1
|
Fernandes MB, Gomes AM, Oliveira ML, Caldas J, Lúcio P, Kim R, Caye-Eude A, Pereira F, De Sousa AB, De Stefano A, Follo MY, Soares MV, Lacerda JF, Desterro J, Cavé H, Clappier E, Duarte X, Ribeiro P, Barata JT. Differential activation of basal and IL-7-induced PI3K/Akt/ mTOR and JAK/STAT5 signaling distinguishes pediatric from adult acute lymphoblastic leukemia. Haematologica 2024; 109:2671-2675. [PMID: 38654666 PMCID: PMC11290547 DOI: 10.3324/haematol.2023.284102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Marta B Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - A Margarida Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - Mariana L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | | | - Paulo Lúcio
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal; Champalimaud Centre for the Unknown, Lisboa
| | - Rathana Kim
- Hematology Laboratory, Saint-Louis Hospital, AP-HP, Paris, France; Saint-Louis Research Institute, Université de Paris, INSERM U944/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7212, Paris
| | - Aurélie Caye-Eude
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Filomena Pereira
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa
| | | | - Alessia De Stefano
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Portugal; University of Bologna, Department of Biomedical and Neuromotor Sciences
| | - Matilde Y Follo
- University of Bologna, Department of Biomedical and Neuromotor Sciences
| | - Maria V Soares
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - João F Lacerda
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - Joana Desterro
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa
| | - Hélène Cavé
- Saint-Louis Research Institute, Université de Paris, INSERM U944/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7212, Paris, France; Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris
| | - Emmanuelle Clappier
- Champalimaud Centre for the Unknown, Lisboa, Portugal; Hematology Laboratory, Saint-Louis Hospital, AP-HP, Paris
| | - Ximo Duarte
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa
| | | | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa.
| |
Collapse
|
2
|
Liang D, Wang Q, Zhang W, Tang H, Song C, Yan Z, Liang Y, Wang H. JAK/STAT in leukemia: a clinical update. Mol Cancer 2024; 23:25. [PMID: 38273387 PMCID: PMC10811937 DOI: 10.1186/s12943-023-01929-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Over the past three decades, considerable efforts have been expended on understanding the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway in leukemia, following the identification of the JAK2V617F mutation in myeloproliferative neoplasms (MPNs). The aim of this review is to summarize the latest progress in our understanding of the involvement of the JAK/STAT signaling pathway in the development of leukemia. We also attempt to provide insights into the current use of JAK/STAT inhibitors in leukemia therapy and explore pertinent clinical trials in this field.
Collapse
Affiliation(s)
- Dong Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qiaoli Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wenbiao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Cailu Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhimin Yan
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
| | - Yang Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Hua Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
3
|
Maninang C, Li J, Li W. Expression and prognostic role of STAT5a across cancer types. Biosci Rep 2023; 43:BSR20230612. [PMID: 37369132 PMCID: PMC10407157 DOI: 10.1042/bsr20230612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023] Open
Abstract
Studies examining the role of signal transducer and activator of transcription 5 (STAT5) in various cancers have produced controversial results. To address this controversy, we examined the prognostic role of STAT5a in cancer patients across multiple cancers. Transcription levels of STAT5a between tumors and normal tissues, obtained from public databases, were analyzed for statistical differences using Cox regression analysis with the outcome as overall survival and covariate of interest as high STAT5a expression. Meta-analysis was then conducted to summarize the hazard ratio estimate from the Cox regression analyses. We found that STAT5a was significantly under-expressed in breast, lung, and ovarian cancers, while STAT5a was significantly overexpressed in lymphoid neoplasm diffuse large B-cell lymphoma, glioblastoma, and glioma. High STAT5a expression was significantly associated with favorable survival in bladder cancer (lnHR = -0.8689 [-1.4087, -0.3292], P-value = 0.0016), breast cancer (lnHR = -0.7805 [-1.1394, -0.4215], P-value < 0.0001) and lung cancer (lnHR = -0.3255 [-0.6427, -0.0083], P-value = 0.0443). After adjusting for clinicopathological factors, high STAT5a expression remained significantly associated with favorable survival in breast cancer (lnHR = -0.6091 [-1.0810, -0.1372], P-value = 0.0114). These results suggest that higher STAT5a expression is associated with favorable overall survival in breast cancer, and therefore might have protective effects, and that STAT5a expression could be a potential prognostic biomarker, especially in breast cancer. However, the prognostic role of STAT5a is dependent on cancer type.
Collapse
Affiliation(s)
- Christine Maninang
- Department of Medicine, University of California San Diego, La Jolla, CA, U.S.A
| | - Jinghong Li
- Department of Medicine, University of California San Diego, La Jolla, CA, U.S.A
| | - Willis X. Li
- Department of Medicine, University of California San Diego, La Jolla, CA, U.S.A
| |
Collapse
|
4
|
Targeting Pim kinases in hematological cancers: molecular and clinical review. Mol Cancer 2023; 22:18. [PMID: 36694243 PMCID: PMC9875428 DOI: 10.1186/s12943-023-01721-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
Decades of research has recognized a solid role for Pim kinases in lymphoproliferative disorders. Often up-regulated following JAK/STAT and tyrosine kinase receptor signaling, Pim kinases regulate cell proliferation, survival, metabolism, cellular trafficking and signaling. Targeting Pim kinases represents an interesting approach since knock-down of Pim kinases leads to non-fatal phenotypes in vivo suggesting clinical inhibition of Pim may have less side effects. In addition, the ATP binding site offers unique characteristics that can be used for the development of small inhibitors targeting one or all Pim isoforms. This review takes a closer look at Pim kinase expression and involvement in hematopoietic cancers. Current and past clinical trials and in vitro characterization of Pim kinase inhibitors are examined and future directions are discussed. Current studies suggest that Pim kinase inhibition may be most valuable when accompanied by multi-drug targeting therapy.
Collapse
|
5
|
Lin M, Ku AT, Dong J, Yue F, Jiang W, Ibrahim AA, Peng F, Creighton CJ, Nagi C, Gutierrez C, Rosen JM, Zhang XHF, Hilsenbeck SG, Chen X, Du YCN, Huang S, Shi A, Fan Z, Li Y. STAT5 confers lactogenic properties in breast tumorigenesis and restricts metastatic potential. Oncogene 2022; 41:5214-5222. [PMID: 36261627 PMCID: PMC9701164 DOI: 10.1038/s41388-022-02500-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/09/2022]
Abstract
Signal transducer and activator of transcription 5 (STAT5) promotes cell survival and instigates breast tumor formation, and in the normal breast it also drives alveolar differentiation and lactogenesis. However, whether STAT5 drives a differentiated phenotype in breast tumorigenesis and therefore impacts cancer spread and metastasis is unclear. We found in two genetically engineered mouse models of breast cancer that constitutively activated Stat5a (Stat5aca) caused precancerous mammary epithelial cells to become lactogenic and evolve into tumors with diminished potential to metastasize. We also showed that STAT5aca reduced the migratory and invasive ability of human breast cancer cell lines in vitro. Furthermore, we demonstrated that STAT5aca overexpression in human breast cancer cells lowered their metastatic burden in xenografted mice. Moreover, RPPA, Western blotting, and studies of ChIPseq data identified several EMT drivers regulated by STAT5. In addition, bioinformatic studies detected a correlation between STAT5 activity and better prognosis of breast cancer patients. Together, we conclude that STAT5 activation during mammary tumorigenesis specifies a tumor phenotype of lactogenic differentiation, suppresses EMT, and diminishes potential for subsequent metastasis.
Collapse
Affiliation(s)
- Meng Lin
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.,Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Amy T Ku
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Jie Dong
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Fei Yue
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Weiyu Jiang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Ahmed Atef Ibrahim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Fanglue Peng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Chandandeep Nagi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Carolina Gutierrez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey M Rosen
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Yi-Chieh Nancy Du
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shixia Huang
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Department of Education, Innovation & Technology, Houston, TX, USA
| | - Aiping Shi
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhimin Fan
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
6
|
段 一, 文 飞. Recent research on the association between signal transducer and activator of transcription 5 and childhood acute lymphoblastic leukemia. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:942-947. [PMID: 36036135 PMCID: PMC9425862 DOI: 10.7499/j.issn.1008-8830.2203117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
Signal transducer and activator of transcription 5 (STAT5) can be involved in the processes such as cell proliferation, differentiation, apoptosis, and hematopoiesis, and its dysregulation is closely associated with the development and progression of malignant tumors including leukemia and may affect the treatment outcome and prognosis of pediatric patients. Identification of STAT5 can facilitate targeted therapy to improve the response rate of children with acute lymphoblastic leukemia. This article reviews the impact of STAT5 on the development/progression, targeted therapy strategies and the prognosis of childhood acute lymphoblastic leukemia.
Collapse
|
7
|
Chiriches C, Khan D, Wieske M, Guillen N, Rokicki M, Guy C, Wilson M, Heesom KJ, Ottmann OG, Ruthardt M. Activation of signaling pathways in models of t(6;9)-acute myeloid leukemia. Ann Hematol 2022; 101:2179-2193. [PMID: 35941390 PMCID: PMC9463248 DOI: 10.1007/s00277-022-04905-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/17/2022] [Indexed: 11/26/2022]
Abstract
Patients within the WHO-subgroup of t(6;9)-positive acute myeloid leukemia (AML) differ from other AML subgroups as they are characterised by younger age and a grim prognosis. Leukemic transformation can often be attributed to single chromosomal aberrations encoding oncogenes, in the case of t(6;9)-AML to the fusion protein DEK-CAN (also called DEK-NUP214). As being a rare disease there is the urgent need for models of t(6;9)-AML. The only cell line derived from a t(6;9)-AML patient currently available is FKH1. By using phospho-proteomics on FKH1 cells, we found a strongly activated ABL1 kinase. Further investigation revealed the presence of ETV6-ABL1. This finding renders necessary to determine DEK-CAN- and ETV6-ABL1-related features when using FKH1. This can be done as ETV6-ABL1 activity in FKH1 is responsive to imatinib. Nevertheless, we provided evidence that both SFK and mTOR activation in FKH1 are DEK-CAN-related features as they were activated also in other t(6;9) and DEK-CAN-positive models. The activation of STAT5 previously shown to be strong in t(6;9)-AML and activated by DEK-CAN is regulated in FKH1 by both DEK-CAN and ETV6-ABL1. In conclusion, FKH1 cells still represent a model for t(6;9)-AML and could serve as model for ETV6-ABL1-positive AML if the presence of these leukemia-inducing oncogenes is adequately considered.Taken together, all our results provide clear evidence of novel and specific interdependencies between leukemia-inducing oncogenes and cancer signaling pathways which will influence the design of therapeutic strategies to better address the complexity of cancer signaling.
Collapse
MESH Headings
- Chromosomal Proteins, Non-Histone/genetics
- Humans
- Imatinib Mesylate
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Oncogene Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Poly-ADP-Ribose Binding Proteins/metabolism
- Signal Transduction
- Translocation, Genetic
Collapse
Affiliation(s)
- Claudia Chiriches
- Division of Cancer and Genetics, Department of Haematology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
- Experimental Clinical Medical Center (ECMC) Cardiff, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
| | - Dilawar Khan
- Department of Hematology, J.W. Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Maria Wieske
- Department of Hematology, J.W. Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Nathalie Guillen
- Department of Hematology, J.W. Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Michal Rokicki
- Division of Cancer and Genetics, Department of Haematology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- Experimental Clinical Medical Center (ECMC) Cardiff, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Carol Guy
- Division of Cancer and Genetics, Department of Haematology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- Experimental Clinical Medical Center (ECMC) Cardiff, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Marieangela Wilson
- Biomedical Sciences Building, University of Bristol Proteomics Facility, Bristol, BS8 1TD, UK
| | - Kate J Heesom
- Biomedical Sciences Building, University of Bristol Proteomics Facility, Bristol, BS8 1TD, UK
| | - Oliver Gerhard Ottmann
- Division of Cancer and Genetics, Department of Haematology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- Experimental Clinical Medical Center (ECMC) Cardiff, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Martin Ruthardt
- Division of Cancer and Genetics, Department of Haematology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
- Experimental Clinical Medical Center (ECMC) Cardiff, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
| |
Collapse
|
8
|
Porazzi P, De Dominici M, Salvino J, Calabretta B. Targeting the CDK6 Dependence of Ph+ Acute Lymphoblastic Leukemia. Genes (Basel) 2021; 12:genes12091355. [PMID: 34573335 PMCID: PMC8467343 DOI: 10.3390/genes12091355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022] Open
Abstract
Ph+ ALL is a poor-prognosis leukemia subtype driven by the BCR-ABL1 oncogene, either the p190- or the p210-BCR/ABL isoform in a 70:30 ratio. Tyrosine Kinase inhibitors (TKIs) are the drugs of choice in the therapy of Ph+ ALL. In combination with standard chemotherapy, TKIs have markedly improved the outcome of Ph+ ALL, in particular if this treatment is followed by bone marrow transplantation. However, resistance to TKIs develops with high frequency, causing leukemia relapse that results in <5-year overall survival. Thus, new therapies are needed to address relapsed/TKI-resistant Ph+ ALL. We have shown that expression of cell cycle regulatory kinase CDK6, but not of the highly related CDK4 kinase, is required for the proliferation and survival of Ph+ ALL cells. Comparison of leukemia suppression induced by treatment with the clinically-approved dual CDK4/6 inhibitor palbociclib versus CDK6 silencing revealed that the latter treatment was markedly more effective, probably reflecting inhibition of CDK6 kinase-independent effects. Thus, we developed CDK4/6-targeted proteolysis-targeting chimeras (PROTACs) that preferentially degrade CDK6 over CDK4. One compound termed PROTAC YX-2-107, which degrades CDK6 by recruiting the Cereblon ubiquitin ligase, markedly suppressed leukemia burden in mice injected with de novo or TKI-resistant Ph+ ALL. The effect of PROTAC YX-2-107 was comparable or superior to that of palbociclib. The development of CDK6-selective PROTACs represents an effective strategy to exploit the “CDK6 dependence” of Ph+ ALL cells while sparing a high proportion of normal hematopoietic progenitors that depend on both CDK6 and CDK6 for their survival. In combination with other agents, CDK6-selective PROTACs may be valuable components of chemotherapy-free protocols for the therapy of Ph+ ALL and other CDK6-dependent hematological malignancies.
Collapse
Affiliation(s)
- Patrizia Porazzi
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
- Correspondence:
| | - Marco De Dominici
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | | | - Bruno Calabretta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
9
|
Therapeutic delivery of siRNA with polymeric carriers to down-regulate STAT5A expression in high-risk B-cell acute lymphoblastic leukemia (B-ALL). PLoS One 2021; 16:e0251719. [PMID: 34157051 PMCID: PMC8219370 DOI: 10.1371/journal.pone.0251719] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/02/2021] [Indexed: 11/19/2022] Open
Abstract
Overexpression and persistent activation of STAT5 play an important role in the development and progression of acute lymphoblastic leukemia (ALL), the most common pediatric cancer. Small interfering RNA (siRNA)-mediated downregulation of STAT5 represents a promising therapeutic approach for ALL to overcome the limitations of current treatment modalities such as high relapse rates and poor prognosis. However, to effectively transport siRNA molecules to target cells, development of potent carriers is of utmost importance to surpass hurdles of delivery. In this study, we investigated the use of lipopolymers as non-viral delivery systems derived from low molecular weight polyethylenimines (PEI) substituted with lauric acid (Lau), linoleic acid (LA) and stearic acid (StA) to deliver siRNA molecules to ALL cell lines and primary samples. Among the lipid-substituted polymers explored, Lau- and LA-substituted PEI displayed excellent siRNA delivery to SUP-B15 and RS4;11 cells. STAT5A gene expression was downregulated (36-92%) in SUP-B15 and (32%) in RS4;11 cells using the polymeric delivery systems, which consequently reduced cell growth and inhibited the formation of colonies in ALL cells. With regard to ALL primary cells, siRNA-mediated STAT5A gene silencing was observed in four of eight patient cells using our leading polymeric delivery system, 1.2PEI-Lau8, accompanied by the significant reduction in colony formation in three of eight patients. In both BCR-ABL positive and negative groups, three of five patients demonstrated marked cell growth inhibition in both MTT and trypan blue exclusion assays using 1.2PEI-Lau8/siRNA complexes in comparison with their control siRNA groups. Three patient samples did not show any positive results with our delivery systems. Differential therapeutic responses to siRNA therapy observed in different patients could result from variable genetic profiles and patient-to-patient variability in delivery. This study supports the potential of siRNA therapy and the designed lipopolymers as a delivery system in ALL therapy.
Collapse
|
10
|
Wang X, Ding X, Yan J, Lu Z, Cao H, Ni X, Ying Y. STAT5 inhibitor attenuates atherosclerosis via inhibition of inflammation: the role of STAT5 in atherosclerosis. Am J Transl Res 2021; 13:1422-1431. [PMID: 33841667 PMCID: PMC8014372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/11/2020] [Indexed: 06/12/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease driven by lipids, which occurs preferentially in the branches or curved areas of the middle and large arteries, contributing to increased morbidity and mortality of cardiovascular disease. Recently, it has been reported that STAT5 and its regulated immune response are closely related to non-tumor diseases. However, the role of STAT5 in the development of atherosclerosis remains unknown. In this study, atherosclerosis was induced by high-fat diet (HFD) in ApoE-/- mice, and STAT5-IN-1, a STAT5 inhibitor, was orally given. Macrophages stimulated by oxLDL were used as cell models in vitro. The effects of STAT5-IN-1 in ApoE-/- mice induced by HFD were assessed, and the underlying mechanisms were investigated by siRNA-induced gene silencing. The results revealed that treatment with STAT5 inhibitor significantly attenuated atherosclerosis in ApoE-/- mice induced by HFD via decreasing inflammation. Furthermore, it was demonstrated that inhibiting STAT5 could decrease oxLDL-induced inflammation. In summary, STAT5-IN-1 may be a potential drug for the treatment of atherosclerosis, and targeting STAT5 has the ability to be a potential therapeutic strategy for reducing atherosclerosis.
Collapse
Affiliation(s)
- Xiaodong Wang
- Sino-German Joint Research Center of Vascular Surgery, Zhejiang Academy of Traditional Chinese MedicineHangzhou 310012, Zhejiang, China
- Department of Vascular Surgery, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Xiaoji Ding
- Department of Pharmacy, Zhejiang Academy of Traditional Chinese MedicineHangzhou 310012, Zhejiang, China
- Department of Pharmacy, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Jin Yan
- Department of Vascular Surgery, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Ziying Lu
- Department of Vascular Surgery, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Haoyang Cao
- Department of Vascular Surgery, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Xiaolong Ni
- Department of Vascular Surgery, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Yin Ying
- Department of Pharmacy, Zhejiang Academy of Traditional Chinese MedicineHangzhou 310012, Zhejiang, China
- Department of Pharmacy, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| |
Collapse
|
11
|
Involvement of STAT5 in Oncogenesis. Biomedicines 2020; 8:biomedicines8090316. [PMID: 32872372 PMCID: PMC7555335 DOI: 10.3390/biomedicines8090316] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022] Open
Abstract
Signal transducer and activator of transcription (STAT) proteins, and in particular STAT3, have been established as heavily implicated in cancer. Recently, the involvement of STAT5 signalling in the pathology of cancer has been shown to be of increasing importance. STAT5 plays a crucial role in the development of the mammary gland and the homeostasis of the immune system. However, in various cancers, aberrant STAT5 signalling promotes the expression of target genes, such as cyclin D, Bcl-2 and MMP-2, that result in increased cell proliferation, survival and metastasis. To target constitutive STAT5 signalling in cancers, there are several STAT5 inhibitors that can prevent STAT5 phosphorylation, dimerisation, or its transcriptional activity. Tyrosine kinase inhibitors (TKIs) that target molecules upstream of STAT5 could also be utilised. Consequently, since STAT5 contributes to tumour aggressiveness and cancer progression, inhibiting STAT5 constitutive activation in cancers that rely on its signalling makes for a promising targeted treatment option.
Collapse
|
12
|
De Dominici M, Porazzi P, Xiao Y, Chao A, Tang HY, Kumar G, Fortina P, Spinelli O, Rambaldi A, Peterson LF, Petruk S, Barletta C, Mazo A, Cingolani G, Salvino JM, Calabretta B. Selective inhibition of Ph-positive ALL cell growth through kinase-dependent and -independent effects by CDK6-specific PROTACs. Blood 2020; 135:1560-1573. [PMID: 32040545 PMCID: PMC7193186 DOI: 10.1182/blood.2019003604] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/23/2020] [Indexed: 12/13/2022] Open
Abstract
Expression of the cell cycle regulatory gene CDK6 is required for Philadelphia-positive (Ph+) acute lymphoblastic leukemia (ALL) cell growth, whereas expression of the closely related CDK4 protein is dispensable. Moreover, CDK6 silencing is more effective than treatment with the dual CDK4/6 inhibitor palbociclib in suppressing Ph+ ALL in mice, suggesting that the growth-promoting effects of CDK6 are, in part, kinase-independent in Ph+ ALL. Accordingly, we developed CDK4/6-targeted proteolysis-targeting chimeras (PROTACs) that inhibit CDK6 enzymatic activity in vitro, promote the rapid and preferential degradation of CDK6 over CDK4 in Ph+ ALL cells, and markedly suppress S-phase cells concomitant with inhibition of CDK6-regulated phospho-RB and FOXM1 expression. No such effects were observed in CD34+ normal hematopoietic progenitors, although CDK6 was efficiently degraded. Treatment with the CDK6-degrading PROTAC YX-2-107 markedly suppressed leukemia burden in mice injected with de novo or tyrosine kinase inhibitor-resistant primary Ph+ ALL cells, and this effect was comparable or superior to that of the CDK4/6 enzymatic inhibitor palbociclib. These studies provide "proof of principle" that targeting CDK6 with PROTACs that inhibit its enzymatic activity and promote its degradation represents an effective strategy to exploit the "CDK6 dependence" of Ph+ ALL and, perhaps, of other hematologic malignancies. Moreover, they suggest that treatment of Ph+ ALL with CDK6-selective PROTACs would spare a high proportion of normal hematopoietic progenitors, preventing the neutropenia induced by treatment with dual CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Marco De Dominici
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Patrizia Porazzi
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | | | | | - Gaurav Kumar
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Paolo Fortina
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Orietta Spinelli
- Hematology and Bone Marrow Transplant Unit, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, Ospedale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology-Oncology, Università Statale Milano, Milan, Italy
| | - Luke F Peterson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI; and
| | - Svetlana Petruk
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Camilla Barletta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Alexander Mazo
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Gino Cingolani
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | - Bruno Calabretta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
13
|
Brachet-Botineau M, Polomski M, Neubauer HA, Juen L, Hédou D, Viaud-Massuard MC, Prié G, Gouilleux F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers (Basel) 2020; 12:E240. [PMID: 31963765 PMCID: PMC7016966 DOI: 10.3390/cancers12010240] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Signal Transducer and Activator of Transcription (STAT) 3 and 5 are important effectors of cellular transformation, and aberrant STAT3 and STAT5 signaling have been demonstrated in hematopoietic cancers. STAT3 and STAT5 are common targets for different tyrosine kinase oncogenes (TKOs). In addition, STAT3 and STAT5 proteins were shown to contain activating mutations in some rare but aggressive leukemias/lymphomas. Both proteins also contribute to drug resistance in hematopoietic malignancies and are now well recognized as major targets in cancer treatment. The development of inhibitors targeting STAT3 and STAT5 has been the subject of intense investigations during the last decade. This review summarizes the current knowledge of oncogenic STAT3 and STAT5 functions in hematopoietic cancers as well as advances in preclinical and clinical development of pharmacological inhibitors.
Collapse
Affiliation(s)
- Marie Brachet-Botineau
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| | - Marion Polomski
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria;
| | - Ludovic Juen
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Damien Hédou
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Marie-Claude Viaud-Massuard
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Gildas Prié
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Fabrice Gouilleux
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| |
Collapse
|
14
|
Maurer B, Kollmann S, Pickem J, Hoelbl-Kovacic A, Sexl V. STAT5A and STAT5B-Twins with Different Personalities in Hematopoiesis and Leukemia. Cancers (Basel) 2019; 11:E1726. [PMID: 31690038 PMCID: PMC6895831 DOI: 10.3390/cancers11111726] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factors STAT5A and STAT5B have essential roles in survival and proliferation of hematopoietic cells-which have been considered largely redundant. Mutations of upstream kinases, copy number gains, or activating mutations in STAT5A, or more frequently in STAT5B, cause altered hematopoiesis and cancer. Interfering with their activity by pharmacological intervention is an up-and-coming therapeutic avenue. Precision medicine requests detailed knowledge of STAT5A's and STAT5B's individual functions. Recent evidence highlights the privileged role for STAT5B over STAT5A in normal and malignant hematopoiesis. Here, we provide an overview on their individual functions within the hematopoietic system.
Collapse
Affiliation(s)
- Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Judith Pickem
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
15
|
Polak KL, Chernosky NM, Smigiel JM, Tamagno I, Jackson MW. Balancing STAT Activity as a Therapeutic Strategy. Cancers (Basel) 2019; 11:cancers11111716. [PMID: 31684144 PMCID: PMC6895889 DOI: 10.3390/cancers11111716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Driven by dysregulated IL-6 family member cytokine signaling in the tumor microenvironment (TME), aberrant signal transducer and activator of transcription (STAT3) and (STAT5) activation have been identified as key contributors to tumorigenesis. Following transformation, persistent STAT3 activation drives the emergence of mesenchymal/cancer-stem cell (CSC) properties, important determinants of metastatic potential and therapy failure. Moreover, STAT3 signaling within tumor-associated macrophages and neutrophils drives secretion of factors that facilitate metastasis and suppress immune cell function. Persistent STAT5 activation is responsible for cancer cell maintenance through suppression of apoptosis and tumor suppressor signaling. Furthermore, STAT5-mediated CD4+/CD25+ regulatory T cells (Tregs) have been implicated in suppression of immunosurveillance. We discuss these roles for STAT3 and STAT5, and weigh the attractiveness of different modes of targeting each cancer therapy. Moreover, we discuss how anti-tumorigenic STATs, including STAT1 and STAT2, may be leveraged to suppress the pro-tumorigenic functions of STAT3/STAT5 signaling.
Collapse
Affiliation(s)
- Kelsey L Polak
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Noah M Chernosky
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Jacob M Smigiel
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Mark W Jackson
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
16
|
Mohseni M, Uludag H, Brandwein JM. Advances in biology of acute lymphoblastic leukemia (ALL) and therapeutic implications. AMERICAN JOURNAL OF BLOOD RESEARCH 2018; 8:29-56. [PMID: 30697448 PMCID: PMC6334189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/06/2018] [Indexed: 06/09/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer and also occurs in adults. Although the outcomes of multi-agent chemotherapy regimens have greatly improved, high toxicity and relapses in many patients necessitate the development of novel therapeutic approaches. Advances in molecular profiling and cytogenetics have identified a broad range of genetic abnormalities, including gene mutations, chromosome translocations and aneuploidy, which has provided a more comprehensive understanding of the biology and pathogenesis of ALL. This understanding has also led to new targeted therapeutic approaches, including the use of selective small molecule inhibitors, nucleic acid-based therapies and immune-based therapies mediated by specific monoclonal antibodies and cellular immunotherapy, which are poised to revolutionize the treatment of various ALL subtypes. The main focus of this review is to highlight the latest advances in ALL biology, including the identification of prognostic factors and putative therapeutic targets. We also review the current status of, and ongoing progress in, the development of targeted therapies for ALL.
Collapse
Affiliation(s)
- Mahsa Mohseni
- Department of Medicine, University of Alberta Edmonton, Alberta, Canada
| | - Hasan Uludag
- Department of Chemical and Materials Engineering, University of Alberta Edmonton, Alberta, Canada
| | | |
Collapse
|
17
|
Targeting the STAT5 pathway in Ph+ acute lymphoblastic leukemia. Oncotarget 2018; 9:36726-36727. [PMID: 30613358 PMCID: PMC6298402 DOI: 10.18632/oncotarget.26412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 11/23/2018] [Indexed: 11/25/2022] Open
|