrome after immunotherapy/haploidentical transplantation, and in the long-term follow-up of acute leukemia survivors with regard to cardiovascular risk. Therapeutic targeting of endocan is now possible, and the possible role of endocan in acute leukemia should be further investigated to clarify whether the therapeutic strategy should also be considered.
Collapse
Affiliation(s)
- Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Kimberley Joanne Hatfield
- Department of Transfusion Medicine and Immunology, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Øystein Wendelbo
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Roald Lindås
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Philippe Lassalle
- Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR9017, University of Lille, 59000 Lille, France;
- Center for Infection and Immunity, le Centre Nationale de la Recherche Scientifique, Univeristy of Lille, 59000 Lille, France
- Centre d’Infection et d’Immunité de Lille, Equipe Immunité Pulmonaire, University of Lille, 59000 Lille, France
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
- Correspondence:
| |
Collapse
|
3
|
Luo D, Liu D, Rao C, Shi S, Zeng X, Liu S, Jiang H, Liu L, Zhang Z, Lu X. Raised SPINK1 levels play a role in angiogenesis and the transendothelial migration of ALL cells. Sci Rep 2022; 12:2999. [PMID: 35194087 PMCID: PMC8864021 DOI: 10.1038/s41598-022-06946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 02/09/2022] [Indexed: 11/23/2022] Open
Abstract
The present study was designed to assess whether raised Serine protease inhibitor Kazal type 1 (SPINK1) expressions modulates angiogenesis. Human umbilical vein endothelial cells (HUVECs) exposed to SPINK1 were noted to exhibit raised expressions of interleukin-8 (IL-8) as well as VCAM-1 and ICAM-1 cell adhesion molecules in a dose-dependent manner. In co-culture system of HUVECs and Acute lymphoblastic leukemia (ALL) cells, SPINK1 exposure also resulted in enhanced endothelial cell motility and ALL cells trans-endothelial migration. High concentrations of SPINK1 caused in vitro cellular reorganization into tubes in Matrigel-cultured HUVECs and induced in vivo vascularization and brain infiltration of NOD/SCID ALL model mice. The further transcriptomic analysis indicated that SPINK1 treatment altered several biological processes of endothelial cells and led to activation of the MAPK pathway. This study is the first to determine the neovascularization effects of raised SPINK1.
Collapse
Affiliation(s)
- Dong Luo
- Medical Laboratory, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan City, Guangdong Province, China.,Department of Medical and Molecular Genetics, Dongguan Institute of Pediatrics, Dongguan, Guangdong, China
| | - Dongqiang Liu
- Department of Hematology, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Chunbao Rao
- Medical Laboratory, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan City, Guangdong Province, China.,Department of Medical and Molecular Genetics, Dongguan Institute of Pediatrics, Dongguan, Guangdong, China
| | - Shanshan Shi
- Department of Neurology, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, Guangdong, China
| | - Xiaomei Zeng
- Medical Laboratory, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan City, Guangdong Province, China.,Department of Medical and Molecular Genetics, Dongguan Institute of Pediatrics, Dongguan, Guangdong, China
| | - Sha Liu
- Department of Hematology-Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Hua Jiang
- Department of Hematology-Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Lishi Liu
- Department of Hematology, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Zhenhong Zhang
- Department of Hematology, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan City, Guangdong Province, China.
| | - Xiaomei Lu
- Medical Laboratory, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan City, Guangdong Province, China. .,Department of Medical and Molecular Genetics, Dongguan Institute of Pediatrics, Dongguan, Guangdong, China.
| |
Collapse
|
4
|
Deng M, Zha J, Jiang Z, Jia X, Shi Y, Li P, Chen XL, Fang Z, Du Z, Xu B. Apatinib exhibits anti-leukemia activity in preclinical models of acute lymphoblastic leukemia. J Transl Med 2018; 16:47. [PMID: 29490645 PMCID: PMC5831852 DOI: 10.1186/s12967-018-1421-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/20/2018] [Indexed: 12/11/2022] Open
Abstract
Background Acute lymphoblastic leukemia (ALL) is a clonal malignant disorder characterized by an uncontrolled proliferation of immature B or T lymphocytes. Extensive studies have suggested an involvement of angiogenesis signaling in ALL progression and resistance to treatment. Thus, targeting angiogenesis with anti-angiogenic drugs may be a promising approach for ALL treatment. In this study, we investigated the effectiveness of Apatinib, a novel receptor tyrosine kinase inhibitor selectively targeting VEGFR-2 in ALL cells. Method ALL cell lines were treated with different concentration of Apatinib and then CCK8 assay, flow cytometry were used to determine the IC50 value and cell apoptosis, respectively. The effect of Apatinib against primary ALL cells from 11 adult patients and normal counterparts were also analyzed by apoptosis with flow cytometry. Next, we used western bolting and mass cytometry (CyTOF) assay to explore the underlying mechanism of the cytotoxicity of Apatinib. Finally, the anti-leukemia activity was further evaluated in an in vivo xenograft model of ALL. Results Our results showed that Apatinib significantly inhibited cell growth and promoted apoptosis in both B and T lineage ALL cell lines in a dose- and time-dependent manner. The IC50 values of Apatinib against Nalm6, Reh, Jurkat and Molt4 for 48 h were 55.76 ± 13.19, 51.53 ± 10.74, 32.43 ± 5.58, 39.91 ± 9.88 μmol/L, and for 72 h were 30.34 ± 2.65, 31.96 ± 3.92, 17.62 ± 5.90, and 17.65 ± 2.17 μmol/L respectively. Similarly, Apatinib shows cytotoxic activity against primary adult ALL cells while sparing their normal counterparts in vitro. Moreover, Apatinib suppressed ALL growth and progression in an in vivo xenograft model. Mechanistically, Apatinib-induced cytotoxicity was closely associated with inhibition of VEGFR2 and its downstream signaling cascades, including the PI3 K, MAPK and STAT3 pathways. Conclusion Our study indicates that Apatinib exerts its anti-leukemia effect by inducing apoptosis through suppressing the VEGFR2 signaling pathway, supporting a potential role for Apatinib in the treatment of ALL. Electronic supplementary material The online version of this article (10.1186/s12967-018-1421-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manman Deng
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Medical College of Xiamen University, Xiamen, 361003, People's Republic of China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Medical College of Xiamen University, Xiamen, 361003, People's Republic of China
| | - Zhiwu Jiang
- Key Laboratory of Regenerative Biology, Southern China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xian Jia
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signalling 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, Medical College of Xiamen University, Xiamen, 361003, People's Republic of China
| | - Peng Li
- Key Laboratory of Regenerative Biology, Southern China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiao Lei Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signalling Network, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Zhihong Fang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Medical College of Xiamen University, Xiamen, 361003, People's Republic of China.
| | - Zhiqiang Du
- Department of Translational Science, Amgen Asia R&D Center, Shanghai, China.
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Medical College of Xiamen University, Xiamen, 361003, People's Republic of China.
| |
Collapse
|
5
|
Cheng Z, Fu J, Liu G, Zhang L, Xu Q, Wang SY. Angiogenesis in JAK2 V617F positive myeloproliferative neoplasms and ruxolitinib decrease VEGF, HIF-1 enesis in JAK2 V617F positive cells. Leuk Lymphoma 2017; 59:196-203. [PMID: 28554272 DOI: 10.1080/10428194.2017.1324155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Angiogenesis and JAK2 V617F mutation are common in BCR-ABL1 negative myeloproliferative neoplasms (MPNs). Ruxolitinib, a JAK inhibitor, is an effective treatment for some MPNs. However, the relationship between angiogenesis and JAK2 V617F and the effects of ruxolitinib on angiogenesis are still unknown. Here, we observed the correlation of JAK2 V617F mutation burden with VEGF, HIF-1a and microvascular density (MVD) in MPNs. We investigate the effect of ruxolitinib on the expression of VEGF and HIF-1α in JAK2 V617F positive cells. We found the expression levels of p-JAK2, VEGF, HIF-1a and MVD in the newly diagnosed MPNs were significantly increased and were related to the JAK2 V617F burden. Ruxolitinib can inhibit p-JAK2, VEGF, HIF-1a expression and suppress blood vessels' formation in chick embryo choriallantoic membrane. Our findings indicated that angiogenesis is related to JAK2 V617F burden and ruxolitinib could decrease VEGF and HIF-1a expression in JAK2 V617F positive cells.
Collapse
Affiliation(s)
- Zhiyong Cheng
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Jianzhu Fu
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Guimin Liu
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Lijun Zhang
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Qian Xu
- a Department of Hematology , The No. 1 Hospital of Baoding , Baoding , China
| | - Su-Yun Wang
- b Department of Hematology , Hebei General Hospital , Shijiazhuang , Hebei Province , China
| |
Collapse
|
6
|
Song Y, Tan Y, Liu L, Wang Q, Zhu J, Liu M. Levels of bone marrow microvessel density are crucial for evaluating the status of acute myeloid leukemia. Oncol Lett 2015; 10:211-215. [PMID: 26171001 DOI: 10.3892/ol.2015.3209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 04/14/2015] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to determine the efficacy of bone marrow microvessel density (BM-MVD) in the evaluation of the status of acute myeloid leukemia (AML). The levels of serum and bone marrow vascular endothelial growth factor (VEGF), and BM-MVD in 28 patients with AML, 10 patients with non-Hodgkin's lymphoma, 10 patients with anemia and 14 patients with AML that achieved complete remission for six months (AML-DFS) subsequent to chemotherapy were determined by ELISA and immunohistochemistry. The levels of serum VEGF in patients with hematological disorders were significantly increased compared with the levels in the healthy controls. The levels of BM VEGF and BM-MVD in AML patients were significantly higher compared with the levels in the patients with non-Hodgkin's lymphoma or anemia. Following chemotherapy, the levels of serum VEGF significantly increased and the levels of BM VEGF decreased in the AML patients, regardless of their therapeutic responses, compared with the levels prior to treatment. By contrast, the levels of BM-MVD in the AML patients were significantly reduced in the patients that completely recovered from AML (AML-DFS group), compared with those in other groups. The present data indicate that the levels of BM-MVD are valuable for evaluating the status of AML.
Collapse
Affiliation(s)
- Yanqiu Song
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Tan
- Department of Oncology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Libo Liu
- Department of Hematology, The Fourth Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qian Wang
- Department of Hematology, The Fourth Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Zhu
- Department of Oncology, Jilin Cancer Hospital, Changchun, Jilin 130021, P.R. China
| | - Min Liu
- Department of Radiation Oncology, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
7
|
Ponce CC, de Lourdes Lopes Ferrari Chauffaille M, Ihara SSM, Silva MRR. Increased angiogenesis in primary myelofibrosis: latent transforming growth factor-β as a possible angiogenic factor. Rev Bras Hematol Hemoter 2014; 36:322-8. [PMID: 25305163 PMCID: PMC4318371 DOI: 10.1016/j.bjhh.2014.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/31/2014] [Indexed: 12/21/2022] Open
Abstract
Objective The aim of this work was to demonstrate a possible relationship between anti-latency-associated peptide human latent transforming growth factor beta 1 (latent TGF-β1) expression in megakaryocytes and microvascular density in bone marrow biopsies from patients with essential thrombocythemia and primary myelofibrosis. Methods Microvascular density was evaluated by immunohistochemical analysis and the expression of latent TGF-β1 in samples (100 megakaryocytes per bone marrow sample) from 18 essential thrombocythemia and 38 primary myelofibrosis (19 prefibrotic and 19 fibrotic) patients. Six bone marrow donor biopsies were used as controls. Fibrosis in the bone marrow biopsies was evaluated according to the European Consensus. Results The average fibrosis grade differed between essential thrombocythemia and primary myelofibrosis groups when compared to the control group. Latent TGF-β1 expression differed significantly between the fibrotic primary myelofibrosis (PMF) group and the control group (p-value < 0.01). A high degree of neo-angiogenesis (demonstrated by analysis of CD34 expression) was detected in patients with myelofibrosis. There were correlations between latent TGF-β1 expression and microvascular density (r = 0.45; p-value < 0.0009) and between degree of microvascular density and fibrosis grade (r = 0.80; p-value < 0.0001). Remarkable differences for neo-angiogenesis were not observed between patients with essential thrombocythemia and controls. Conclusion Angiogenesis participates in the pathogenesis of primary myelofibrosis, in both the prefibrotic and fibrotic stages, while latent TGF-β is differentially expressed only in the prefibrotic stage.
Collapse
|
8
|
Xu Z, Zhang S, Zhou Q, Wang Y, Xia R. Endocan, a potential prognostic and diagnostic biomarker of acute leukemia. Mol Cell Biochem 2014; 395:117-23. [DOI: 10.1007/s11010-014-2117-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 06/02/2014] [Indexed: 01/07/2023]
|
9
|
Basu SK, Remick SC, Monga M, Gibson LF. Breaking and entering into the CNS: clues from solid tumor and nonmalignant models with relevance to hematopoietic malignancies. Clin Exp Metastasis 2013; 31:257-67. [PMID: 24306183 DOI: 10.1007/s10585-013-9623-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 11/01/2013] [Indexed: 12/16/2022]
Abstract
Various malignancies invade the CNS sanctuary site, accounting for the vast majority of CNS neoplastic foci and contributing to significant morbidity as well as mortality. The blood-brain barrier (BBB) exhibits considerable impermeability to chemotherapeutic agents, severely limiting therapeutic options available for patients developing metastatic CNS involvement, accounting for poor outcomes. The mechanisms by which malignant cells breach the highly exclusive BBB and subsequently survive in this unique anatomical site remain poorly understood, with most of the current knowledge stemming from nonmalignant and solid malignancy models. While solid and hematologic malignancies may face different challenges once within the CNS (e.g., solid tumor parenchymal metastasis compared to masses/nodules/leptomeningeal disease in hematologic malignancies), commonality exists in the process of migrating across the BBB from the circulation. Specifically considering this last point, this review aims to survey the current mechanistic knowledge regarding malignant migration across the BBB, necessarily emphasizing the better studied solid tumor and nonmalignant models with the intention of highlighting both the current knowledge gap and additional work required to effectively consider how hematopoietic malignancies breach the CNS.
Collapse
Affiliation(s)
- Soumit K Basu
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program, Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, WV, USA,
| | | | | | | |
Collapse
|