1
|
Casado P, Marfa S, Hadi MM, Gerdes H, Martin-Guerrero SM, Miraki-Moud F, Rajeeve V, Cutillas PR. Phosphoproteomics identifies determinants of PAK inhibitor sensitivity in leukaemia cells. Cell Commun Signal 2025; 23:135. [PMID: 40082888 PMCID: PMC11907924 DOI: 10.1186/s12964-025-02107-0] [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: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The P21 activated kinases (PAK) are frequently dysregulated in cancer and have central roles in oncogenic signalling, prompting the development of PAK inhibitors (PAKi) as anticancer agents. However, such compounds have not reached clinical use because, at least partially, there is a limited mechanistic understanding of their mode of action. Here, we aimed to characterize functional and molecular responses to PAKi (PF-3758309, FRAX-486 and IPA-3) in multiple acute myeloid leukaemia (AML) models to gain insights on the biochemical pathways affected by these inhibitors in this disease and identify determinants of response in patient samples. METHODS We mined phosphoproteomic datasets of primary AML, and used proteomics and phosphoproteomics to profile PAKi impact in immortalized (P31/Fuj and MV4-11), and primary AML cells from 8 AML patients. These omics datasets were integrated with gene dependency data to identify which proteins targeted by PAKi are necessary for the proliferation of AML. We studied the effect PAKi on cell cycle progression, proliferation, differentiation and apoptosis. Finally, we used phosphoproteomics data as input for machine learning models that predicted ex vivo response in two independent datasets of primary AML cells (with 36 and 50 cases, respectively) to PF-3758309 and identify markers of response. RESULTS We found that PAK1 activation- measured from phosphoproteomics data- was predictive of poor prognosis in primary AML cases. PF-3758309 was the most effective PAKi in reducing proliferation and inducing apoptosis in AML cell lines. In cell lines and primary cells, PF-3758309 inhibited PAK, AMPK and PKCA activities, reduced c-MYC transcriptional activity and the expression of ribosomal proteins, and targeted the FLT3 pathway in FLT3-ITD mutated cells. In primary cells, PF-3758309 reduced STAT5 phosphorylation at Tyr699. Functionally, PF-3758309 reduced cell-growth, induced apoptosis, blocked cell cycle progression and promoted differentiation in a model-dependent manner. ML modelling accurately classified primary AML samples as sensitive or resistant to PF-3758309 ex vivo treatment, and highlighted PHF2 phosphorylation at Ser705 as a robust response biomarker. CONCLUSIONS In summary, our data define the proteomic, molecular and functional responses of primary and immortalised AML cells to PF-3758309 and suggest a route to personalise AML treatments based on PAK inhibitors.
Collapse
Affiliation(s)
- Pedro Casado
- Centre for Cancer Evolution, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK.
| | - Santiago Marfa
- Centre for Cancer Evolution, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Marym M Hadi
- Centre for Cancer Evolution, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Henry Gerdes
- Centre for Cancer Evolution, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Sandra M Martin-Guerrero
- Centre for Cancer Evolution, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Farideh Miraki-Moud
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Vinothini Rajeeve
- Centre for Cancer Evolution, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Pedro R Cutillas
- Centre for Cancer Evolution, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK.
| |
Collapse
|
2
|
Bogdanov K, Kudryavtseva E, Fomicheva Y, Churkina I, Lomaia E, Girshova L, Osipov Y, Zaritskey A. Shift of N-MYC Oncogene Expression in AML Patients Carrying the FLT3-ITD Mutation. PATHOPHYSIOLOGY 2023; 30:296-313. [PMID: 37606386 PMCID: PMC10443239 DOI: 10.3390/pathophysiology30030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/20/2023] [Accepted: 07/29/2023] [Indexed: 08/23/2023] Open
Abstract
Mutations in the FLT3 gene not only lead to abnormalities in its structure and function, but also affect the expression of other genes involved in leukemogenesis. This study evaluated the expression of genes that are more characteristic of neuroblastoma but less studied in leukemia. N-MYC oncogene expression was found to be more than 3-fold higher in primary AML patients carrying the FLT3-ITD mutation compared to carriers of other mutations as well as patients with normal karyotype (p = 0.03946). In contrast to the expression of several genes (C-MYC, SPT16, AURKA, AURKB) directly correlated to the allelic load of FLT3-ITD, the expression of the N-MYC oncogene is extremely weakly related or independent of it (p = 0.0405). Monitoring of N-MYC expression in some patients with high FLT3-ITD allelic load receiving therapy showed that a decrease in FLT3-ITD allelic load is not always accompanied by a decrease in N-MYC expression. On the contrary, N-MYC expression may remain elevated during the first three months after therapy, which is additional evidence of the emergence of resistance to therapy and progression of AML.
Collapse
Affiliation(s)
- Konstantin Bogdanov
- Almazov National Medical Research Centre, 2 Akkuratova Str., Saint Petersburg 197341, Russia; (E.K.); (Y.F.); (I.C.); (E.L.); (L.G.); (Y.O.); (A.Z.)
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Li CF, Chan TC, Fang FM, Yu SC, Huang HY. PAK1 overexpression promotes myxofibrosarcoma angiogenesis through STAT5B-mediated CSF2 transactivation: clinical and therapeutic relevance of amplification and nuclear entry. Int J Biol Sci 2023; 19:3920-3936. [PMID: 37564209 PMCID: PMC10411477 DOI: 10.7150/ijbs.83467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
Myxofibrosarcoma is genetically complex without established nonsurgical therapies. In public datasets, PAK1 was recurrently gained with mRNA upregulation. Using myxofibrosarcoma cells, we explored the oncogenic underpinning of PAK1 with genetic manipulation and a pan-PAK inhibitor (PF3758309). Myxofibrosarcoma specimens were analyzed for the levels of PAK1, phospho-PAKT423, CSF2 and microvascular density (MVD) and those of PAK1 gene and mRNA. PAK1-expressing xenografts were assessed for the effects of PF3758309 and CSF2 silencing. Besides pro-proliferative and pro-migrator/pro-invasive attributes, PAK1 strongly enhanced angiogenesis in vitro, which, not phenocopied by PAK2-4, was identified as CSF2-mediated using antibody arrays. PAK1 underwent phosphorylation at tyrosines153,201,285 and threonine423 to facilitate nuclear entry, whereby nuclear PAK1 bound STAT5B to co-transactivate the CSF2 promoter, increasing CSF2 secretion needed for angiogenesis. Angiogenesis driven by PAK1-upregulated CSF2 was negated by CSF2 silencing, anti-CSF2, and PF3758309. Clinically, overexpressed whole-cell phospho-PAKT423, related to PAK1 amplification, was associated with increased grades, stages, and PAK1 mRNA, higher MVD, and CSF2 overexpression. Overexpressed whole-cell phospho-PAKT423 and CSF2 independently portended shorter metastasis-free survival and disease-specific survival, respectively. In vivo, both CSF2 silencing and PF3758309 suppressed PAK1-driven tumor proliferation and angiogenesis. Conclusively, the nuclear entry of overexpressed/activated PAK1 endows myxofibrosarcomas with pro-angiogenic function, highlighting the vulnerable PAK1/STAT5B/CSF2 regulatory axis.
Collapse
Affiliation(s)
- Chien-Feng Li
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ti-Chun Chan
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Fu-Min Fang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Chen Yu
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsuan-Ying Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
4
|
Azlan A, Khor KZ, Rajasegaran Y, Rosli AA, Said MSM, Yusoff NM, Moses EJ. RUNX1/ETO regulates reactive oxygen species (ROS) levels in t(8,21) acute myeloid leukaemia via FLT3 and RAC1. Med Oncol 2023; 40:208. [PMID: 37341821 DOI: 10.1007/s12032-023-02075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023]
Abstract
Reactive oxygen species (ROS) homeostasis is crucial for leukaemogenesisand deregulation would hamper leukaemic progression. Although the regulatory effects of RUNX1/ETO has been extensively studied, its underlying molecular mechanims in ROS production in t(8,21) AML is yet to be fully elucidated. Here, we report that RUNX1/ETO could directly control FLT3 by occupying several DNA elements on FLT3 locus. The possible hijacking mechanism by RUNX1/ETO over FLT3 mediated ROS modulation in AML t(8;21) was made apparent when suppression of RUNX1/ETO led to decrement in ROS levels and the direct oxidative marker FOXO3 but not in FLT3 and RAC1 suppressed t(8,21) AML cell line Furthermore, nuclear import of RUNX1/ETO was aberrated following RUNX1/ETO and RAC1 suppression suggesting association in ROS control. A different picture was depicted in non t(8;21) cells where suppression of RAC1 and FLT3 led to decreased levels of FOXO3a and ROS. Results alltogether indicate a possible dysregulation of ROS levels by RUNX1/ETO in t(8,21) AML.
Collapse
Affiliation(s)
- Adam Azlan
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Kang Zi Khor
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Yaashini Rajasegaran
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Aliaa Arina Rosli
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | | | - Narazah Mohd Yusoff
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Emmanuel Jairaj Moses
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia.
| |
Collapse
|
5
|
Ye J, Wu J, Liu B. Therapeutic strategies of dual-target small molecules to overcome drug resistance in cancer therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:188866. [PMID: 36842765 DOI: 10.1016/j.bbcan.2023.188866] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/12/2023] [Accepted: 01/31/2023] [Indexed: 02/28/2023]
Abstract
Despite some advances in targeted therapeutics of human cancers, curative cancer treatment still remains a tremendous challenge due to the occurrence of drug resistance. A variety of underlying resistance mechanisms to targeted cancer drugs have recently revealed that the dual-target therapeutic strategy would be an attractive avenue. Compared to drug combination strategies, one agent simultaneously modulating two druggable targets generally shows fewer adverse reactions and lower toxicity. As a consequence, the dual-target small molecule has been extensively explored to overcome drug resistance in cancer therapy. Thus, in this review, we focus on summarizing drug resistance mechanisms of cancer cells, such as enhanced drug efflux, deregulated cell death, DNA damage repair, and epigenetic alterations. Based upon the resistance mechanisms, we further discuss the current therapeutic strategies of dual-target small molecules to overcome drug resistance, which will shed new light on exploiting more intricate mechanisms and relevant dual-target drugs for future cancer therapeutics.
Collapse
Affiliation(s)
- Jing Ye
- State Key Laboratory of Biotherapy and Cancer Center and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhao Wu
- State Key Laboratory of Biotherapy and Cancer Center and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center and Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Santos JC, Profitós-Pelejà N, Sánchez-Vinces S, Roué G. RHOA Therapeutic Targeting in Hematological Cancers. Cells 2023; 12:cells12030433. [PMID: 36766776 PMCID: PMC9914237 DOI: 10.3390/cells12030433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Primarily identified as an important regulator of cytoskeletal dynamics, the small GTPase Ras homolog gene family member A (RHOA) has been implicated in the transduction of signals regulating a broad range of cellular functions such as cell survival, migration, adhesion and proliferation. Deregulated activity of RHOA has been linked to the growth, progression and metastasis of various cancer types. Recent cancer genome-wide sequencing studies have unveiled both RHOA gain and loss-of-function mutations in primary leukemia/lymphoma, suggesting that this GTPase may exert tumor-promoting or tumor-suppressive functions depending on the cellular context. Based on these observations, RHOA signaling represents an attractive therapeutic target for the development of selective anticancer strategies. In this review, we will summarize the molecular mechanisms underlying RHOA GTPase functions in immune regulation and in the development of hematological neoplasms and will discuss the current strategies aimed at modulating RHOA functions in these diseases.
Collapse
Affiliation(s)
- Juliana Carvalho Santos
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Núria Profitós-Pelejà
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Salvador Sánchez-Vinces
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista 01246-100, São Paulo, Brazil
| | - Gaël Roué
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
- Correspondence: ; Tel.: +34-935572835
| |
Collapse
|
7
|
Sakai J, Yang J, Chou CK, Wu WW, Akkoyunlu M. B cell receptor-induced IL-10 production from neonatal mouse CD19 +CD43 - cells depends on STAT5-mediated IL-6 secretion. eLife 2023; 12:83561. [PMID: 36735294 PMCID: PMC9934864 DOI: 10.7554/elife.83561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/31/2023] [Indexed: 02/04/2023] Open
Abstract
Newborns are unable to reach the adult-level humoral immune response partly due to the potent immunoregulatory role of IL-10. Increased IL-10 production by neonatal B cells has been attributed to the larger population of IL-10-producting CD43+ B-1 cells in neonates. Here, we show that neonatal mouse CD43- non-B-1 cells also produce substantial amounts of IL-10 following B cell antigen receptor (BCR) activation. In neonatal mouse CD43- non-B-1 cells, BCR engagement activated STAT5 under the control of phosphorylated forms of signaling molecules Syk, Btk, PKC, FAK, and Rac1. Neonatal STAT5 activation led to IL-6 production, which in turn was responsible for IL-10 production in an autocrine/paracrine fashion through the activation of STAT3. In addition to the increased IL-6 production in response to BCR stimulation, elevated expression of IL-6Rα expression in neonatal B cells rendered them highly susceptible to IL-6-mediated STAT3 phosphorylation and IL-10 production. Finally, IL-10 secreted from neonatal mouse CD43- non-B-1 cells was sufficient to inhibit TNF-α secretion by macrophages. Our results unveil a distinct mechanism of IL-6-dependent IL-10 production in BCR-stimulated neonatal CD19+CD43- B cells.
Collapse
Affiliation(s)
- Jiro Sakai
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, The US Food and Drug AdministrationSilver SpringUnited States
| | - Jiyeon Yang
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, The US Food and Drug AdministrationSilver SpringUnited States
| | - Chao-Kai Chou
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, United States Food and Drug AdministrationSilver SpringUnited States
| | - Wells W Wu
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, United States Food and Drug AdministrationSilver SpringUnited States
| | - Mustafa Akkoyunlu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, The US Food and Drug AdministrationSilver SpringUnited States
| |
Collapse
|
8
|
Sorger H, Dey S, Vieyra‐Garcia PA, Pölöske D, Teufelberger AR, de Araujo ED, Sedighi A, Graf R, Spiegl B, Lazzeri I, Braun T, Garces de los Fayos Alonso I, Schlederer M, Timelthaler G, Kodajova P, Pirker C, Surbek M, Machtinger M, Graier T, Perchthaler I, Pan Y, Fink‐Puches R, Cerroni L, Ober J, Otte M, Albrecht JD, Tin G, Abdeldayem A, Manaswiyoungkul P, Olaoye OO, Metzelder ML, Orlova A, Berger W, Wobser M, Nicolay JP, André F, Nguyen VA, Neubauer HA, Fleck R, Merkel O, Herling M, Heitzer E, Gunning PT, Kenner L, Moriggl R, Wolf P. Blocking STAT3/5 through direct or upstream kinase targeting in leukemic cutaneous T-cell lymphoma. EMBO Mol Med 2022; 14:e15200. [PMID: 36341492 PMCID: PMC9727928 DOI: 10.15252/emmm.202115200] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 09/26/2022] [Accepted: 10/02/2022] [Indexed: 11/09/2022] Open
Abstract
Leukemic cutaneous T-cell lymphomas (L-CTCL) are lymphoproliferative disorders of skin-homing mature T-cells causing severe symptoms and high mortality through chronic inflammation, tissue destruction, and serious infections. Despite numerous genomic sequencing efforts, recurrent driver mutations have not been identified, but chromosomal losses and gains are frequent and dominant. We integrated genomic landscape analyses with innovative pharmacologic interference studies to identify key vulnerable nodes in L-CTCL. We detected copy number gains of loci containing the STAT3/5 oncogenes in 74% (n = 17/23) of L-CTCL, which correlated with the increased clonal T-cell count in the blood. Dual inhibition of STAT3/5 using small-molecule degraders and multi-kinase blockers abolished L-CTCL cell growth in vitro and ex vivo, whereby PAK kinase inhibition was specifically selective for L-CTCL patient cells carrying STAT3/5 gains. Importantly, the PAK inhibitor FRAx597 demonstrated encouraging anti-leukemic activity in vivo by inhibiting tumor growth and disease dissemination in intradermally xenografted mice. We conclude that STAT3/5 and PAK kinase interaction represents a new therapeutic node to be further explored in L-CTCL.
Collapse
Affiliation(s)
- Helena Sorger
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
- Department of Pediatric and Adolescent Surgery, Vienna General HospitalMedical University of ViennaViennaAustria
| | - Saptaswa Dey
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
- Department of PathologyMedical University of ViennaViennaAustria
| | | | - Daniel Pölöske
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | | | - Elvin D de Araujo
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Abootaleb Sedighi
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Ricarda Graf
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Benjamin Spiegl
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Isaac Lazzeri
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Till Braun
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
| | - Ines Garces de los Fayos Alonso
- Department of PathologyMedical University of ViennaViennaAustria
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | | | | | - Petra Kodajova
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Christine Pirker
- Centre for Cancer ResearchMedical University of ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Marta Surbek
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Michael Machtinger
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Thomas Graier
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | | | - Yi Pan
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Regina Fink‐Puches
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Lorenzo Cerroni
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Jennifer Ober
- Core Facility Flow Cytometry, Center for Medical Research (ZMF)Medical University of GrazGrazAustria
| | - Moritz Otte
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
| | - Jana D Albrecht
- Department of DermatologyUniversity Hospital MannheimMannheimGermany
| | - Gary Tin
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Ayah Abdeldayem
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Pimyupa Manaswiyoungkul
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Olasunkanmi O Olaoye
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Martin L Metzelder
- Department of Pediatric and Adolescent Surgery, Vienna General HospitalMedical University of ViennaViennaAustria
| | - Anna Orlova
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Walter Berger
- Centre for Cancer ResearchMedical University of ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Marion Wobser
- Department of DermatologyUniversity Hospital WuerzburgWuerzburgGermany
| | - Jan P Nicolay
- Department of DermatologyUniversity Hospital MannheimMannheimGermany
| | - Fiona André
- University Clinic for Dermatology, Venereology and Allergology InnsbruckMedical University of InnsbruckInnsbruckAustria
| | - Van Anh Nguyen
- University Clinic for Dermatology, Venereology and Allergology InnsbruckMedical University of InnsbruckInnsbruckAustria
| | - Heidi A Neubauer
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | | | - Olaf Merkel
- Department of PathologyMedical University of ViennaViennaAustria
| | - Marco Herling
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
- Department of Hematology, Cellular Therapy, and HemostaseologyUniversity of LeipzigLeipzigGermany
| | - Ellen Heitzer
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Patrick T Gunning
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
- Janpix, a Centessa CompanyLondonUK
| | - Lukas Kenner
- Department of PathologyMedical University of ViennaViennaAustria
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for Applied Metabolomics (CDL‐AM), Division of Nuclear MedicineMedical University of ViennaViennaAustria
- CBmed GmbH Center for Biomarker Research in MedicineGrazAustria
| | - Richard Moriggl
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Peter Wolf
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
- BioTechMed GrazGrazAustria
| |
Collapse
|
9
|
p21-Activated kinases as promising therapeutic targets in hematological malignancies. Leukemia 2022; 36:315-326. [PMID: 34697424 DOI: 10.1038/s41375-021-01451-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 01/12/2023]
Abstract
The p21-Activated Kinases (PAKs) are a family of six serine/threonine kinases that were originally identified as downstream effectors of the Rho GTPases Cdc42 and Rac. Since the first PAK was discovered in 1994, studies have revealed their fundamental and biological importance in the development of physiological systems. Within the cell, PAKs also play significant roles in regulating essential cellular processes such as cytoskeletal dynamics, gene expression, cell survival, and cell cycle progression. These processes are often deregulated in numerous cancers when different PAKs are overexpressed or amplified at the chromosomal level. Furthermore, PAKs modulate multiple oncogenic signaling pathways which facilitate apoptosis escape, uncontrolled proliferation, and drug resistance. There is growing insight into the critical roles of PAKs in regulating steady-state hematopoiesis, including the properties of hematopoietic stem cells (HSC), and the initiation and progression of hematological malignancies. This review will focus on the most recent studies that provide experimental evidence showing how specific PAKs regulate the properties of leukemic stem cells (LSCs) and drug-resistant cells to initiate and maintain hematological malignancies. The current understanding of the molecular and cellular mechanisms by which the PAKs operate in specific human leukemia or lymphomas will be discussed. From a translational point of view, PAKs have been suggested to be critical therapeutic targets and potential prognosis markers; thus, this review will also discuss current therapeutic strategies against hematological malignancies using existing small-molecule PAK inhibitors, as well as promising combination treatments, to sensitize drug-resistant cells to conventional therapies. The challenges of toxicity and non-specific targeting associated with some PAK inhibitors, as well as how future approaches for PAK inhibition to overcome these limitations, will also be addressed.
Collapse
|
10
|
Spohr C, Poggio T, Andrieux G, Schönberger K, Cabezas-Wallscheid N, Boerries M, Halbach S, Illert AL, Brummer T. Gab2 deficiency prevents Flt3-ITD driven acute myeloid leukemia in vivo. Leukemia 2022; 36:970-982. [PMID: 34903841 PMCID: PMC8979819 DOI: 10.1038/s41375-021-01490-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 11/09/2022]
Abstract
Internal tandem duplications (ITD) of the FMS-like tyrosine kinase 3 (FLT3) predict poor prognosis in acute myeloid leukemia (AML) and often co-exist with inactivating DNMT3A mutations. In vitro studies implicated Grb2-associated binder 2 (GAB2) as FLT3-ITD effector. Utilizing a Flt3-ITD knock-in, Dnmt3a haploinsufficient mouse model, we demonstrate that Gab2 is essential for the development of Flt3-ITD driven AML in vivo, as Gab2 deficient mice displayed prolonged survival, presented with attenuated liver and spleen pathology and reduced blast counts. Furthermore, leukemic bone marrow from Gab2 deficient mice exhibited reduced colony-forming unit capacity and increased FLT3 inhibitor sensitivity. Using transcriptomics, we identify the genes encoding for Axl and the Ret co-receptor Gfra2 as targets of the Flt3-ITD/Gab2/Stat5 axis. We propose a pathomechanism in which Gab2 increases signaling of these receptors by inducing their expression and by serving as downstream effector. Thereby, Gab2 promotes AML aggressiveness and drug resistance as it incorporates these receptor tyrosine kinases into the Flt3-ITD signaling network. Consequently, our data identify GAB2 as a promising biomarker and therapeutic target in human AML.
Collapse
Affiliation(s)
- Corinna Spohr
- grid.5963.9Institute of Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany ,grid.5963.9Faculty of Biology, University of Freiburg, Freiburg, Germany ,grid.5963.9Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Teresa Poggio
- grid.5963.9Faculty of Biology, University of Freiburg, Freiburg, Germany ,grid.5963.9Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Geoffroy Andrieux
- grid.5963.9Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Katharina Schönberger
- grid.5963.9Faculty of Biology, University of Freiburg, Freiburg, Germany ,grid.429509.30000 0004 0491 4256Max-Planck-Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany ,grid.4372.20000 0001 2105 1091International Max Planck Research School for Molecular and Cellular Biology (IMPRS-MCB), Freiburg, Germany
| | - Nina Cabezas-Wallscheid
- grid.429509.30000 0004 0491 4256Max-Planck-Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany ,Centre for Integrative Biological Signaling Studies (CIBSS), 79104 Freiburg, Germany
| | - Melanie Boerries
- grid.5963.9Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Comprehensive Cancer Center Freiburg (CCCF), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sebastian Halbach
- grid.5963.9Institute of Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Anna L. Illert
- grid.5963.9Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany ,grid.5963.9Comprehensive Cancer Center Freiburg (CCCF), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany. .,Comprehensive Cancer Center Freiburg (CCCF), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany. .,Center for Biological Signalling Studies BIOSS, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
11
|
FLT3-ITD transduces autonomous growth signals during its biosynthetic trafficking in acute myelogenous leukemia cells. Sci Rep 2021; 11:22678. [PMID: 34811450 PMCID: PMC8608843 DOI: 10.1038/s41598-021-02221-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) in hematopoietic cells binds to its ligand at the plasma membrane (PM), then transduces growth signals. FLT3 gene alterations that lead the kinase to assume its permanently active form, such as internal tandem duplication (ITD) and D835Y substitution, are found in 30–40% of acute myelogenous leukemia (AML) patients. Thus, drugs for molecular targeting of FLT3 mutants have been developed for the treatment of AML. Several groups have reported that compared with wild-type FLT3 (FLT3-wt), FLT3 mutants are retained in organelles, resulting in low levels of PM localization of the receptor. However, the precise subcellular localization of mutant FLT3 remains unclear, and the relationship between oncogenic signaling and the mislocalization is not completely understood. In this study, we show that in cell lines established from leukemia patients, endogenous FLT3-ITD but not FLT3-wt clearly accumulates in the perinuclear region. Our co-immunofluorescence assays demonstrate that Golgi markers are co-localized with the perinuclear region, indicating that FLT3-ITD mainly localizes to the Golgi region in AML cells. FLT3-ITD biosynthetically traffics to the Golgi apparatus and remains there in a manner dependent on its tyrosine kinase activity. Tyrosine kinase inhibitors, such as quizartinib (AC220) and midostaurin (PKC412), markedly decrease FLT3-ITD retention and increase PM levels of the mutant. FLT3-ITD activates downstream in the endoplasmic reticulum (ER) and the Golgi apparatus during its biosynthetic trafficking. Results of our trafficking inhibitor treatment assays show that FLT3-ITD in the ER activates STAT5, whereas that in the Golgi can cause the activation of AKT and ERK. We provide evidence that FLT3-ITD signals from the early secretory compartments before reaching the PM in AML cells.
Collapse
|
12
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
13
|
Cho H, Shin I, Yoon H, Jeon E, Lee J, Kim Y, Ryu S, Song C, Kwon NH, Moon Y, Kim S, Kim ND, Choi HG, Sim T. Identification of Thieno[3,2- d]pyrimidine Derivatives as Dual Inhibitors of Focal Adhesion Kinase and FMS-like Tyrosine Kinase 3. J Med Chem 2021; 64:11934-11957. [PMID: 34324343 DOI: 10.1021/acs.jmedchem.1c00459] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Focal adhesion kinase (FAK) is overexpressed in highly invasive and metastatic cancers. To identify novel FAK inhibitors, we designed and synthesized various thieno[3,2-d]pyrimidine derivatives. An intensive structure-activity relationship (SAR) study led to the identification of 26 as a lead. Moreover, 26, a multitargeted kinase inhibitor, possesses excellent potencies against FLT3 mutants as well as FAK. Gratifyingly, 26 remarkably inhibits recalcitrant FLT3 mutants, including F691L, that cause drug resistance. Importantly, 26 is superior to PF-562271 in terms of apoptosis induction, anchorage-independent growth inhibition, and tumor burden reduction in the MDA-MB-231 xenograft mouse model. Also, 26 causes regression of tumor growth in the MV4-11 xenograft mouse model, indicating that it could be effective against acute myeloid leukemia (AML). Finally, in an orthotopic mouse model using MDA-MB-231, 26 remarkably prevents metastasis of orthotopic tumors to lymph nodes. Taken together, the results indicate that 26 possesses potential therapeutic value against highly invasive cancers and relapsed AML.
Collapse
Affiliation(s)
- Hanna Cho
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Injae Shin
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hojong Yoon
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Eunhye Jeon
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jiwon Lee
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Younghoon Kim
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - SeongShick Ryu
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Chiman Song
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Nam Hoon Kwon
- Medicinal Bioconvergence Research Center, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Institute for Artificial Intelligence and Biomedical Research, College of Pharmacy & College of Medicine, Gangnam Severance Hospital, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Youngji Moon
- Medicinal Bioconvergence Research Center, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Institute for Artificial Intelligence and Biomedical Research, College of Pharmacy & College of Medicine, Gangnam Severance Hospital, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Nam Doo Kim
- Voronoibio Inc., 32 Songdogwahak-ro, Yeonsu-gu, Incheon 21984, Republic of Korea
| | - Hwan Geun Choi
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- B2Sbio Inc., 32 Songdogwahak-ro, Yeonsu-gu, Incheon 21984, Republic of Korea
| | - Taebo Sim
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
14
|
Sudha T, Godugu K, Darwish NHE, Nazeer T, Mousa SA. Novel Polyethylene Glycol-Conjugated Triazole Derivative with High Thyrointegrin αvβ3 Affinity in Acute Myeloid Leukemia Management. Cancers (Basel) 2021; 13:cancers13164070. [PMID: 34439224 PMCID: PMC8392871 DOI: 10.3390/cancers13164070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023] Open
Abstract
(1) Background: Acute myeloid leukemia (AML) accounts for up to one-third of more than 60,000 leukemia cases diagnosed annually in the U.S. Primary AML cells express membrane αvβ3 integrin, which is associated with adverse prognosis and resistance to chemotherapies. A novel anticancer compound Polyethylene glycol-conjugated bi-TriAzole Tetraiodothyroacetic acid (P-bi-TAT) interacts with high affinity (Ki 0.3 nM) and specificity with the thyrointegrin αvβ3. We evaluated P-bi-TAT activities in two different AML models representing monocytic and myelocytic forms of acute leukemia. (2) Methods and Results: The in vivo AML models were established prior to initiation of treatment protocols by grafting human leukemia cells in immunocompromised mice. IVIS imaging scans revealed that leukemic colonies were extensively established throughout the bone marrow, liver, and lung of the untreated animals. In animals treated with P-bi-TAT at daily doses ranging from 1-10 mg/kg, subcutaneously for 2-3 weeks, IVIS imaging scans revealed 95% reduction in bone marrow colonies and leukemic colonies in liver and lung. Also, the leukemic cells were not detected in bone marrow samples of P-bi-TAT-treated animals. The anti-neoplastic effect of P-bi-TAT administration on leukemic cells was associated with marked inhibition of NF-κB activity. We conclude that experimental P-bi-TAT therapy in vivo appears extraordinarily effective against the two forms of human AML models in mice. Because the P-bi-TAT molecular target, thyrointegrin αvβ3, is consistently expressed in many, if not all, clinical AML samples, P-bi-TAT-based therapy seems to have significant clinical potential in treating most AML sub-types. Hence, P-bi-TAT represents a promising targeted therapeutic agent for AML patients.
Collapse
Affiliation(s)
- Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
| | - Noureldien H. E. Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Tipu Nazeer
- Albany Medical Center, Pathology Department, AMC Hospital, Albany, NY 12208, USA;
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
- Correspondence:
| |
Collapse
|
15
|
Actin cytoskeleton deregulation confers midostaurin resistance in FLT3-mutant acute myeloid leukemia. Commun Biol 2021; 4:799. [PMID: 34172833 PMCID: PMC8233337 DOI: 10.1038/s42003-021-02215-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
The presence of FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) is one of the most frequent mutations in acute myeloid leukemia (AML) and is associated with an unfavorable prognosis. FLT3 inhibitors, such as midostaurin, are used clinically but fail to entirely eradicate FLT3-ITD + AML. This study introduces a new perspective and highlights the impact of RAC1-dependent actin cytoskeleton remodeling on resistance to midostaurin in AML. RAC1 hyperactivation leads resistance via hyperphosphorylation of the positive regulator of actin polymerization N-WASP and antiapoptotic BCL-2. RAC1/N-WASP, through ARP2/3 complex activation, increases the number of actin filaments, cell stiffness and adhesion forces to mesenchymal stromal cells (MSCs) being identified as a biomarker of resistance. Midostaurin resistance can be overcome by a combination of midostaruin, the BCL-2 inhibitor venetoclax and the RAC1 inhibitor Eht1864 in midostaurin-resistant AML cell lines and primary samples, providing the first evidence of a potential new treatment approach to eradicate FLT3-ITD + AML. Garitano-Trojaola et al. used a combination of human acute myeloid leukemia (AML) cell lines and primary samples to show that RAC1-dependent actin cytoskeleton remodeling through BCL2 family plays a key role in resistance to the FLT3 inhibitor, Midostaurin in AML. They showed that by targeting RAC1 and BCL2, Midostaurin resistance was diminished, which potentially paves the way for an innovate treatment approach for FLT3 mutant AML.
Collapse
|
16
|
Synergistic targeting of FLT3 mutations in AML via combined menin-MLL and FLT3 inhibition. Blood 2021; 136:2442-2456. [PMID: 32589720 DOI: 10.1182/blood.2020005037] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022] Open
Abstract
The interaction of menin (MEN1) and MLL (MLL1, KMT2A) is a dependency and provides a potential opportunity for treatment of NPM1-mutant (NPM1mut) and MLL-rearranged (MLL-r) leukemias. Concomitant activating driver mutations in the gene encoding the tyrosine kinase FLT3 occur in both leukemias and are particularly common in the NPM1mut subtype. In this study, transcriptional profiling after pharmacological inhibition of the menin-MLL complex revealed specific changes in gene expression, with downregulation of the MEIS1 transcription factor and its transcriptional target gene FLT3 being the most pronounced. Combining menin-MLL inhibition with specific small-molecule kinase inhibitors of FLT3 phosphorylation resulted in a significantly superior reduction of phosphorylated FLT3 and transcriptional suppression of genes downstream of FLT3 signaling. The drug combination induced synergistic inhibition of proliferation, as well as enhanced apoptosis, compared with single-drug treatment in models of human and murine NPM1mut and MLL-r leukemias harboring an FLT3 mutation. Primary acute myeloid leukemia (AML) cells harvested from patients with NPM1mutFLT3mut AML showed significantly better responses to combined menin and FLT3 inhibition than to single-drug or vehicle control treatment, whereas AML cells with wild-type NPM1, MLL, and FLT3 were not affected by either of the 2 drugs. In vivo treatment of leukemic animals with MLL-r FLT3mut leukemia reduced leukemia burden significantly and prolonged survival compared with results in the single-drug and vehicle control groups. Our data suggest that combined menin-MLL and FLT3 inhibition represents a novel and promising therapeutic strategy for patients with NPM1mut or MLL-r leukemia and concurrent FLT3 mutation.
Collapse
|
17
|
Liu H, Liu K, Dong Z. The Role of p21-Activated Kinases in Cancer and Beyond: Where Are We Heading? Front Cell Dev Biol 2021; 9:641381. [PMID: 33796531 PMCID: PMC8007885 DOI: 10.3389/fcell.2021.641381] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/03/2021] [Indexed: 12/12/2022] Open
Abstract
The p21-activated kinases (PAKs), downstream effectors of Ras-related Rho GTPase Cdc42 and Rac, are serine/threonine kinases. Biologically, PAKs participate in various cellular processes, including growth, apoptosis, mitosis, immune response, motility, inflammation, and gene expression, making PAKs the nexus of several pathogenic and oncogenic signaling pathways. PAKs were proved to play critical roles in human diseases, including cancer, infectious diseases, neurological disorders, diabetes, pancreatic acinar diseases, and cardiac disorders. In this review, we systematically discuss the structure, function, alteration, and molecular mechanisms of PAKs that are involved in the pathogenic and oncogenic effects, as well as PAK inhibitors, which may be developed and deployed in cancer therapy, anti-viral infection, and other diseases. Furthermore, we highlight the critical questions of PAKs in future research, which provide an opportunity to offer input and guidance on new directions for PAKs in pathogenic, oncogenic, and drug discovery research.
Collapse
Affiliation(s)
- Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| |
Collapse
|
18
|
Kuželová K, Obr A, Röselová P, Grebeňová D, Otevřelová P, Brodská B, Holoubek A. Group I p21-activated kinases in leukemia cell adhesion to fibronectin. Cell Adh Migr 2021; 15:18-36. [PMID: 33464167 PMCID: PMC7834095 DOI: 10.1080/19336918.2021.1872760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
P21-activated kinases (PAK) regulate processes associated with cytoskeleton dynamics. PAK expression in leukemia cells was measured on protein and mRNA levels. In functional assays, we analyzed the effect of PAK inhibitors IPA-3 and FRAX597 on cell adhesivity and viability. PAK2 was dominant in cell lines, whereas primary cells also expressed comparable amount of PAK1 transcription isoforms: PAK1-full and PAK1Δ15. PAK1Δ15 and PAK2 levels correlated with surface density of integrins β1 and αVβ3. PAK1-full, but not PAK2, was present in membrane protrusions. IPA-3, which prevents PAK activation, induced cell contraction in semi-adherent HEL cells only. FRAX597, which inhibits PAK kinase activity, increased cell-surface contact area in all leukemia cells. Both inhibitors reduced the stability of cell attachment and induced cell death.
Collapse
Affiliation(s)
- Kateřina Kuželová
- Department of Proteomics, Institute of Hematology and Blood Transfusion , Prague, Czech Republic
| | - Adam Obr
- Department of Proteomics, Institute of Hematology and Blood Transfusion , Prague, Czech Republic
| | - Pavla Röselová
- Department of Proteomics, Institute of Hematology and Blood Transfusion , Prague, Czech Republic
| | - Dana Grebeňová
- Department of Proteomics, Institute of Hematology and Blood Transfusion , Prague, Czech Republic
| | - Petra Otevřelová
- Department of Proteomics, Institute of Hematology and Blood Transfusion , Prague, Czech Republic
| | - Barbora Brodská
- Department of Proteomics, Institute of Hematology and Blood Transfusion , Prague, Czech Republic
| | - Aleš Holoubek
- Department of Proteomics, Institute of Hematology and Blood Transfusion , Prague, Czech Republic
| |
Collapse
|
19
|
Coordinated dysregulation of cancer progression by the HER family and p21-activated kinases. Cancer Metastasis Rev 2020; 39:583-601. [PMID: 32820388 DOI: 10.1007/s10555-020-09922-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/20/2022]
Abstract
Most epithelial cancer types are polygenic in nature and are driven by coordinated dysregulation of multiple regulatory pathways, genes, and protein modifications. The process of coordinated regulation of cancer promoting pathways in response to extrinsic and intrinsic signals facilitates the dysregulation of several pathways with complementary functions, contributing to the hallmarks of cancer. Dysregulation and hyperactivation of cell surface human epidermal growth factor receptors (HERs) and cytoskeleton remodeling by p21-activated kinases (PAKs) are two prominent interconnected aspects of oncogenesis. We briefly discuss the discoveries and significant advances in the area of coordinated regulation of HERs and PAKs in the development and progression of breast and other epithelial cancers. We also discuss how initial studies involving heregulin signaling via HER3-HER2 axis and HER2-overexpressing breast cancer cells not only discovered a mechanistic role of PAK1 in breast cancer pathobiology but also acted as a bridge in generating a broader cancer research interest in other PAK family members and cancer types and catalyzed establishing the role of PAKs in human cancer, at-large. In addition, growth factor stimulation of the PAK pathway also helped to recognize new facets of PAKs, connecting the PAK pathway to oncogenesis, nuclear signaling, gene expression, mitotic progression, DNA damage response, among other phenotypic responses, and shaped the field of PAK cancer research. Finally, we recount some of the current limitations of HER- and PAK-directed therapeutics in counteracting acquired therapeutic resistance and discuss how cancer's as a polygenic disease may be best targeted with a polygenic approach.
Collapse
|
20
|
Napolitano R, De Matteis S, Carloni S, Bruno S, Abbati G, Capelli L, Ghetti M, Bochicchio MT, Liverani C, Mercatali L, Calistri D, Cuneo A, Menon K, Musuraca G, Martinelli G, Simonetti G. Kevetrin induces apoptosis in TP53 wild‑type and mutant acute myeloid leukemia cells. Oncol Rep 2020; 44:1561-1573. [PMID: 32945487 PMCID: PMC7448420 DOI: 10.3892/or.2020.7730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor protein p53 is a key regulator of several cellular pathways, including DNA repair, cell cycle and angiogenesis. Kevetrin exhibits p53-dependent as well as-independent activity in solid tumors, while its effects on leukemic cells remain unknown. The aim of the present study was to analyze the response of acute myeloid leukemia (AML) cell lines (TP53 wild-type: OCI-AML3 and MOLM-13; and TP53-mutant: KASUMI-1 and NOMO-1) to kevetrin at a concentration range of 85–340 µM. The cellular and molecular effects of the treatment were analyzed in terms of cell growth, viability [Annexin V-propidium iodide (PI) staining] and cell cycle alterations (PI staining). Gene expression profiling, western blotting and immunofluorescence were performed to elucidate the pathways underlying kevetrin activity. Pulsed exposure exerted no effect on the wild-type cells, but was effective on mutant cells. After continuous treatment, significant cell growth arrest and apoptosis were observed in all cell lines, with TP53-mutant models displaying a higher sensitivity and p53 induction. Kevetrin also displayed efficacy against TP53 wild-type and mutant primary AML, with a preferential cytotoxic activity against blast cells. Gene expression profiling revealed a common core transcriptional program altered by drug exposure and the downregulation of glycolysis, DNA repair and unfolded protein response signatures. These findings suggest that kevetrin may be a promising therapeutic option for patients with both wild-type and TP53-mutant AML.
Collapse
Affiliation(s)
- Roberta Napolitano
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Serena De Matteis
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Silvia Carloni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Samantha Bruno
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna and Institute of Hematology 'L. e A. Seràgnoli', I‑40138 Bologna, Italy
| | - Giulia Abbati
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Laura Capelli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Martina Ghetti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Chiara Liverani
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Laura Mercatali
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Antonio Cuneo
- Department of Medical Sciences, University of Ferrara‑Arcispedale Sant'Anna, I‑44124 Ferrara, Italy
| | | | - Gerardo Musuraca
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Giovanni Martinelli
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna and Institute of Hematology 'L. e A. Seràgnoli', I‑40138 Bologna, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| |
Collapse
|
21
|
Wang X, Mak PY, Mu H, Tao W, Rao A, Visweswaran R, Ruvolo V, Pachter JA, Weaver DT, Andreeff M, Xu B, Carter BZ. Combinatorial Inhibition of Focal Adhesion Kinase and BCL-2 Enhances Antileukemia Activity of Venetoclax in Acute Myeloid Leukemia. Mol Cancer Ther 2020; 19:1636-1648. [PMID: 32404407 PMCID: PMC7416436 DOI: 10.1158/1535-7163.mct-19-0841] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/05/2019] [Accepted: 05/07/2020] [Indexed: 01/07/2023]
Abstract
Focal adhesion kinase (FAK) promotes cancer cell growth and metastasis. We previously reported that FAK inhibition by the selective inhibitor VS-4718 exerted antileukemia activities in acute myeloid leukemia (AML). The mechanisms involved, and whether VS-4718 potentiates efficacy of other therapeutic agents, have not been investigated. Resistance to apoptosis inducted by the BCL-2 inhibitor ABT-199 (venetoclax) in AML is mediated by preexisting and ABT-199-induced overexpression of MCL-1 and BCL-XL. We observed that VS-4718 or silencing FAK with siRNA decreased MCL-1 and BCL-XL levels. Importantly, VS-4718 antagonized ABT-199-induced MCL-1 and BCL-XL. VS-4718 markedly synergized with ABT-199 to induce apoptosis in AML cells, including primary AML CD34+ cells and AML cells overexpressing MCL-1 or BCL-XL. In a patient-derived xenograft (PDX) model derived from a patient sample with NPM1/FLT3-ITD/TET2/DNMT3A/WT1 mutations and complex karyotype, VS-4718 statistically significantly reduced leukemia tissue infiltration and extended survival (72 vs. control 36 days, P = 0.0002), and only its combination with ABT-199 effectively decreased systemic leukemia tissue infiltration and circulating blasts, and prolonged survival (65.5 vs. control 36 days, P = 0.0119). Furthermore, the combination decreased NFκB signaling and induced the expression of IFN genes in vivo The combination also markedly extended survival of a second PDX model developed from an aggressive, TP53-mutated complex karyotype AML sample. The data suggest that the combined inhibition of FAK and BCL-2 enhances antileukemia activity in AML at least in part by suppressing MCL-1 and BCL-XL and that this combination may be effective in AML with TP53 and other mutations, and thus benefit patients with high-risk AML.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis
- Biomarkers, Tumor/antagonists & inhibitors
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Proliferation
- Focal Adhesion Kinase 1/antagonists & inhibitors
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Nucleophosmin
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Sulfonamides/pharmacology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xiangmeng Wang
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Po Yee Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenjing Tao
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arvind Rao
- The Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ravikumar Visweswaran
- The Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivian Ruvolo
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Bing Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China.
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, P.R. China
| | - Bing Z Carter
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
22
|
Yao D, Li C, Rajoka MSR, He Z, Huang J, Wang J, Zhang J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Am J Cancer Res 2020; 10:9741-9766. [PMID: 32863957 PMCID: PMC7449905 DOI: 10.7150/thno.46913] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
The p21-Activated kinase 1 (PAK1), a member of serine-threonine kinases family, was initially identified as an interactor of the Rho GTPases RAC1 and CDC42, which affect a wide range of processes associated with cell motility, survival, metabolism, cell cycle, proliferation, transformation, stress, inflammation, and gene expression. Recently, the PAK1 has emerged as a potential therapeutic target in cancer due to its role in many oncogenic signaling pathways. Many PAK1 inhibitors have been developed as potential preclinical agents for cancer therapy. Here, we provide an overview of essential roles that PAK1 plays in cancer, including its structure and autoactivation mechanism, its crucial function from onset to progression to metastasis, metabolism, immune escape and even drug resistance in cancer; endogenous regulators; and cancer-related pathways. We also summarize the reported PAK1 small-molecule inhibitors based on their structure types and their potential application in cancer. In addition, we provide overviews on current progress and future challenges of PAK1 in cancer, hoping to provide new ideas for the diagnosis and treatment of cancer.
Collapse
|
23
|
Kuželová K, Obr A, Marková J, Gašová Z. Integrin expression and adhesivity to fibronectin in primary acute myeloid leukemia cells: Impact of NPM1 and FLT3 mutations. Eur J Haematol 2020; 105:578-587. [PMID: 32668024 DOI: 10.1111/ejh.13488] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Interaction of leukemia cells with the bone marrow extracellular matrix promotes cell survival and resistance to chemotherapy. In this work, we analyzed integrin expression and adhesivity to fibronectin in primary cells from patients with acute myeloid leukemia. METHODS Surface expression of integrins β1 and αVβ3 on primary leukemia cells (N = 46) was correlated with the stem cell marker CD34, as well as with cell adhesivity to fibronectin. The results were analyzed with regard to the mutational status of NPM1 and FLT3 genes. RESULTS The integrin β1 was omnipresent, whereas αVβ3 was often more expressed on CD34-positive cells. In particular, higher αVβ3 expression on CD34+ cells was associated with NPM1 mutation (P = .0018). Monocytic leukemias had significantly higher αVβ3 expression compared to less maturated cases (P = .0008). Cells from patients with internal tandem duplications in FLT3 (FLT3-ITD) had lower adhesivity to fibronectin compared to cells with wild-type FLT3 (P = .031), specifically in less differentiated myeloblasts. Inhibition of a putative FLT3-ITD target, EZH2, increased cell adhesivity in MV4-11 cell line (P = .024). CONCLUSIONS The integrin αVβ3 is expressed in particular on CD34+ cells with NPM1 mutation and might have a prognostic value in patients with mutated NPM1. FLT3-ITD is associated with lower cell adhesivity, especially in patients with less differentiated leukemias.
Collapse
Affiliation(s)
- Kateřina Kuželová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Adam Obr
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Jana Marková
- Clinical Department, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Zdenka Gašová
- Department of Apheresis, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| |
Collapse
|
24
|
Zhu XY, Guo QY, Zhu M, Chen BG, Wang LY, Zhang DQ, Zhang L, Shao YP, Luo WD. HLX affects cell cycle and proliferation in AML cells via the JAK/STAT signaling pathway. Oncol Lett 2020; 20:1888-1896. [PMID: 32724432 PMCID: PMC7377103 DOI: 10.3892/ol.2020.11718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/13/2020] [Indexed: 01/11/2023] Open
Abstract
Acute myelogenous leukemia (AML) is a class of malignant tumors derived from hematopoietic stem or progenitor cells. The H2.0-like homeobox gene (HLX) encodes transcription factors that function in promoting normal hematopoietic cell proliferation and tumor immunity. The present study analyzed the effect of downregulating the HLX on cell cycle distribution and cell proliferation in AML. Moreover, the current study detected changes in the expression of genes and proteins in the Janus kinase (JAK)/STAT signaling pathway to investigate the mechanism of the action of HLX in tumor immunity in AML. HLX expression in AML cell lines was silenced using small interfering siRNA, and MTS/PMS-assay colorimetric assays were used to assess the effect of knockdown of HLX on AML cell proliferation. Flow cytometry was used to analyze changes in cell cycle distribution, while reverse transcription-quantitative PCR and western blotting were used to detect changes in the expression levels of key components of the JAK/STAT signaling pathway, such as p21-activated kinase 1 (PAK1), neuropilin 1 (NRP1), B-cell translocation gene 1 (BTG1) and STAT5. It was found that HLX was differentially expressed in AML cell lines of various subtypes, and HLX expression was higher in the AML/M3 subtype NB4 cell line compared with the control group. Knockdown of HLX in NB4 cells significantly inhibited cell proliferation and arrested cells in the G0/G1 phase. Moreover, STAT5 protein expression, as well as NRP1 and PAK1 expression levels were downregulated, while BTG1 expression was upregulated when HLX was knocked out by siRNA. Collectively, the results suggested that downregulation of HLX may cause G0/G1 phase arrest and inhibit the proliferation of AML cells by activating the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Xia-Yin Zhu
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Qun-Yi Guo
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Min Zhu
- Department of Central Laboratory, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Bao-Guo Chen
- Department of Central Laboratory, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Ling-Yan Wang
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Dan-Qiong Zhang
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Li Zhang
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Yan-Ping Shao
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Wen-Da Luo
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| |
Collapse
|
25
|
Martelli M, Monaldi C, De Santis S, Bruno S, Mancini M, Cavo M, Soverini S. Recent Advances in the Molecular Biology of Systemic Mastocytosis: Implications for Diagnosis, Prognosis, and Therapy. Int J Mol Sci 2020; 21:E3987. [PMID: 32498255 PMCID: PMC7312790 DOI: 10.3390/ijms21113987] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/28/2020] [Accepted: 05/30/2020] [Indexed: 12/20/2022] Open
Abstract
In recent years, molecular characterization and management of patients with systemic mastocytosis (SM) have greatly benefited from the application of advanced technologies. Highly sensitive and accurate assays for KIT D816V mutation detection and quantification have allowed the switch to non-invasive peripheral blood testing for patient screening; allele burden has prognostic implications and may be used to monitor therapeutic efficacy. Progress in genetic profiling of KIT, together with the use of next-generation sequencing panels for the characterization of associated gene mutations, have allowed the stratification of patients into three subgroups differing in terms of pathogenesis and prognosis: i) patients with mast cell-restricted KIT D816V; ii) patients with multilineage KIT D816V-involvement; iii) patients with "multi-mutated disease". Thanks to these findings, new prognostic scoring systems combining clinical and molecular data have been developed. Finally, non-genetic SETD2 histone methyltransferase loss of function has recently been identified in advanced SM. Assessment of SETD2 protein levels and activity might provide prognostic information and has opened new research avenues exploring alternative targeted therapeutic strategies. This review discusses how progress in recent years has rapidly complemented previous knowledge improving the molecular characterization of SM, and how this has the potential to impact on patient diagnosis and management.
Collapse
Affiliation(s)
- Margherita Martelli
- Department of Experimental, Diagnostic and Specialty Medicine, Hematology/Oncology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy; (C.M.); (S.D.S.); (S.B.); (M.M.); (M.C.); (S.S.)
| | | | | | | | | | | | | |
Collapse
|
26
|
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: 52] [Impact Index Per Article: 10.4] [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
|
27
|
Pandey R, Kapur R. DNA damage on the DOCK in FLT3-ITD-driven acute myeloid leukemia. Haematologica 2019; 104:2330-2332. [PMID: 31787614 DOI: 10.3324/haematol.2019.231340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Ruchi Pandey
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, IN, USA
| | - Reuben Kapur
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, IN, USA
| |
Collapse
|
28
|
Ma G, Gezer D, Herrmann O, Feldberg K, Schemionek M, Jawhar M, Reiter A, Brümmendorf TH, Koschmieder S, Chatain N. LCP1 triggers mTORC2/AKT activity and is pharmacologically targeted by enzastaurin in hypereosinophilia. Mol Carcinog 2019; 59:87-103. [DOI: 10.1002/mc.23131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Guangxin Ma
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
- Hematology and Oncology Unit, Department of Geriatrics Qilu Hospital of Shandong University Jinan Shandong China
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Oliver Herrmann
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Kristina Feldberg
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Mirle Schemionek
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Mohamad Jawhar
- Department of Hematology and Oncology University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | - Andreas Reiter
- Department of Hematology and Oncology University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | - Tim H. Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine RWTH Aachen University Aachen Germany
| |
Collapse
|
29
|
Combined inhibition of receptor tyrosine and p21-activated kinases as a therapeutic strategy in childhood ALL. Blood Adv 2019; 2:2554-2567. [PMID: 30301811 DOI: 10.1182/bloodadvances.2018020693] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/02/2018] [Indexed: 11/20/2022] Open
Abstract
Receptor tyrosine kinase (RTK)-dependent signaling has been implicated in the pathogenesis of acute lymphoblastic leukemia (ALL) of childhood. However, the RTK-dependent signaling state and its interpretation with regard to biological behavior are often elusive. To decipher signaling circuits that link RTK activity with biological output in vivo, we established patient-derived xenograft ALL (PDX-ALL) models with dependencies on fms-like tyrosine kinase 3 (FLT3) and platelet-derived growth factor receptor β (PDGFRB), which were interrogated by phosphoproteomics using iTRAQ mass spectrometry. Signaling circuits were determined by receptor type and cellular context with few generic features, among which we identified group I p21-activated kinases (PAKs) as potential therapeutic targets. Growth factor stimulation markedly increased catalytic activities of PAK1 and PAK2. RNA interference (RNAi)-mediated or pharmacological inhibition of PAKs using allosteric or adenosine triphosphate (ATP)-competitive compounds attenuated cell growth and increased apoptosis in vitro. Notably, PAK1- or PAK2-directed RNAi enhanced the antiproliferative effects of the type III RTK and protein kinase C inhibitor midostaurin. Treatment of FLT3- or PDGFRB-dependent ALLs with ATP-competitive PAK inhibitors markedly decreased catalytic activities of both PAK isoforms. In FLT3-driven ALL, this effect was augmented by coadministration of midostaurin resulting in synergistic effects on growth inhibition and apoptosis. Finally, combined treatment of FLT3 D835H PDX-ALL with the ATP-competitive group I PAK inhibitor FRAX486 and midostaurin in vivo significantly prolonged leukemia progression-free survival compared with midostaurin monotherapy or control. Our study establishes PAKs as potential downstream targets in RTK-dependent ALL of childhood, the inhibition of which might help prevent the selection or acquisition of resistance mutations toward tyrosine kinase inhibitors.
Collapse
|
30
|
Ziyad S, Riordan JD, Cavanaugh AM, Su T, Hernandez GE, Hilfenhaus G, Morselli M, Huynh K, Wang K, Chen JN, Dupuy AJ, Iruela-Arispe ML. A Forward Genetic Screen Targeting the Endothelium Reveals a Regulatory Role for the Lipid Kinase Pi4ka in Myelo- and Erythropoiesis. Cell Rep 2019; 22:1211-1224. [PMID: 29386109 PMCID: PMC5828030 DOI: 10.1016/j.celrep.2018.01.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/05/2017] [Accepted: 01/05/2018] [Indexed: 11/19/2022] Open
Abstract
Given its role as the source of definitive hematopoietic cells, we sought to determine whether mutations initiated in the hemogenic endothelium would yield hematopoietic abnormalities or malignancies. Here, we find that endothelium-specific transposon mutagenesis in mice promotes hematopoietic pathologies that are both myeloid and lymphoid in nature. Frequently mutated genes included previously recognized cancer drivers and additional candidates, such as Pi4ka, a lipid kinase whose mutation was found to promote myeloid and erythroid dysfunction. Subsequent validation experiments showed that targeted inactivation of the Pi4ka catalytic domain or reduction in mRNA expression inhibited myeloid and erythroid cell differentiation in vitro and promoted anemia in vivo through a mechanism involving deregulation of AKT, MAPK, SRC, and JAK-STAT signaling. Finally, we provide evidence linking PI4KAP2, previously considered a pseudogene, to human myeloid and erythroid leukemia.
Collapse
Affiliation(s)
- Safiyyah Ziyad
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jesse D Riordan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Ann M Cavanaugh
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Trent Su
- Institute for Quantitative and Computational Biology and Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gloria E Hernandez
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Georg Hilfenhaus
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marco Morselli
- Institute for Quantitative and Computational Biology and Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute of Genomics and Proteomics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kristine Huynh
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kevin Wang
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jau-Nian Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adam J Dupuy
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
31
|
Chung EY, Mai Y, Shah UA, Wei Y, Ishida E, Kataoka K, Ren X, Pradhan K, Bartholdy B, Wei X, Zou Y, Zhang J, Ogawa S, Steidl U, Zang X, Verma A, Janakiram M, Ye BH. PAK Kinase Inhibition Has Therapeutic Activity in Novel Preclinical Models of Adult T-Cell Leukemia/Lymphoma. Clin Cancer Res 2019; 25:3589-3601. [PMID: 30862694 DOI: 10.1158/1078-0432.ccr-18-3033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/11/2019] [Accepted: 03/06/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE To evaluate therapeutic activity of PAK inhibition in ATLL and to characterize the role of PAK isoforms in cell proliferation, survival, and adhesion of ATLL cells in preclinical models. EXPERIMENTAL DESIGN Frequency and prognostic impact of PAK2 amplification were evaluated in an ATLL cohort of 370 cases. Novel long-term cultures and in vivo xenograft models were developed using primary ATLL cells from North American patients. Two PAK inhibitors were used to block PAK kinase activity pharmacologically. siRNA-based gene silencing approach was used to genetically knockdown (KD) PAK1 and PAK2 in ATLL cell lines. RESULTS PAK1/2/4 are the three most abundantly expressed PAK family members in ATLL. PAK2 amplifications are seen in 24% of ATLLs and are associated with worse prognosis in a large patient cohort. The pan-PAK inhibitor PF-3758309 (PF) has strong in vitro and in vivo activity in a variety of ATLL preclinical models. These activities of PF are likely attributed to its ability to target several PAK isoforms simultaneously because genetic silencing of either PAK1 or PAK2 produced more modest effects. PAK2 plays a major role in CADM1-mediated stromal interaction, which is an important step in systemic dissemination of the disease. This finding is consistent with the observation that PAK2 amplification is more frequent in aggressive ATLLs and correlates with inferior outcome. CONCLUSIONS PAK2, a gene frequently amplified in ATLL, facilitates CADM1-mediated stromal interaction and promotes survival of ATLL cells. Taken together, PAK inhibition may hold significant promise as a targeted therapy for aggressive ATLLs.
Collapse
Affiliation(s)
- Elaine Y Chung
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Yun Mai
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Urvi A Shah
- Department of Oncology, Montefiore Medical Center, Bronx, New York
| | - Yongqiang Wei
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York.,Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Elise Ishida
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Xiaoxin Ren
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Kith Pradhan
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Boris Bartholdy
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Xiaolei Wei
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York.,Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyu Zou
- Department of Oncology, Montefiore Medical Center, Bronx, New York
| | - Jinghang Zhang
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Xingxing Zang
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Amit Verma
- Department of Oncology, Montefiore Medical Center, Bronx, New York.,Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Murali Janakiram
- Department of Oncology, Montefiore Medical Center, Bronx, New York
| | - B Hilda Ye
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
32
|
Yuanxin Y, Yanhong Z, Qin Z, Sishi T, Yang D, Yi Z, Minjin W, Juan Z, Xiaojun L, Lanlan W, Binwu Y. Pak1 gene functioned differentially in different BCR-ABL subtypes in leukemiagenesis and treatment response through STAT5 pathway. Leuk Res 2019; 79:6-16. [PMID: 30784762 DOI: 10.1016/j.leukres.2019.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 02/05/2023]
Abstract
The BCR-ABL fusion gene (BCR-ABL) has different subtypes such as p210 and p190 with p190 appear to lead to a worse prognosis. To explore the mechanism of difference in pathogenesis and prognosis in different BCR-ABL subtype-related leukemia, expression profile microarray analysis was conducted between p190 and p210 patients and verified by RT-PCR. The p21-activated kinase (PAK1) gene was chosen and regulation of the PAK1-STAT5 biological axis and its influence on proliferation and apoptosis in leukemia cells were also analyzed. The results showed that PAK1 might be an important molecular mechanism of the pathogenic difference between different BCR-ABL subtypes. In P210 (+) chronic myelogenous leukemia (CML), down-regulated PAK1 gene expressions may lead to the suppression of cell proliferation and promotion of apoptosis through phosphorylation of STAT5, with a reverse effect in P190 (+) acute lymphoblastic leukemia(ALL), especially acute B lymphoblastic leukemia (B-ALL). Additionally, in P210 (+) CML, down-regulated PAK1 expression may enhance the effect of TKI, whereas the reverse is true in P190 (+) B-ALL, demonstrating that PAK1 might also be an important therapeutic target between different BCR-ABL subtypes.
Collapse
Affiliation(s)
- Ye Yuanxin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Zhou Yanhong
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Zheng Qin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Tang Sishi
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Dai Yang
- Department of hematology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Zhou Yi
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Wang Minjin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Zhou Juan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Lu Xiaojun
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Wang Lanlan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| | - Ying Binwu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, People's Republic of China.
| |
Collapse
|
33
|
Durand-Onaylı V, Haslauer T, Härzschel A, Hartmann TN. Rac GTPases in Hematological Malignancies. Int J Mol Sci 2018; 19:ijms19124041. [PMID: 30558116 PMCID: PMC6321480 DOI: 10.3390/ijms19124041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence suggests that crosstalk between hematologic tumor cells and the tumor microenvironment contributes to leukemia and lymphoma cell migration, survival, and proliferation. The supportive tumor cell-microenvironment interactions and the resulting cellular processes require adaptations and modulations of the cytoskeleton. The Rac subfamily of the Rho family GTPases includes key regulators of the cytoskeleton, with essential functions in both normal and transformed leukocytes. Rac proteins function downstream of receptor tyrosine kinases, chemokine receptors, and integrins, orchestrating a multitude of signals arising from the microenvironment. As such, it is not surprising that deregulation of Rac expression and activation plays a role in the development and progression of hematological malignancies. In this review, we will give an overview of the specific contribution of the deregulation of Rac GTPases in hematologic malignancies.
Collapse
Affiliation(s)
- Valerie Durand-Onaylı
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Theresa Haslauer
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Andrea Härzschel
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Tanja Nicole Hartmann
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Disease, Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
34
|
Pallarès V, Hoyos M, Chillón MC, Barragán E, Prieto Conde MI, Llop M, Falgàs A, Céspedes MV, Montesinos P, Nomdedeu JF, Brunet S, Sanz MÁ, González-Díaz M, Sierra J, Mangues R, Casanova I. Focal Adhesion Genes Refine the Intermediate-Risk Cytogenetic Classification of Acute Myeloid Leukemia. Cancers (Basel) 2018; 10:cancers10110436. [PMID: 30428571 PMCID: PMC6265715 DOI: 10.3390/cancers10110436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/05/2018] [Accepted: 11/10/2018] [Indexed: 02/04/2023] Open
Abstract
In recent years, several attempts have been made to identify novel prognostic markers in patients with intermediate-risk acute myeloid leukemia (IR-AML), to implement risk-adapted strategies. The non-receptor tyrosine kinases are proteins involved in regulation of cell growth, adhesion, migration and apoptosis. They associate with metastatic dissemination in solid tumors and poor prognosis. However, their role in haematological malignancies has been scarcely studied. We hypothesized that PTK2/FAK, PTK2B/PYK2, LYN or SRC could be new prognostic markers in IR-AML. We assessed PTK2, PTK2B, LYN and SRC gene expression in a cohort of 324 patients, adults up to the age of 70, classified in the IR-AML cytogenetic group. Univariate and multivariate analyses showed that PTK2B, LYN and PTK2 gene expression are independent prognostic factors in IR-AML patients. PTK2B and LYN identify a patient subgroup with good prognosis within the cohort with non-favorable FLT3/NPM1 combined mutations. In contrast, PTK2 identifies a patient subgroup with poor prognosis within the worst prognosis cohort who display non-favorable FLT3/NPM1 combined mutations and underexpression of PTK2B or LYN. The combined use of these markers can refine the highly heterogeneous intermediate-risk subgroup of AML patients, and allow the development of risk-adapted post-remission chemotherapy protocols to improve their response to treatment.
Collapse
Affiliation(s)
- Victor Pallarès
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Pavelló 11, 2n pis, 08025 Barcelona, Spain.
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Mas Casanovas nº 90, 08041 Barcelona, Spain.
| | - Montserrat Hoyos
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Pavelló 11, 2n pis, 08025 Barcelona, Spain.
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Mas Casanovas nº 90, 08041 Barcelona, Spain.
| | - M Carmen Chillón
- Servicio de Hematología, IBSAL-Hospital Universitario, Centro de Investigación del Cáncer (CIC)-IBMCC, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Universidad de Salamanca, 37007 Salamanca, Spain.
| | - Eva Barragán
- Hematology Department, Hospital Universitari i Politècnic La Fe, Department of Medicine, University of Valencia, and Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, 46026 Valencia, Spain.
| | - M Isabel Prieto Conde
- Servicio de Hematología, IBSAL-Hospital Universitario, Centro de Investigación del Cáncer (CIC)-IBMCC, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Universidad de Salamanca, 37007 Salamanca, Spain.
| | - Marta Llop
- Hematology Department, Hospital Universitari i Politècnic La Fe, Department of Medicine, University of Valencia, and Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, 46026 Valencia, Spain.
| | - Aïda Falgàs
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Pavelló 11, 2n pis, 08025 Barcelona, Spain.
| | - María Virtudes Céspedes
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Pavelló 11, 2n pis, 08025 Barcelona, Spain.
- CIBER en Bioinginiería, Biomateriales y Nanomedicina (CIBER-BBN), 08025 Barcelona, Spain.
| | - Pau Montesinos
- Hematology Department, Hospital Universitari i Politècnic La Fe, Department of Medicine, University of Valencia, and Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, 46026 Valencia, Spain.
| | - Josep F Nomdedeu
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Mas Casanovas nº 90, 08041 Barcelona, Spain.
| | - Salut Brunet
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Mas Casanovas nº 90, 08041 Barcelona, Spain.
| | - Miguel Ángel Sanz
- Hematology Department, Hospital Universitari i Politècnic La Fe, Department of Medicine, University of Valencia, and Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, 46026 Valencia, Spain.
| | - Marcos González-Díaz
- Servicio de Hematología, IBSAL-Hospital Universitario, Centro de Investigación del Cáncer (CIC)-IBMCC, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Universidad de Salamanca, 37007 Salamanca, Spain.
| | - Jorge Sierra
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Mas Casanovas nº 90, 08041 Barcelona, Spain.
- Josep Carreras Leukemia Research Institute, 08021 Barcelona, Spain.
- Hematology Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Pavelló 11, 2n pis, 08025 Barcelona, Spain.
- CIBER en Bioinginiería, Biomateriales y Nanomedicina (CIBER-BBN), 08025 Barcelona, Spain.
- Josep Carreras Leukemia Research Institute, 08021 Barcelona, Spain.
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Pavelló 11, 2n pis, 08025 Barcelona, Spain.
- CIBER en Bioinginiería, Biomateriales y Nanomedicina (CIBER-BBN), 08025 Barcelona, Spain.
- Josep Carreras Leukemia Research Institute, 08021 Barcelona, Spain.
| |
Collapse
|
35
|
Potluri S, Coleman D, Bonifer C. Pharmacological inhibition of aberrant transcription factor complexes in inversion 16 acute myeloid leukemia. Stem Cell Investig 2018; 5:30. [PMID: 30363728 PMCID: PMC6182015 DOI: 10.21037/sci.2018.09.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Sandeep Potluri
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Daniel Coleman
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Constanze Bonifer
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
36
|
Koblish H, Li YL, Shin N, Hall L, Wang Q, Wang K, Covington M, Marando C, Bowman K, Boer J, Burke K, Wynn R, Margulis A, Reuther GW, Lambert QT, Dostalik Roman V, Zhang K, Feng H, Xue CB, Diamond S, Hollis G, Yeleswaram S, Yao W, Huber R, Vaddi K, Scherle P. Preclinical characterization of INCB053914, a novel pan-PIM kinase inhibitor, alone and in combination with anticancer agents, in models of hematologic malignancies. PLoS One 2018; 13:e0199108. [PMID: 29927999 PMCID: PMC6013247 DOI: 10.1371/journal.pone.0199108] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/17/2022] Open
Abstract
The Proviral Integration site of Moloney murine leukemia virus (PIM) serine/threonine protein kinases are overexpressed in many hematologic and solid tumor malignancies and play central roles in intracellular signaling networks important in tumorigenesis, including the Janus kinase-signal transducer and activator of transcription (JAK/STAT) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. The three PIM kinase isozymes (PIM1, PIM2, and PIM3) share similar downstream substrates with other key oncogenic kinases and have differing but mutually compensatory functions across tumors. This supports the therapeutic potential of pan-PIM kinase inhibitors, especially in combination with other anticancer agents chosen based on their role in overlapping signaling networks. Reported here is a preclinical characterization of INCB053914, a novel, potent, and selective adenosine triphosphate-competitive pan-PIM kinase inhibitor. In vitro, INCB053914 inhibited proliferation and the phosphorylation of downstream substrates in cell lines from multiple hematologic malignancies. Effects were confirmed in primary bone marrow blasts from patients with acute myeloid leukemia treated ex vivo and in blood samples from patients receiving INCB053914 in an ongoing phase 1 dose-escalation study. In vivo, single-agent INCB053914 inhibited Bcl-2-associated death promoter protein phosphorylation and dose-dependently inhibited tumor growth in acute myeloid leukemia and multiple myeloma xenografts. Additive or synergistic inhibition of tumor growth was observed when INCB053914 was combined with selective PI3Kδ inhibition, selective JAK1 or JAK1/2 inhibition, or cytarabine. Based on these data, pan-PIM kinase inhibitors, including INCB053914, may have therapeutic utility in hematologic malignancies when combined with other inhibitors of oncogenic kinases or standard chemotherapeutics.
Collapse
Affiliation(s)
- Holly Koblish
- Incyte Corporation, Wilmington, Delaware, United States of America
- * E-mail:
| | - Yun-long Li
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Niu Shin
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Leslie Hall
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Qian Wang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kathy Wang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | | | - Cindy Marando
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kevin Bowman
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Jason Boer
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Krista Burke
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Richard Wynn
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Alex Margulis
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Gary W. Reuther
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Que T. Lambert
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | | | - Ke Zhang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Hao Feng
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Chu-Biao Xue
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Sharon Diamond
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Greg Hollis
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Swamy Yeleswaram
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Wenqing Yao
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Reid Huber
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kris Vaddi
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Peggy Scherle
- Incyte Corporation, Wilmington, Delaware, United States of America
| |
Collapse
|
37
|
Johansen S, Brenner AK, Bartaula-Brevik S, Reikvam H, Bruserud Ø. The Possible Importance of β3 Integrins for Leukemogenesis and Chemoresistance in Acute Myeloid Leukemia. Int J Mol Sci 2018; 19:ijms19010251. [PMID: 29342970 PMCID: PMC5796198 DOI: 10.3390/ijms19010251] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 12/25/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive bone marrow malignancy where the immature leukemia cells communicate with neighboring cells through constitutive cytokine release and through their cell surface adhesion molecules. The primary AML cells express various integrins. These heterodimeric molecules containing an α and a β chain are cell surface molecules that bind extracellular matrix molecules, cell surface molecules and soluble mediators. The β3 integrin (ITGB3) chain can form heterodimers only with the two α chains αIIb and αV. These integrins are among the most promiscuous and bind to a large number of ligands, including extracellular matrix molecules, cell surface molecules and soluble mediators. Recent studies suggest that the two β3 integrins are important for leukemogenesis and chemosensitivity in human AML. Firstly, αIIb and β3 are both important for adhesion of AML cells to vitronectin and fibronectin. Secondly, β3 is important for the development of murine AML and also for the homing and maintenance of the proliferation for xenografted primary human AML cells, and for maintaining a stem cell transcriptional program. These last effects seem to be mediated through Syk kinase. The β3 expression seems to be regulated by HomeboxA9 (HoxA9) and HoxA10, and the increased β3 expression then activates spleen tyrosine kinase (Syk) and thereby contributes to cytokine hypersensitivity and activation of β2 integrins. Finally, high integrin αV/β3 expression is associated with an adverse prognosis in AML and decreased sensitivity to the kinase inhibitor sorafenib; this integrin can also be essential for osteopontin-induced sorafenib resistance in AML. In the present article, we review the experimental and clinical evidence for a role of β3 integrins for leukemogenesis and chemosensitivity in AML.
Collapse
MESH Headings
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Integrin beta3/chemistry
- Integrin beta3/genetics
- Integrin beta3/metabolism
- Integrins/chemistry
- Integrins/genetics
- Integrins/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Ligands
- Multigene Family
- Prognosis
- Protein Binding
- Signal Transduction
Collapse
Affiliation(s)
- Silje Johansen
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Annette K Brenner
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Sushma Bartaula-Brevik
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Håkon Reikvam
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Øystein Bruserud
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| |
Collapse
|
38
|
Dovey OM, Cooper JL, Mupo A, Grove CS, Lynn C, Conte N, Andrews RM, Pacharne S, Tzelepis K, Vijayabaskar MS, Green P, Rad R, Arends M, Wright P, Yusa K, Bradley A, Varela I, Vassiliou GS. Molecular synergy underlies the co-occurrence patterns and phenotype of NPM1-mutant acute myeloid leukemia. Blood 2017; 130:1911-1922. [PMID: 28835438 PMCID: PMC5672315 DOI: 10.1182/blood-2017-01-760595] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/23/2017] [Indexed: 02/06/2023] Open
Abstract
NPM1 mutations define the commonest subgroup of acute myeloid leukemia (AML) and frequently co-occur with FLT3 internal tandem duplications (ITD) or, less commonly, NRAS or KRAS mutations. Co-occurrence of mutant NPM1 with FLT3-ITD carries a significantly worse prognosis than NPM1-RAS combinations. To understand the molecular basis of these observations, we compare the effects of the 2 combinations on hematopoiesis and leukemogenesis in knock-in mice. Early effects of these mutations on hematopoiesis show that compound Npm1cA/+;NrasG12D/+ or Npm1cA;Flt3ITD share a number of features: Hox gene overexpression, enhanced self-renewal, expansion of hematopoietic progenitors, and myeloid differentiation bias. However, Npm1cA;Flt3ITD mutants displayed significantly higher peripheral leukocyte counts, early depletion of common lymphoid progenitors, and a monocytic bias in comparison with the granulocytic bias in Npm1cA/+;NrasG12D/+ mutants. Underlying this was a striking molecular synergy manifested as a dramatically altered gene expression profile in Npm1cA;Flt3ITD , but not Npm1cA/+;NrasG12D/+ , progenitors compared with wild-type. Both double-mutant models developed high-penetrance AML, although latency was significantly longer with Npm1cA/+;NrasG12D/+ During AML evolution, both models acquired additional copies of the mutant Flt3 or Nras alleles, but only Npm1cA/+;NrasG12D/+ mice showed acquisition of other human AML mutations, including IDH1 R132Q. We also find, using primary Cas9-expressing AMLs, that Hoxa genes and selected interactors or downstream targets are required for survival of both types of double-mutant AML. Our results show that molecular complementarity underlies the higher frequency and significantly worse prognosis associated with NPM1c/FLT3-ITD vs NPM1/NRAS-G12D-mutant AML and functionally confirm the role of HOXA genes in NPM1c-driven AML.
Collapse
Affiliation(s)
- Oliver M Dovey
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Jonathan L Cooper
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Annalisa Mupo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Carolyn S Grove
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Australia
- PathWest Division of Clinical Pathology, Queen Elizabeth II Medical Centre, Nedlands, Australia
| | - Claire Lynn
- Leukemia and Stem Cell Biology Group, Division of Cancer Studies, Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Nathalie Conte
- Sample Phenotype Ontology Team, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Robert M Andrews
- Institute of Translation, Innovation, Methodology, and Engagement, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Suruchi Pacharne
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Konstantinos Tzelepis
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - M S Vijayabaskar
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Paul Green
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Roland Rad
- Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
- German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Mark Arends
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Penny Wright
- Department of Haematology, Cambridge University Hospitals NHS Trust, Cambridge, United Kingdom; and
| | - Kosuke Yusa
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Allan Bradley
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Ignacio Varela
- Instituto de Biomedicina y Biotecnología de Cantabria, Santander, Spain
| | - George S Vassiliou
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Haematology, Cambridge University Hospitals NHS Trust, Cambridge, United Kingdom; and
| |
Collapse
|
39
|
Kumar R, Deivendran S, Santhoshkumar TR, Pillai MR. Signaling coupled epigenomic regulation of gene expression. Oncogene 2017. [DOI: 10.1038/onc.2017.201] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Hospital MA, Green AS, Maciel TT, Moura IC, Leung AY, Bouscary D, Tamburini J. FLT3 inhibitors: clinical potential in acute myeloid leukemia. Onco Targets Ther 2017; 10:607-615. [PMID: 28223820 PMCID: PMC5304990 DOI: 10.2147/ott.s103790] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy that is cured in as few as 15%–40% of cases. Tremendous improvements in AML prognostication arose from a comprehensive analysis of leukemia cell genomes. Among normal karyotype AML cases, mutations in the FLT3 gene are the ones most commonly detected as having a deleterious prognostic impact. FLT3 is a transmembrane tyrosine kinase receptor, and alterations of the FLT3 gene such as internal tandem duplications (FLT3-ITD) deregulate FLT3 downstream signaling pathways in favor of increased cell proliferation and survival. FLT3 tyrosine kinase inhibitors (TKI) emerged as a new therapeutic option in FLT3-ITD AML, and clinical trials are ongoing with a variety of TKI either alone, combined with chemotherapy, or even as maintenance after allogenic stem cell transplantation. However, a wide range of molecular resistance mechanisms are activated upon TKI therapy, thus limiting their clinical impact. Massive research efforts are now ongoing to develop more efficient FLT3 TKI and/or new therapies targeting these resistance mechanisms to improve the prognosis of FLT3-ITD AML patients in the future.
Collapse
Affiliation(s)
- Marie-Anne Hospital
- Département Développement, Reproduction, Cancer, Institut Cochin, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016; Faculté de Médecine Sorbonne Paris Cité, Université Paris Descartes; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC)
| | - Alexa S Green
- Département Développement, Reproduction, Cancer, Institut Cochin, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016; Faculté de Médecine Sorbonne Paris Cité, Université Paris Descartes; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC)
| | - Thiago T Maciel
- INSERM UMR 1163, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications; Paris Descartes - Sorbonne Paris Cité University; CNRS ERL 8254, Imagine Institute; Laboratory of Excellence GR-Ex, Paris, France
| | - Ivan C Moura
- INSERM UMR 1163, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications; Paris Descartes - Sorbonne Paris Cité University; CNRS ERL 8254, Imagine Institute; Laboratory of Excellence GR-Ex, Paris, France
| | - Anskar Y Leung
- Department of Medicine, Division of Hematology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Didier Bouscary
- Département Développement, Reproduction, Cancer, Institut Cochin, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016; Faculté de Médecine Sorbonne Paris Cité, Université Paris Descartes; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC)
| | - Jerome Tamburini
- Département Développement, Reproduction, Cancer, Institut Cochin, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016; Faculté de Médecine Sorbonne Paris Cité, Université Paris Descartes; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC)
| |
Collapse
|
41
|
O-GlcNAcylation of STAT5 controls tyrosine phosphorylation and oncogenic transcription in STAT5-dependent malignancies. Leukemia 2017; 31:2132-2142. [PMID: 28074064 PMCID: PMC5629373 DOI: 10.1038/leu.2017.4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 12/14/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022]
Abstract
The signal transducer and activator of transcription 5 (STAT5) regulates differentiation, survival, proliferation and transformation of hematopoietic cells. Upon cytokine stimulation, STAT5 tyrosine phosphorylation (pYSTAT5) is transient, while in diverse neoplastic cells persistent overexpression and enhanced pYSTAT5 are frequently found. Post-translational modifications might contribute to enhanced STAT5 activation in the context of transformation, but the strength and duration of pYSTAT5 are incompletely understood. We found that O-GlcNAcylation and tyrosine phosphorylation act together to trigger pYSTAT5 levels and oncogenic transcription in neoplastic cells. The expression of a mutated hyperactive gain-of-function (GOF) STAT5 without O-GlcNAcylation resulted in decreased tyrosine phosphorylation, oligomerization and transactivation potential and complete loss of oncogenic transformation capacity. The lack of O-GlcNAcylation diminished phospho-ERK and phospho-AKT levels. Our data show that O-GlcNAcylation of STAT5 is an important process that contributes to oncogenic transcription through enhanced STAT5 tyrosine phosphorylation and oligomerization driving myeloid transformation. O-GlcNAcylation of STAT5 could be required for nutrient sensing and metabolism of cancer cells.
Collapse
|
42
|
Kumar R, Sanawar R, Li X, Li F. Structure, biochemistry, and biology of PAK kinases. Gene 2016; 605:20-31. [PMID: 28007610 DOI: 10.1016/j.gene.2016.12.014] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/24/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
PAKs, p21-activated kinases, play central roles and act as converging junctions for discrete signals elicited on the cell surface and for a number of intracellular signaling cascades. PAKs phosphorylate a vast number of substrates and act by remodeling cytoskeleton, employing scaffolding, and relocating to distinct subcellular compartments. PAKs affect wide range of processes that are crucial to the cell from regulation of cell motility, survival, redox, metabolism, cell cycle, proliferation, transformation, stress, inflammation, to gene expression. Understandably, their dysregulation disrupts cellular homeostasis and severely impacts key cell functions, and many of those are implicated in a number of human diseases including cancers, neurological disorders, and cardiac disorders. Here we provide an overview of the members of the PAK family and their current status. We give special emphasis to PAK1 and PAK4, the prototypes of groups I and II, for their profound roles in cancer, the nervous system, and the heart. We also highlight other family members. We provide our perspective on the current advancements, their growing importance as strategic therapeutic targets, and our vision on the future of PAKs.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA; Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India.
| | - Rahul Sanawar
- Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India
| | - Xiaodong Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China.
| |
Collapse
|
43
|
Wu M, Hamaker M, Li L, Small D, Duffield AS. DOCK2 interacts with FLT3 and modulates the survival of FLT3-expressing leukemia cells. Leukemia 2016; 31:688-696. [PMID: 27748370 PMCID: PMC5332301 DOI: 10.1038/leu.2016.284] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 08/12/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022]
Abstract
The FMS-like tyrosine kinase-3 (FLT3) gene is the most commonly mutated gene in acute myeloid leukemia (AML), and patients carrying internal tandem duplication (ITD) mutations have a poor prognosis. Long-term inhibition of FLT3 activity in these patients has been elusive. To provide a more complete understanding of FLT3 biology, a mass spectroscopy-based screen was performed to search for FLT3-interacting proteins. The screen identified dedicator of cytokinesis 2 (DOCK2), which is a guanine nucleotide exchange factor for Rho GTPases, and its expression is limited to hematolymphoid cells. We show that DOCK2 is expressed in leukemia cell lines and primary AML samples, and DOCK2 co-immunoprecipitates with wild-type FLT3 and FLT3/ITD. Knock-down (KD) of DOCK2 by shRNA selectively reduced cell proliferation and colony formation in leukemia cell lines with increased FLT3 activity, and greatly sensitized these cells to cytarabine treatment, alone and in combination with FLT3 tyrosine kinase inhibitors. DOCK2 KD in a FLT3/ITD-positive leukemia cell line also significantly prolonged survival in a mouse xenograft model. These findings suggest that DOCK2 is a potential therapeutic target for novel AML treatments, as this protein regulates the survival of leukemia cells with elevated FLT3 activity and sensitizes FLT3/ITD leukemic cells to conventional anti-leukemic agents.
Collapse
Affiliation(s)
- M Wu
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - M Hamaker
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - L Li
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - D Small
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - A S Duffield
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
44
|
Chi M, Evans H, Gilchrist J, Mayhew J, Hoffman A, Pearsall EA, Jankowski H, Brzozowski JS, Skelding KA. Phosphorylation of calcium/calmodulin-stimulated protein kinase II at T286 enhances invasion and migration of human breast cancer cells. Sci Rep 2016; 6:33132. [PMID: 27605043 PMCID: PMC5015093 DOI: 10.1038/srep33132] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/22/2016] [Indexed: 01/01/2023] Open
Abstract
Calcium/calmodulin-stimulated protein kinase II (CaMKII) is a multi-functional kinase that controls a range of cellular functions, including proliferation, differentiation and apoptosis. The biological properties of CaMKII are regulated by multi-site phosphorylation. However, the role that CaMKII phosphorylation plays in cancer cell metastasis has not been examined. We demonstrate herein that CaMKII expression and phosphorylation at T286 is increased in breast cancer when compared to normal breast tissue, and that increased CAMK2 mRNA is associated with poor breast cancer patient prognosis (worse overall and distant metastasis free survival). Additionally, we show that overexpression of WT, T286D and T286V forms of CaMKII in MDA-MB-231 and MCF-7 breast cancer cells increases invasion, migration and anchorage independent growth, and that overexpression of the T286D phosphomimic leads to a further increase in the invasive, migratory and anchorage independent growth capacity of these cells. Pharmacological inhibition of CaMKII decreases MDA-MB-231 migration and invasion. Furthermore, we demonstrate that overexpression of T286D, but not WT or T286V-CaMKII, leads to phosphorylation of FAK, STAT5a, and Akt. These results demonstrate a novel function for phosphorylation of CaMKII at T286 in the control of breast cancer metastasis, offering a promising target for the development of therapeutics to prevent breast cancer metastasis.
Collapse
Affiliation(s)
- Mengna Chi
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Cancer, Hunter Medical Research Institute, Faculty of Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Hamish Evans
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Jackson Gilchrist
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Jack Mayhew
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Alexander Hoffman
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Elizabeth Ann Pearsall
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Cancer, Hunter Medical Research Institute, Faculty of Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Helen Jankowski
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Cancer, Hunter Medical Research Institute, Faculty of Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Joshua Stephen Brzozowski
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Cancer, Hunter Medical Research Institute, Faculty of Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Kathryn Anne Skelding
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Cancer, Hunter Medical Research Institute, Faculty of Health, The University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
45
|
Chatterjee A, Ghosh J, Kapur R. Mastocytosis: a mutated KIT receptor induced myeloproliferative disorder. Oncotarget 2016; 6:18250-64. [PMID: 26158763 PMCID: PMC4621888 DOI: 10.18632/oncotarget.4213] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/23/2015] [Indexed: 12/20/2022] Open
Abstract
Although more than 90% systemic mastocytosis (SM) patients express gain of function mutations in the KIT receptor, recent next generation sequencing has revealed the presence of several additional genetic and epigenetic mutations in a subset of these patients, which confer poor prognosis and inferior overall survival. A clear understanding of how genetic and epigenetic mutations cooperate in regulating the tremendous heterogeneity observed in these patients will be essential for designing effective treatment strategies for this complex disease. In this review, we describe the clinical heterogeneity observed in patients with mastocytosis, the nature of relatively novel mutations identified in these patients, therapeutic strategies to target molecules downstream from activating KIT receptor and finally we speculate on potential novel strategies to interfere with the function of not only the oncogenic KIT receptor but also epigenetic mutations seen in these patients.
Collapse
Affiliation(s)
- Anindya Chatterjee
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Joydeep Ghosh
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Reuben Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Molecular Biology and Biochemistry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
46
|
Kumar R, Li DQ. PAKs in Human Cancer Progression: From Inception to Cancer Therapeutic to Future Oncobiology. Adv Cancer Res 2016; 130:137-209. [PMID: 27037753 DOI: 10.1016/bs.acr.2016.01.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since the initial recognition of a mechanistic role of p21-activated kinase 1 (PAK1) in breast cancer invasion, PAK1 has emerged as one of the widely overexpressed or hyperactivated kinases in human cancer at-large, allowing the PAK family to make in-roads in cancer biology, tumorigenesis, and cancer therapeutics. Much of our current understanding of the PAK family in cancer progression relates to a central role of the PAK family in the integration of cancer-promoting signals from cell membrane receptors as well as function as a key nexus-modifier of complex, cytoplasmic signaling network. Another core aspect of PAK signaling that highlights its importance in cancer progression is through PAK's central role in the cross talk with signaling and interacting proteins, as well as PAK's position as a key player in the phosphorylation of effector substrates to engage downstream components that ultimately leads to the development cancerous phenotypes. Here we provide a comprehensive review of the recent advances in PAK cancer research and its downstream substrates in the context of invasion, nuclear signaling and localization, gene expression, and DNA damage response. We discuss how a deeper understanding of PAK1's pathobiology over the years has widened research interest to the PAK family and human cancer, and positioning the PAK family as a promising cancer therapeutic target either alone or in combination with other therapies. With many landmark findings and leaps in the progress of PAK cancer research since the infancy of this field nearly 20 years ago, we also discuss postulated advances in the coming decade as the PAK family continues to shape the future of oncobiology.
Collapse
Affiliation(s)
- R Kumar
- School of Medicine and Health Sciences, George Washington University, Washington, DC, United States; Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram, India.
| | - D-Q Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China; Key Laboratory of Epigenetics in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
47
|
Berenstein R. Class III Receptor Tyrosine Kinases in Acute Leukemia - Biological Functions and Modern Laboratory Analysis. Biomark Insights 2015; 10:1-14. [PMID: 26309392 PMCID: PMC4527365 DOI: 10.4137/bmi.s22433] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/02/2015] [Accepted: 07/04/2015] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a complex disease caused by deregulation of multiple signaling pathways. Mutations in class III receptor tyrosine kinases (RTKs) have been implicated in alteration of cell signals concerning the growth and differentiation of leukemic cells. Point mutations, insertions, or deletions of RTKs as well as chromosomal translocations induce constitutive activation of the receptor, leading to uncontrolled proliferation of undifferentiated myeloid blasts. Aberrations can occur in all domains of RTKs causing either the ligand-independent activation or mimicking the activated conformation. The World Health Organization recommended including RTK mutations in the AML classification since their detection in routine laboratory diagnostics is a major factor for prognostic stratification of patients. Polymerase chain reaction (PCR)-based methods are well-validated for the detection of fms-related tyrosine kinase 3 (FLT3) mutations and can easily be applied for other RTKs. However, when methodological limitations are reached, accessory techniques can be applied. For a higher resolution and more quantitative approach compared to agarose gel electrophoresis, PCR fragments can be separated by capillary electrophoresis. Furthermore, high-resolution melting and denaturing high-pressure liquid chromatography are reliable presequencing screening methods that reduce the sample amount for Sanger sequencing. Because traditional DNA sequencing is time-consuming, next-generation sequencing (NGS) is an innovative modern possibility to analyze a high amount of samples simultaneously in a short period of time. At present, standardized procedures for NGS are not established, but when this barrier is resolved, it will provide a new platform for rapid and reliable laboratory diagnostic of RTK mutations in patients with AML. In this article, the biological and physiological role of RTK mutations in AML as well as possible laboratory methods for their detection will be reviewed.
Collapse
Affiliation(s)
- Rimma Berenstein
- Department of Hematology, Oncology and Tumourimmunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
48
|
Cauchy P, James SR, Zacarias-Cabeza J, Ptasinska A, Imperato MR, Assi SA, Piper J, Canestraro M, Hoogenkamp M, Raghavan M, Loke J, Akiki S, Clokie SJ, Richards SJ, Westhead DR, Griffiths MJ, Ott S, Bonifer C, Cockerill PN. Chronic FLT3-ITD Signaling in Acute Myeloid Leukemia Is Connected to a Specific Chromatin Signature. Cell Rep 2015; 12:821-36. [PMID: 26212328 PMCID: PMC4726916 DOI: 10.1016/j.celrep.2015.06.069] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 05/20/2015] [Accepted: 06/19/2015] [Indexed: 12/24/2022] Open
Abstract
Acute myeloid leukemia (AML) is characterized by recurrent mutations that affect the epigenetic regulatory machinery and signaling molecules, leading to a block in hematopoietic differentiation. Constitutive signaling from mutated growth factor receptors is a major driver of leukemic growth, but how aberrant signaling affects the epigenome in AML is less understood. Furthermore, AML cells undergo extensive clonal evolution, and the mutations in signaling genes are often secondary events. To elucidate how chronic growth factor signaling alters the transcriptional network in AML, we performed a system-wide multi-omics study of primary cells from patients suffering from AML with internal tandem duplications in the FLT3 transmembrane domain (FLT3-ITD). This strategy revealed cooperation between the MAP kinase (MAPK) inducible transcription factor AP-1 and RUNX1 as a major driver of a common, FLT3-ITD-specific gene expression and chromatin signature, demonstrating a major impact of MAPK signaling pathways in shaping the epigenome of FLT3-ITD AML.
Collapse
Affiliation(s)
- Pierre Cauchy
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK
| | - Sally R James
- Section of Experimental Haematology, Leeds Institute for Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Joaquin Zacarias-Cabeza
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK
| | - Anetta Ptasinska
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK
| | - Maria Rosaria Imperato
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK
| | - Salam A Assi
- Section of Experimental Haematology, Leeds Institute for Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Jason Piper
- Warwick Systems Biology Centre, University of Warwick, Coventry CV4 7AL, UK
| | - Martina Canestraro
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK
| | - Maarten Hoogenkamp
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK
| | - Manoj Raghavan
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK; Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham B15 2TH, UK
| | - Justin Loke
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK
| | - Susanna Akiki
- West Midlands Regional Genetics Laboratory, Birmingham Women's NHS Foundation Trust, Birmingham B15 2TG, UK
| | - Samuel J Clokie
- West Midlands Regional Genetics Laboratory, Birmingham Women's NHS Foundation Trust, Birmingham B15 2TG, UK
| | - Stephen J Richards
- Haematological Malignancy Diagnostic Service, St. James's University Hospital, Leeds LS9 7TF, UK
| | - David R Westhead
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Michael J Griffiths
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK; West Midlands Regional Genetics Laboratory, Birmingham Women's NHS Foundation Trust, Birmingham B15 2TG, UK
| | - Sascha Ott
- Warwick Systems Biology Centre, University of Warwick, Coventry CV4 7AL, UK
| | - Constanze Bonifer
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK.
| | - Peter N Cockerill
- School of Immunity and Infection, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
49
|
PAK1 is a therapeutic target in acute myeloid leukemia and myelodysplastic syndrome. Blood 2015; 126:1118-27. [PMID: 26170031 DOI: 10.1182/blood-2014-12-618801] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/26/2015] [Indexed: 12/14/2022] Open
Abstract
Poor clinical outcome of acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) has been attributed to failure of current chemotherapeutic regimens to target leukemic stem cells. We recently identified p21-activated kinase (PAK1) as a downstream effector molecule of H2.0-like homeobox (HLX), a gene functionally relevant for AML pathogenesis. In this study, we find that inhibition of PAK1 activity by small molecule inhibitors or by RNA interference leads to profound leukemia inhibitory effects both in vitro and in vivo. Inhibition of PAK1 induces differentiation and apoptosis of AML cells through downregulation of the MYC oncogene and a core network of MYC target genes. Importantly, we find that inhibition of PAK1 inhibits primary human leukemic cells including immature leukemic stem cell-enriched populations. Moreover, we find that PAK1 upregulation occurs during disease progression and is relevant for patient survival in MDS. Our studies highlight PAK1 as a novel target in AML and MDS and support the use of PAK1 inhibitors as a therapeutic strategy in these diseases.
Collapse
|
50
|
Pandey R, Kapur R. Targeting phosphatidylinositol-3-kinase pathway for the treatment of Philadelphia-negative myeloproliferative neoplasms. Mol Cancer 2015; 14:118. [PMID: 26062813 PMCID: PMC4464249 DOI: 10.1186/s12943-015-0388-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/18/2015] [Indexed: 12/24/2022] Open
Abstract
Myeloproliferative neoplasms (MPN) are a diverse group of chronic hematological disorders that involve unregulated clonal proliferation of white blood cells. Sevearl of them are associated with mutations in receptor tyrosine kinases or cytokine receptor associated tyrosine kinases rendering them independent of cytokine-mediated regulation. Classically they have been broadly divided into BCR-ABL1 fusion + ve (Ph + ve) or -ve (Ph-ve) MPNs. Identification of BCR-ABL1 tyrosine kinase as a driver of chronic myeloid leukemia (CML) and successful application of small molecule inhibitors of the tyrosine kinases in the clinic have triggered the search for kinase dependent pathways in other Ph-ve MPNs. In the past few years, identification of mutations in JAK2 associated with a majority of MPNs raised the hopes for similar success with specific targeting of JAK2. However, targeting JAK2 kinase activity has met with limited success. Subsequently, mutations in genes other than JAK2 have been identified. These mutations specifically associate with certain MPNs and can drive cytokine independent growth. Therefore, targeting alternate molecules and pathways may be more successful in management of MPNs. Among other pathways, phosphatidylinositol -3 kinase (PI3K) has emerged as a promising target as different cell surface receptor induced signaling pathways converge on the PI3K signaling axis to regulate cell metabolism, growth, proliferation, and survival. Herein, we will review the clinically relevant inhibitors of the PI3K pathway that have been evaluated or hold promise for the treatment of Ph-ve MPNs.
Collapse
Affiliation(s)
- Ruchi Pandey
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Reuben Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Molecular Biology and Biochemistry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|