1
|
Lu C, Zhang J, Wang B, Gao Q, Ma K, Pei S, Li J, Cui S. Casein kinase 1α is required to maintain murine hypothalamic pro-opiomelanocortin expression. iScience 2023; 26:106670. [PMID: 37168577 PMCID: PMC10165255 DOI: 10.1016/j.isci.2023.106670] [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: 12/13/2022] [Revised: 02/08/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Hypothalamic pro-opiomelanocortin (POMC) neuron development is considered to play an essential role in the development of obesity. However, the underlying mechanisms remain unclear. Casein kinase 1α (CK1α) was expressed in the embryonic mouse hypothalamus at high levels and colocalized with POMC neurons. CK1α deletion in POMC neurons caused weight gain, metabolic defects, and increased food intake. The number of POMC-expressing cells was considerably decreased in Csnk1a1fl/fl;POMCcre (PKO) mice from embryonic day 15.5 to postnatal day 60, while apoptosis of POMC neurons was not affected. Furthermore, unchanged POMC progenitor cells and a decreased POMC phenotype established CK1α function in hypothalamic POMC neuron development. CK1α deletion led to elevated Notch intracellular domain (NICD) protein expression, and NICD inhibition rescued the PKO mouse phenotype. In summary, CK1α is involved in hypothalamic POMC expression via NICD-POMC signaling, deepening our understanding of POMC neuron development and control of systemic metabolic functions.
Collapse
Affiliation(s)
- Chenyang Lu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Jinglin Zhang
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Bingjie Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Qiao Gao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Kezhe Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Shaona Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People’s Republic of China
- Corresponding author
| |
Collapse
|
2
|
CRL4 CRBN E3 Ligase Complex as a Therapeutic Target in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14184492. [PMID: 36139651 PMCID: PMC9496858 DOI: 10.3390/cancers14184492] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Immunomodulatory drugs (IMiDs) are effective in the treatment of multiple myeloma (MM) and other hematological malignancies. Cereblon (CRBN), a target of IMiDs, forms the CRL4 E3 ubiquitin ligase complex (CRL4CRBN) with DDB1, CUL4A and RBX1. The insight into the molecular mechanism of IMiDs action has advanced dramatically since the identification of cereblon (CRBN) as their direct target. Targeting CRBN by IMiDs modifies CRL4CRBN substrate specificity towards non-physiological protein targets which are subsequently ubiquitinated and degraded by the proteasome. To date, IMiDs are the only known group of protein degraders used in clinical practice. This review provides the current state of knowledge about thalidomide and its derivatives’ mechanisms of action, and highlights the future perspectives for targeted protein degraders. Abstract Multiple myeloma (MM) is the second most common hematological malignancy with a recurrent clinical course. The introduction of immunomodulatory drugs (IMiDs) was one of the milestones in MM therapy leading to a significant improvement in patients’ prognosis. Currently, IMiDs are the backbone of MM therapy in newly diagnosed and relapsed/refractory settings. It is now known that IMiDs exert their anti-myeloma activity mainly by binding cereblon (CRBN), the substrate receptor protein of the CRL4 E3 ubiquitin ligase (CRL4CRBN) complex. By binding CRBN, IMiDs alter its substrate specificity, leading to ubiquitination and proteasomal degradation of proteins essential for MM cell survival. Following the success of IMiDs, it is not surprising that the possibility of using the CRL4CRBN complex’s activity to treat MM is being further explored. In this review, we summarize the current state of knowledge about novel players in the MM therapeutic landscape, namely the CRBN E3 ligase modulators (CELMoDs), the next generation of IMiDs with broader biological activity. In addition, we discuss a new strategy of tailored proteolysis called proteolysis targeting chimeras (PROTACs) using the CRL4CRBN to degrade typically undruggable proteins, which may have relevance for the treatment of MM and other malignancies in the future.
Collapse
|
3
|
Fregnani A, Saggin L, Gianesin K, Quotti Tubi L, Carraro M, Barilà G, Scapinello G, Bonetto G, Pesavento M, Berno T, Branca A, Gurrieri C, Zambello R, Semenzato G, Trentin L, Manni S, Piazza F. CK1α/RUNX2 Axis in the Bone Marrow Microenvironment: A Novel Therapeutic Target in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14174173. [PMID: 36077711 PMCID: PMC9454895 DOI: 10.3390/cancers14174173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is an incurable disease for which novel therapeutic approaches targeting the malignant cells and the associated bone disease are urgently needed. CK1α is a protein kinase that plays a crucial role in the signaling network that sustains plasma cell (PC) survival and bone disease. This protein regulates Wnt/β-catenin signaling, which is fundamental for both MM cell survival and mesenchymal stromal cell (MSC) osteogenic differentiation. In this study, we investigated its involvement in MM–MSC cross-talk. We found that, by lowering CK1α expression levels in co-cultures of MM and MSC cells, expression of RUNX2—the master regulator of osteogenic differentiation—was regulated differently in the two cell types. Our data suggest the possibility of using a specific CK1α inhibitor as part of a novel therapeutic approach to selectively kill malignant PCs and overcome the blocking of osteogenic differentiation induced by MM cells in MSCs. Abstract Multiple myeloma (MM) is a malignant plasma cell (PC) neoplasm, which also displays pathological bone involvement. Clonal expansion of MM cells in the bone marrow causes a perturbation of bone homeostasis that culminates in MM-associated bone disease (MMABD). We previously demonstrated that the S/T kinase CK1α sustains MM cell survival through the activation of AKT and β-catenin signaling. CK1α is a negative regulator of the Wnt/β-catenin cascade, the activation of which promotes osteogenesis by directly stimulating the expression of RUNX2, the master gene regulator of osteoblastogenesis. In this study, we investigated the role of CK1α in the osteoblastogenic potential of mesenchymal stromal cells (MSCs) and its involvement in MM–MSC cross-talk. We found that CK1α silencing in in vitro co-cultures of MMs and MSCs modulated RUNX2 expression differently in PCs and in MSCs, mainly through the regulation of Wnt/β-catenin signaling. Our findings suggest that the CK1α/RUNX2 axis could be a potential therapeutic target for constraining malignant PC expansion and supporting the osteoblastic transcriptional program of MSCs, with potential for ameliorating MMABD. Moreover, considering that Lenalidomide treatment leads to MM cell death through Ikaros, Aiolos and CK1α proteasomal degradation, we examined its effects on the osteoblastogenic potential of MSC compartments.
Collapse
Affiliation(s)
- Anna Fregnani
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Lara Saggin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Ketty Gianesin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Laura Quotti Tubi
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Marco Carraro
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Gregorio Barilà
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Greta Scapinello
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Giorgia Bonetto
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Maria Pesavento
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Tamara Berno
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Antonio Branca
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Carmela Gurrieri
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Renato Zambello
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Gianpietro Semenzato
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Sabrina Manni
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Correspondence: (S.M.); (F.P.); Tel.: +39-049-7923263 (S.M. & F.P.); Fax: +39-049-7923250 (S.M. & F.P.)
| | - Francesco Piazza
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Correspondence: (S.M.); (F.P.); Tel.: +39-049-7923263 (S.M. & F.P.); Fax: +39-049-7923250 (S.M. & F.P.)
| |
Collapse
|
4
|
Luo H, Zhang D, Wang F, Wang Q, Wu Y, Gou M, Hu Y, Zhang W, Huang J, Gong Y, Pan L, Li T, Zhao P, Zhang D, Qu Y, Liu Z, Jiang T, Dai Y, Guo T, Zhu J, Ye L, Zhang L, Liu W, Yi Q, Zheng Y. ALCAM-EGFR interaction regulates myelomagenesis. Blood Adv 2021; 5:5269-5282. [PMID: 34592762 PMCID: PMC9152994 DOI: 10.1182/bloodadvances.2021004695] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/28/2021] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma, a plasma cell malignancy in the bone marrow, remains largely incurable with currently available therapeutics. In this study, we discovered that the activated leukocyte cell adhesion molecule (ALCAM) interacted with epidermal growth factor receptor (EGFR), and regulated myelomagenesis. ALCAM was a negative regulator of myeloma clonogenicity. ALCAM expression was positively correlated with patients' survival. ALCAM-knockdown myeloma cells displayed enhanced colony formation in the presence of bone marrow stromal cells (BMSCs). BMSCs supported myeloma colony formation by secreted epidermal growth factor (EGF), which bound with its receptor (EGFR) on myeloma cells and activated Mek/Erk cell signaling, PI3K/Akt cell signaling, and hedgehog pathway. ALCAM could also bind with EGFR, block EGF from binding to EGFR, and abolish EGFR-initiated cell signaling. Hence, our study identifies ALCAM as a novel negative regulator of myeloma pathogenesis.
Collapse
Affiliation(s)
- Hongmei Luo
- Department of Hematology, West China Hospital
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Dan Zhang
- Department of Hematology, West China Hospital
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Qiang Wang
- Center for Translational Research in Hematological Malignancies, Cancer Center, Houston Methodist Hospital, Houston, TX
| | - Yu Wu
- Department of Hematology, West China Hospital
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Yiguo Hu
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | | | - Jingcao Huang
- Department of Hematology, West China Hospital
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Yuping Gong
- Department of Hematology, West China Hospital
| | - Ling Pan
- Department of Hematology, West China Hospital
| | - Tianshu Li
- Center for Translational Research in Hematological Malignancies, Cancer Center, Houston Methodist Hospital, Houston, TX
| | - Pan Zhao
- Department of Hematology, West China Hospital
| | | | - Ying Qu
- Department of Hematology, West China Hospital
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Zhigang Liu
- Department of Hematology, West China Hospital
| | - Tao Jiang
- Department of Hematology, West China Hospital
| | - Yang Dai
- Department of Hematology, West China Hospital
| | | | - Jiang Zhu
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Lingqun Ye
- Center for Translational Research in Hematological Malignancies, Cancer Center, Houston Methodist Hospital, Houston, TX
| | - Li Zhang
- Department of Hematology, West China Hospital
| | | | - Qing Yi
- Center for Translational Research in Hematological Malignancies, Cancer Center, Houston Methodist Hospital, Houston, TX
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Molecular Mechanisms of Cereblon-Interacting Small Molecules in Multiple Myeloma Therapy. J Pers Med 2021; 11:jpm11111185. [PMID: 34834536 PMCID: PMC8623651 DOI: 10.3390/jpm11111185] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Thalidomide analogues (or immunomodulatory imide drugs, IMiDs) are cornerstones in the treatment of multiple myeloma (MM). These drugs bind Cereblon (CRBN), a receptor for the Cullin-ring 4 ubiquitin-ligase (CRL4) complex, to modify its substrate specificity. IMiDs mediate CRBN-dependent engagement and proteasomal degradation of ‘neosubstrates’, Ikaros (IKZF1) and Aiolos (IKZF3), conveying concurrent antimyeloma activity and T-cell costimulation. There is now a greater understanding of physiological CRBN functions, including endogenous substrates and chaperone activity. CRISPR Cas9-based genome-wide screening has further elucidated the complex cellular machinery implicated in IMiD sensitivity, including IKZF1/3-independent mechanisms. New-generation IMiD derivatives with more potent anti-cancer properties—the CELMoDs (Cereblon E3 ligase modulators)—are now being evaluated. Rational drug design also allows ‘hijacking’ of CRL4CRBN utilising proteolysis targeting chimeras (PROTACs) to convey entirely distinct substrate repertoires. As all these chemotypes—thalidomide, IMiDs, CELMoDs and PROTACs—engage CRBN and modify its functions, we describe them here in aggregate as ‘CRBN-interacting small molecules’ (CISMs). In this review, we provide a contemporary summary of the biological consequences of CRBN modulation by CISMs. Detailed molecular insight into CRBN–CISM interactions now provides an opportunity to more effectively target previously elusive cancer dependencies, representing a new and powerful tool for the implementation of precision medicine.
Collapse
|
6
|
Manni S, Fregnani A, Quotti Tubi L, Spinello Z, Carraro M, Scapinello G, Visentin A, Barilà G, Pizzi M, Dei Tos AP, Vianello F, Zambello R, Gurrieri C, Semenzato G, Trentin L, Piazza F. Protein Kinase CK1α Sustains B-Cell Receptor Signaling in Mantle Cell Lymphoma. Front Oncol 2021; 11:733848. [PMID: 34722279 PMCID: PMC8551451 DOI: 10.3389/fonc.2021.733848] [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: 06/30/2021] [Accepted: 09/06/2021] [Indexed: 12/25/2022] Open
Abstract
Mantle Cell Lymphoma (MCL) is still an incurable B-cell malignancy characterized by poor prognosis and frequent relapses. B Cell Receptor (BCR) signaling inhibitors, in particular of the kinases BTK and PI3Kγ/δ, have demonstrated clinically meaningful anti-proliferative effects in B cell tumors. However, refractoriness to these drugs may develop, portending a dismal prognosis. Protein kinase CK1α is an emerging pro-growth enzyme in B cell malignancies. In multiple myeloma, this kinase sustains β-catenin and AKT-dependent survival and is involved in the activation of NF-κB in B cells. In this study, we analyzed the role of CK1α on MCL cell survival and proliferation, on the regulation of BCR-related BTK, NF-κB, PI3K/AKT signaling cascades and the effects of CK1α chemical inhibition or gene silencing in association with the BTK inhibitor Ibrutinib or the PI3Kγ/δ inhibitor Duvelisib. CK1α was found highly expressed in MCL cells as compared to normal B cells. The inactivation/loss of CK1α caused MCL cell apoptosis and proliferation arrest. CK1α sustained BCR signaling, in particular the NF-κB, AKT and BTK pathways by modulating the phosphorylation of Ser 652 on CARD11, Ser 536 p65 on NF-κB, Ser 473 on AKT, Tyr 223 on BTK, as well as the protein levels. We also provided evidence that CK1α-mediated regulation of CARD11 and BTK likely implicates a physical interaction. The combination of CK1α inhibition with Ibrutinib or Duvelisib synergistically increased cytotoxicity, leading to a further decrease of the activation of BCR signaling pathways. Therefore, CK1α sustains MCL growth through the regulation of BCR-linked survival signaling cascades and protects from Ibrutinib/Duvelisib-induced apoptosis. Thus, CK1α could be considered as a rational molecular target for the treatment of MCL, in association with novel agents.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Anna Fregnani
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Zaira Spinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Carraro
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Greta Scapinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Andrea Visentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gregorio Barilà
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Pizzi
- Department of Medicine-DIMED, Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine-DIMED, Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Fabrizio Vianello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
| | - Renato Zambello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Livio Trentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
7
|
Nahi H, Kashif M, Klimkowska M, Karvouni M, Wallblom A, Gran C, Hauenstein J, Frengen N, Gustafsson C, Afram G, Uttervall K, Lund J, Månsson R, Wagner AK, Alici E. Low dose venetoclax as a single agent treatment of plasma cell malignancies harboring t(11;14). Am J Hematol 2021; 96:925-933. [PMID: 33901326 DOI: 10.1002/ajh.26207] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Approximately 20% of newly diagnosed multiple myeloma (NDMM) patients harbor t(11;14), a marker of inferior prognosis, resulting in up-regulation of CCND1. These patients respond to BCL2 inhibitor experimental drug venetoclax. Furthermore, t(11;14) is reported to be associated with increased BCL2/MCL1 ratio. We investigated the use of venetoclax (400 mg daily) in a cohort of 25 multiple myeloma (MM) and AL-amyloidosis patients harboring t(11;14) and assessed safety and efficacy. Efficacy was assessed by response rate (RR) and time on treatment. Furthermore, immunohistochemistry (IHC), for BCL2 family member expression was assessed at diagnosis and relapse in the venetoclax-treated group and analyzed for correlation with clinical RR. Additionally, patient material from venetoclax non-treated group including non-t(11;14) diagnosis (n = 27), t(11;14) diagnosis (n = 17), t(11;14) relapse (n = 7), hyperdiploidy (n = 6) and hyperdiploidy + t(11;14) (n = 6) was used for RNA sequencing (RNASeq) and validation by qPCR. Venetoclax treatment in t(11;14) patients demonstrated manageable safety and promising efficacy. Partial responses or better were observed in eleven patients (44%). Responding patients had significantly higher BCL2/MCL1 (p = 0.031) as well as BCL2/BCL-XL (p = 0.021) ratio, regardless of time of measurement before venetoclax treatment. Furthermore, an IRF5 motif was enriched (p < .001) in the downregulated genes in t(11;14) relapses vs diagnoses. The RR with single agent venetoclax was 71% in AL-amyloidosis and 33% in MM, and IHC proved useful in prediction of treatment outcome. We could also demonstrate possible resistance mechanisms of t(11;14), downregulation of IRF5 targeted genes, which can be exploited for therapeutic advantages.
Collapse
Affiliation(s)
- Hareth Nahi
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Muhammad Kashif
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
| | - Monika Klimkowska
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
- Department of Clinical Pathology and Cytology Karolinska University Hospital Stockholm Sweden
| | - Maria Karvouni
- Department of Medicine Karolinska Institutet Stockholm Sweden
| | - Ann Wallblom
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
| | - Charlotte Gran
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Clinical Chemistry Karolinska University Hospital Stockholm Sweden
| | - Julia Hauenstein
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Nicolai Frengen
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Charlotte Gustafsson
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Gabriel Afram
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Katarina Uttervall
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Johan Lund
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Robert Månsson
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | | | - Evren Alici
- Department of Medicine Karolinska Institutet Stockholm Sweden
| |
Collapse
|
8
|
Pisano M, Cheng Y, Sun F, Dhakal B, D’Souza A, Chhabra S, Knight JM, Rao S, Zhan F, Hari P, Janz S. Laboratory Mice - A Driving Force in Immunopathology and Immunotherapy Studies of Human Multiple Myeloma. Front Immunol 2021; 12:667054. [PMID: 34149703 PMCID: PMC8206561 DOI: 10.3389/fimmu.2021.667054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
Mouse models of human cancer provide an important research tool for elucidating the natural history of neoplastic growth and developing new treatment and prevention approaches. This is particularly true for multiple myeloma (MM), a common and largely incurable neoplasm of post-germinal center, immunoglobulin-producing B lymphocytes, called plasma cells, that reside in the hematopoietic bone marrow (BM) and cause osteolytic lesions and kidney failure among other forms of end-organ damage. The most widely used mouse models used to aid drug and immunotherapy development rely on in vivo propagation of human myeloma cells in immunodeficient hosts (xenografting) or myeloma-like mouse plasma cells in immunocompetent hosts (autografting). Both strategies have made and continue to make valuable contributions to preclinical myeloma, including immune research, yet are ill-suited for studies on tumor development (oncogenesis). Genetically engineered mouse models (GEMMs), such as the widely known Vκ*MYC, may overcome this shortcoming because plasma cell tumors (PCTs) develop de novo (spontaneously) in a highly predictable fashion and accurately recapitulate many hallmarks of human myeloma. Moreover, PCTs arise in an intact organism able to mount a complete innate and adaptive immune response and tumor development reproduces the natural course of human myelomagenesis, beginning with monoclonal gammopathy of undetermined significance (MGUS), progressing to smoldering myeloma (SMM), and eventually transitioning to frank neoplasia. Here we review the utility of transplantation-based and transgenic mouse models of human MM for research on immunopathology and -therapy of plasma cell malignancies, discuss strengths and weaknesses of different experimental approaches, and outline opportunities for closing knowledge gaps, improving the outcome of patients with myeloma, and working towards a cure.
Collapse
Affiliation(s)
- Michael Pisano
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Yan Cheng
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Fumou Sun
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Binod Dhakal
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anita D’Souza
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Saurabh Chhabra
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jennifer M. Knight
- Departments of Psychiatry, Medicine, and Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sridhar Rao
- Division of Hematology, Oncology and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, United States
| | - Fenghuang Zhan
- Myeloma Center, Department of Internal Medicine and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Parameswaran Hari
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Siegfried Janz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
9
|
Li SS, Dong YH, Liu ZP. Recent Advances in the Development of Casein Kinase 1 Inhibitors. Curr Med Chem 2021; 28:1585-1604. [PMID: 32660395 DOI: 10.2174/0929867327666200713185413] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/21/2020] [Accepted: 05/28/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The casein kinase 1 (CK1) family is involved in regulating many cellular processes, including membrane trafficking, DNA damage repair, cytoskeleton dynamics, cytoskeleton maintenance and apoptosis. CK1 isoforms, especially CK1δ and CK1ε have emerged as important therapeutic targets for severe disorders such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), familial advanced sleep phase syndrome and cancer. Due to the importance of CK1 for the pathogenesis of disorders, there are great interests in the development of CK1 inhibitors. METHODS Using SciFinder® as a tool, the publications about the biology of CK1 and the recent developments of CK1 inhibitors were surveyed with an exclusion of those published as patents. RESULTS This review presents the current state of knowledge on the development of CK1 inhibitors, including both synthetic small molecular inhibitors that were divided into 7 categories according to structural features, and the natural compounds. An overview of the advancement of CK1 inhibitors was given, with the introduction of various existing CK1 inhibitors, their inhibitory activities, and the structure-activity relationships. CONCLUSION Through physicochemical characterization and biological investigations, it is possible to understand the structure-activity relationship of CK1 inhibitors, which will contribute to better design and discovery of potent and selective CK1 inhibitors as potential agents for severe disorders such as AD, ALS and cancer.
Collapse
Affiliation(s)
- Sha-Sha Li
- Department of Medicinal Chemistry, Key laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yue-Hui Dong
- Jinan Vocational College of Nursing, Jinan 250102, China
| | - Zhao-Peng Liu
- Department of Medicinal Chemistry, Key laboratory of Chemical Biology (Ministry of Education), School of pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
10
|
Spinello Z, Fregnani A, Quotti Tubi L, Trentin L, Piazza F, Manni S. Targeting Protein Kinases in Blood Cancer: Focusing on CK1α and CK2. Int J Mol Sci 2021; 22:ijms22073716. [PMID: 33918307 PMCID: PMC8038136 DOI: 10.3390/ijms22073716] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
Disturbance of protein kinase activity may result in dramatic consequences that often lead to cancer development and progression. In tumors of blood origin, both tyrosine kinases and serine/threonine kinases are altered by different types of mutations, critically regulating cancer hallmarks. CK1α and CK2 are highly conserved, ubiquitously expressed and constitutively active pleiotropic kinases, which participate in multiple biological processes. The involvement of these kinases in solid and blood cancers is well documented. CK1α and CK2 are overactive in multiple myeloma, leukemias and lymphomas. Intriguingly, they are not required to the same degree for the viability of normal cells, corroborating the idea of “druggable” kinases. Different to other kinases, mutations on the gene encoding CK1α and CK2 are rare or not reported. Actually, these two kinases are outside the paradigm of oncogene addiction, since cancer cells’ dependency on these proteins resembles the phenomenon of “non-oncogene” addiction. In this review, we will summarize the general features of CK1α and CK2 and the most relevant oncogenic and stress-related signaling nodes, regulated by kinase phosphorylation, that may lead to tumor progression. Finally, we will report the current data, which support the positioning of these two kinases in the therapeutic scene of hematological cancers.
Collapse
Affiliation(s)
- Zaira Spinello
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Anna Fregnani
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
- Correspondence: (F.P.); (S.M.); Tel.: +39-049-792-3263 (F.P. & S.M.); Fax: +39-049-792-3250 (F.P. & S.M.)
| | - Sabrina Manni
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
- Correspondence: (F.P.); (S.M.); Tel.: +39-049-792-3263 (F.P. & S.M.); Fax: +39-049-792-3250 (F.P. & S.M.)
| |
Collapse
|
11
|
Gao HS, Lin SY, Han X, Xu HZ, Gao YL, Qin ZY. Casein kinase 1 (CK1) promotes the proliferation and metastasis of glioma cells via the phosphatidylinositol 3 kinase-matrix metalloproteinase 2 (AKT-MMP2) pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:659. [PMID: 33987357 PMCID: PMC8106055 DOI: 10.21037/atm-21-935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Glioma is a type of tumor that usually occurs in the adult central nervous system. Protein kinases have become important targets for oncotherapy since they are closely correlated with signal transduction. The role of the casein kinase 1 (CK1) gene in glioma remains to be fully elucidated. Methods The mRNA and protein expression of CK1 were analyzed by Realtime PCR, Western blot and immunohistochemistry. The cell behavior was assayed by MTT, Transwell and cell scratch methods. Cell cycle and cell apoptosis were performed by flow cytometer. Construction of stable cell line was completed by lentivirus infection. The nude mouse model was used for in vivo analysis on the role of CK1 by injecting the cells into subcutaneous tissue, tail vein and cerebral cortex. The prognostic role of CK1 in glioma was evaluated using Kaplan-Meier and Cox regression analyses. Results immunohistochemical staining demonstrated that the expression of CK1 in glioma samples was correlated with the grade of glioma. Survival analysis using Kaplan-Meier and multivariate analysis by Cox regression indicated that CK1 could be used as an independent prognostic marker for glioma. The methyl thiazolyl tetrazolium (MTT), transwell, and cell scratch assays demonstrated that the CK1 gene promoted cell proliferation and invasion through the phosphatidylinositol 3 kinase/matrix metalloproteinase 2 (AKT-MMP2) signaling pathway. In vivo experiments in mice also confirmed the ability of CK1 to enhance tumor proliferation and metastasis, with the metastatic site being the small intestine. Conclusions the expression of CK1 was correlated with glioma grade and patient survival and it may enhance glioma proliferation and metastasis via AKT-MMP2 pathway.
Collapse
Affiliation(s)
- Hua-Song Gao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - She-Yu Lin
- Department of Biological Sciences, School of Life Sciences, Nantong University, Nantong, China
| | - Xi Han
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Zhi Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Lu Gao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhi-Yong Qin
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Achieving effective and selective CK1 inhibitors through structure modification. Future Med Chem 2021; 13:505-528. [PMID: 33438471 DOI: 10.4155/fmc-2020-0215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Casein kinase 1 (CK1) is an extensively expressed serine/threonine kinase family, with six highly conserved isoforms of human CK1. Due to its involvement in many biological processes, CK1 is a promising target for several pathological states, including circadian sleep disorder, neurodegenerative diseases, cancer and inflammation. However, due to the structural similarities between the six CK1 members, the design of CK1 inhibitors is intricate. So far, no effective CK1 inhibitors are reported to reach clinical trials; thus, approaches to obtaining both selective and effective CK1 inhibitors are in great demand. Here we analyze several CK1 inhibitors that provide successful experience for structure-based drug design and rational structure modification, which could provide references for further drug design.
Collapse
|
13
|
Janovská P, Normant E, Miskin H, Bryja V. Targeting Casein Kinase 1 (CK1) in Hematological Cancers. Int J Mol Sci 2020; 21:E9026. [PMID: 33261128 PMCID: PMC7730698 DOI: 10.3390/ijms21239026] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
The casein kinase 1 enzymes (CK1) form a family of serine/threonine kinases with seven CK1 isoforms identified in humans. The most important substrates of CK1 kinases are proteins that act in the regulatory nodes essential for tumorigenesis of hematological malignancies. Among those, the most important are the functions of CK1s in the regulation of Wnt pathways, cell proliferation, apoptosis and autophagy. In this review we summarize the recent developments in the understanding of biology and therapeutic potential of the inhibition of CK1 isoforms in the pathogenesis of chronic lymphocytic leukemia (CLL), other non-Hodgkin lymphomas (NHL), myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and multiple myeloma (MM). CK1δ/ε inhibitors block CLL development in preclinical models via inhibition of WNT-5A/ROR1-driven non-canonical Wnt pathway. While no selective CK1 inhibitors have reached clinical stage to date, one dual PI3Kδ and CK1ε inhibitor, umbralisib, is currently in clinical trials for CLL and NHL patients. In MDS, AML and MM, inhibition of CK1α, acting via activation of p53 pathway, showed promising preclinical activities and the first CK1α inhibitor has now entered the clinical trials.
Collapse
Affiliation(s)
- Pavlína Janovská
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
| | | | - Hari Miskin
- TG Therapeutics, New York, NY 10014, USA; (E.N.); (H.M.)
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, 61265 Brno, Czech Republic
| |
Collapse
|
14
|
Martinez-Høyer S, Karsan A. Mechanisms of lenalidomide sensitivity and resistance. Exp Hematol 2020; 91:22-31. [PMID: 32976949 DOI: 10.1016/j.exphem.2020.09.196] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022]
Abstract
The discovery that the immunomodulatory imide drugs (IMiDs) possess antitumor properties revolutionized the treatment of specific types of hematological cancers. Since then, much progress has been made in understanding why the IMiDs are so efficient in targeting the malignant clones in difficult-to-treat diseases. Despite their efficacy, IMiD resistance arises eventually. Herein we summarize the mechanisms of sensitivity and resistance to lenalidomide in del(5q) myelodysplastic syndrome and multiple myeloma, two diseases in which these drugs are at the therapeutic frontline. Understanding the molecular and cellular mechanisms underlying IMiD efficacy and resistance may allow development of specific strategies to eliminate the malignant clone in otherwise incurable diseases.
Collapse
Affiliation(s)
- Sergio Martinez-Høyer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada; Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
15
|
Mogollón P, Díaz-Tejedor A, Algarín EM, Paíno T, Garayoa M, Ocio EM. Biological Background of Resistance to Current Standards of Care in Multiple Myeloma. Cells 2019; 8:cells8111432. [PMID: 31766279 PMCID: PMC6912619 DOI: 10.3390/cells8111432] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/03/2019] [Accepted: 11/09/2019] [Indexed: 12/16/2022] Open
Abstract
A high priority problem in multiple myeloma (MM) management is the development of resistance to administered therapies, with most myeloma patients facing successively shorter periods of response and relapse. Herewith, we review the current knowledge on the mechanisms of resistance to the standard backbones in MM treatment: proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies (mAbs). In some cases, strategies to overcome resistance have been discerned, and an effort should be made to evaluate whether resensitization to these agents is feasible in the clinical setting. Additionally, at a time in which we are moving towards precision medicine in MM, it is equally important to identify reliable and accurate biomarkers of sensitivity/refractoriness to these main therapeutic agents with the goal of having more efficacious treatments and, if possible, prevent the development of relapse.
Collapse
Affiliation(s)
- Pedro Mogollón
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Andrea Díaz-Tejedor
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Esperanza M. Algarín
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Teresa Paíno
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Mercedes Garayoa
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Enrique M. Ocio
- Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, 39008 Santander, Spain
- Correspondence: ; Tel.: +34-942202520
| |
Collapse
|
16
|
Small molecule modulators targeting protein kinase CK1 and CK2. Eur J Med Chem 2019; 181:111581. [DOI: 10.1016/j.ejmech.2019.111581] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 12/31/2022]
|
17
|
Qiu Q, Li M, Yang L, Tang M, Zheng L, Wang F, Qiu H, Liang C, Li N, Yi D, Yi Y, Pan C, Yang S, Chen L, Hu Y. Targeting glutaminase1 and synergizing with clinical drugs achieved more promising antitumor activity on multiple myeloma. Oncotarget 2019; 10:5993-6005. [PMID: 31666930 PMCID: PMC6800263 DOI: 10.18632/oncotarget.27243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/10/2019] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) pathogenesis remains incompletely understood and biomarkers predicting treatment response still remain lacking. Here we describe the rational mechanisms of combining targeting glautaminase1 (GLS1) with other chemo-reagents for MM treatment. Gls1 is highly expressed cMYC/KRAS12V-drived plasmacytoma (PCT) cells. Down-regulation of Gls1 with miRNAi in cMYC/KRAS12V-expressing BaF3 cells prevented them from growing independence of interleukin 3 (IL3). By using our cMYC/KRAS12V-transduced adoptive plasmacytoma mouse model, we found that Gls1 is involved in PCT pathogenesis. Down-regulation of Gls1 significantly prolonged the survival of PCT recipients. Knockdown of Gls1 increased the expression of Cdkn1a and Cdkn1b and decreased the expression of some critical oncogenes for cancer cell survival, such as c-Myc, Cdk4, and NfκB, as well as some genes which are essential for MM cell survival, such as Irf4, Prdm1, Csnk1α1, and Rassf5. Combination of Gls1 inhibition with LBH589, Bortezomib, or Lenalidomide significantly impaired tumor growth in a MM xenograft mouse model. Our data strongly suggest that Gls1 plays an important role for MM pathogenesis and that combination of GLS1 inhibitor with other MM therapy agents could benefit to MM patients.
Collapse
Affiliation(s)
- Qiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Mengyuan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Li Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Fang Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Huandi Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Cailing Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Ning Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Dongni Yi
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuyao Yi
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cong Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China.,Guizhou Normal College, Guiyang, China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yiguo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China.,Department of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Thomsen H, Chattopadhyay S, Weinhold N, Vodicka P, Vodickova L, Hoffmann P, Nöthen MM, Jöckel KH, Langer C, Hajek R, Hallmans G, Pettersson-Kymmer U, Ohlsson C, Späth F, Houlston R, Goldschmidt H, Hemminki K, Försti A. Genome-wide association study of monoclonal gammopathy of unknown significance (MGUS): comparison with multiple myeloma. Leukemia 2019; 33:1817-1821. [PMID: 30737484 DOI: 10.1038/s41375-019-0396-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/10/2019] [Accepted: 01/21/2019] [Indexed: 02/08/2023]
Affiliation(s)
- Hauke Thomsen
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany
| | - Subhayan Chattopadhyay
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Pavel Vodicka
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 00, Prague, Czech Republic
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University, Albertov 4, 128 00, Prague, Czech Republic
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605, Pilsen, Czech Republic
| | - Ludmila Vodickova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 00, Prague, Czech Republic
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University, Albertov 4, 128 00, Prague, Czech Republic
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605, Pilsen, Czech Republic
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Research Center, University of Bonn, Bonn, Germany
| | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Langer
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava, 17. listopadu 1790, 708 52, Ostrava, Czech Republic
| | - Göran Hallmans
- Department of Medical Biosciences/Pathology, University of Umea, Umea, Sweden
| | - Ulrika Pettersson-Kymmer
- Clinical Pharmacology, Department of Pharmacology and Clinical Neuroscience, Umea University, Umea, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Florentin Späth
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- National Centre of Tumor Diseases, Heidelberg, Germany
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany.
| |
Collapse
|
19
|
Carrino M, Quotti Tubi L, Fregnani A, Canovas Nunes S, Barilà G, Trentin L, Zambello R, Semenzato G, Manni S, Piazza F. Prosurvival autophagy is regulated by protein kinase CK1 alpha in multiple myeloma. Cell Death Discov 2019; 5:98. [PMID: 31123604 PMCID: PMC6529432 DOI: 10.1038/s41420-019-0179-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023] Open
Abstract
Multiple myeloma (MM) is a tumor of plasma cells (PCs). Due to the intense immunoglobulin secretion, PCs are prone to endoplasmic reticulum stress and activate several stress-managing pathways, including autophagy. Indeed, autophagy deregulation is maladaptive for MM cells, resulting in cell death. CK1α, a pro-survival kinase in MM, has recently been involved as a regulator of the autophagic flux and of the transcriptional competence of the autophagy-related transcription factor FOXO3a in several cancers. In this study, we investigated the role of CK1α in autophagy in MM. To study the autophagic flux we generated clones of MM cell lines expressing the mCherry-eGFP-LC3B fusion protein. We observed that CK1 inhibition with the chemical ATP-competitive CK1 α/δ inhibitor D4476 resulted in an impaired autophagic flux, likely due to an alteration of lysosomes acidification. However, D4476 caused the accumulation of the transcription factor FOXO3a in the nucleus, and this was paralleled by the upregulation of mRNA coding for autophagic genes. Surprisingly, silencing of CK1α by RNA interference triggered the autophagic flux. However, FOXO3a did not shuttle into the nucleus and the transcription of autophagy-related FOXO3a-dependent genes was not observed. Thus, while the chemical inhibition with the dual CK1α/δ inhibitor D4476 induced cell death as a consequence of an accumulation of ineffective autophagic vesicles, on the opposite, CK1α silencing, although it also determined apoptosis, triggered a full activation of the early autophagic flux, which was then not supported by the upregulation of autophagic genes. Taken together, our results indicate that the family of CK1 kinases may profoundly influence MM cells survival also through the modulation of the autophagic pathway.
Collapse
Affiliation(s)
- Marilena Carrino
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Laura Quotti Tubi
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Anna Fregnani
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sara Canovas Nunes
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy.,Boston Children's Hospital/Harvard Medical School, Boston, MA USA
| | - Gregorio Barilà
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Livio Trentin
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Renato Zambello
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Gianpietro Semenzato
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sabrina Manni
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Francesco Piazza
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
20
|
CK1α and IRF4 are essential and independent effectors of immunomodulatory drugs in primary effusion lymphoma. Blood 2018; 132:577-586. [PMID: 29954751 DOI: 10.1182/blood-2018-01-828418] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022] Open
Abstract
Primary effusion lymphoma (PEL) is an aggressive cancer with few treatment options. The immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide have recently been shown to kill PEL cell lines, and lenalidomide is in clinical trials against PEL. IMiDs bind to the CRL4CRBN E3 ubiquitin ligase complex, leading to the acquisition of the Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3), casein kinase 1 α (CK1α), and zinc finger protein 91 (ZFP91) as neosubstrates. IMiDs are effective against multiple myeloma because of degradation of IKZF1 and IKZF3 and the consequent loss of interferon regulatory factor 4 (IRF4) and MYC expression. Lenalidomide is also effective in chromosome 5q deletion-associated myelodysplastic syndrome as a result of degradation of CK1α. An essential IKZF1-IRF4-MYC axis has recently been proposed to underlie the toxicity of IMiDs in PEL. Here, we further investigate IMiD effectors in PEL cell lines, based on genome-wide CRISPR/Cas9 screens for essential human genes. These screens and extensive validation experiments show that, of the 4 neosubstrates, only CK1α is essential for the survival of PEL cell lines. In contrast, IKZF1 and IKZF3 are dispensable, individually or in combination. IRF4 was critical in all 8 PEL cell lines tested, and surprisingly, IMiDs triggered downregulation of IRF4 expression independently of both IKZF1 and IKZF3. Reexpression of CK1α and/or IRF4 partially rescued PEL cell lines from IMiD-mediated toxicity. In conclusion, IMiD toxicity in PEL cell lines is independent of IKZF1 and IKZF3 but proceeds through degradation of the neosubstrate CK1α and downregulation of IRF4.
Collapse
|
21
|
Jiang S, Zhang M, Sun J, Yang X. Casein kinase 1α: biological mechanisms and theranostic potential. Cell Commun Signal 2018; 16:23. [PMID: 29793495 PMCID: PMC5968562 DOI: 10.1186/s12964-018-0236-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/16/2018] [Indexed: 02/07/2023] Open
Abstract
Casein kinase 1α (CK1α) is a multifunctional protein belonging to the CK1 protein family that is conserved in eukaryotes from yeast to humans. It regulates signaling pathways related to membrane trafficking, cell cycle progression, chromosome segregation, apoptosis, autophagy, cell metabolism, and differentiation in development, circadian rhythm, and the immune response as well as neurodegeneration and cancer. Given its involvement in diverse cellular, physiological, and pathological processes, CK1α is a promising therapeutic target. In this review, we summarize what is known of the biological functions of CK1α, and provide an overview of existing challenges and potential opportunities for advancing theranostics.
Collapse
Affiliation(s)
- Shaojie Jiang
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, 310016, Hangzhou, China
| | - Miaofeng Zhang
- Department of Orthopaedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, 310016, Hangzhou, China
| | - Xiaoming Yang
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, 310016, Hangzhou, China. .,Image-Guided Bio-Molecular Intervention Research, Department of Radiology, University of Washington School of Medicine, Seattle, WA, 98109, USA.
| |
Collapse
|
22
|
Cohen K, Abadi U, Hercbergs A, Davis PJ, Ellis M, Ashur-Fabian O. The induction of myeloma cell death and DNA damage by tetrac, a thyroid hormone derivative. Endocr Relat Cancer 2018; 25:21-34. [PMID: 29018054 DOI: 10.1530/erc-17-0246] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 11/08/2022]
Abstract
Multiple myeloma (MM) is a plasma cell malignancy in which involvement of the thyroid hormone-integrin αvβ3 pathway was shown, and pharmacologic inhibition of this pathway is a rational approach to disease management. A thyroid hormone derivative, tetraiodothyroacetic acid (tetrac), which inhibits l-thyroxine (T4) and 3,5,3'-triiodo-l-thyronine (T3) binding to αvβ3 integrin, was studied in five MM cell lines and primary bone marrow (BM) MM cells. Tetrac inhibited MM cell proliferation (absolute cell number/viability) and induced caspase-dependent apoptosis (annexin-V/PI and cell cycle). Activation of caspase-9 and caspase-3 was further demonstrated. Moreover, DNA damage markers, ataxia-telangiectasia-mutated (ATM) kinase, poly ADP-ribose polymerase (PARP-1) and histone γH2AX were induced by tetrac. The various tetrac-initiated effects were attenuated by Arg-Gly-Asp (RGD) peptide, suggesting integrin involvement. Primary BM mononuclear cells were harvested from MM patients (n = 39) at various disease stages. Tetrac-induced apoptosis (12/17 samples) and sensitized the cytotoxic action of bortezomib (6/9 samples). Lastly, expression of plasma membrane integrin αvβ3 was shown not only in the malignant plasma clone, but also in other cell populations within the BM samples (n = 25). Tetrac is anti-proliferative and pro-apoptotic in MM and cells may offer a therapeutic approach for this disease.
Collapse
Affiliation(s)
- Keren Cohen
- Translational Hemato-Oncology LaboratoryThe Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel
- Department of Human Molecular Genetics and BiochemistryTel Aviv University, Tel Aviv, Israel
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
| | - Uri Abadi
- Translational Hemato-Oncology LaboratoryThe Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
| | | | - Paul J Davis
- Department of MedicineAlbany Medical College, Albany, New York, USA
| | - Martin Ellis
- Translational Hemato-Oncology LaboratoryThe Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
| | - Osnat Ashur-Fabian
- Translational Hemato-Oncology LaboratoryThe Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel
- Department of Human Molecular Genetics and BiochemistryTel Aviv University, Tel Aviv, Israel
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Manni S, Carrino M, Piazza F. Role of protein kinases CK1α and CK2 in multiple myeloma: regulation of pivotal survival and stress-managing pathways. J Hematol Oncol 2017; 10:157. [PMID: 28969692 PMCID: PMC5625791 DOI: 10.1186/s13045-017-0529-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/22/2017] [Indexed: 01/07/2023] Open
Abstract
Multiple myeloma (MM) is a malignant tumor of transformed plasma cells. MM pathogenesis is a multistep process. This cancer can occur de novo (rarely) or it can develop from monoclonal gammopathy of undetermined significance (most of the cases). MM can be asymptomatic (smoldering myeloma) or clinically active. Malignant plasma cells exploit intrinsic and extrinsic bone marrow microenvironment-derived growth signals. Upregulation of stress-coping pathways is also instrumental to maintain MM cell growth. The phylogenetically related Ser/Thr kinases CSNK1A1 (CK1α) and CSNK2 (CK2) have recently gained a growing importance in hematologic malignancies arising both from precursors and from mature blood cells. In multiple myeloma, CK1α or CK2 sustain oncogenic cascades, such as the PI3K/AKT, JAK/STAT, and NF-κB, as well as propel stress-related signaling that help in coping with different noxae. Data also suggest that these kinases modulate the delivery of growth factors and cytokines from the bone marrow stroma. The “non-oncogene addiction” phenotype generated by the increased activity of CK1α and CK2 in multiple myeloma contributes to malignant plasma cell proliferation and survival and represents an Achilles’ heel for the activity of small ATP competitive CK1α or CK2 inhibitors.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology Section, University of Padova, Via Giustiniani 2, 35128, Padova, Italy. .,Venetian Institute of Molecular Medicine, Padova, Italy.
| | - Marilena Carrino
- Department of Medicine, Hematology Section, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology Section, University of Padova, Via Giustiniani 2, 35128, Padova, Italy. .,Venetian Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
24
|
Manni S, Carrino M, Manzoni M, Gianesin K, Nunes SC, Costacurta M, Tubi LQ, Macaccaro P, Taiana E, Cabrelle A, Barilà G, Martines A, Zambello R, Bonaldi L, Trentin L, Neri A, Semenzato G, Piazza F. Inactivation of CK1α in multiple myeloma empowers drug cytotoxicity by affecting AKT and β-catenin survival signaling pathways. Oncotarget 2017; 8:14604-14619. [PMID: 28099937 PMCID: PMC5362429 DOI: 10.18632/oncotarget.14654] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/07/2017] [Indexed: 11/25/2022] Open
Abstract
Recent evidence indicates that protein kinase CK1α may support the growth of multiple myeloma (MM) plasma cells. Here, by analyzing a large cohort of MM cases, we found that high CK1α mRNA levels are virtually associated with all MM patients. Moreover, we provided functional evidence that CK1α activity is essential for malignant plasma cell survival even in the protective niche generated by co-cultures with bone marrow stromal cells. We demonstrated that CK1α inactivation, while toxic for myeloma cells, is dispensable for the survival of healthy B lymphocytes and stromal cells. Disruption of CK1α function in myeloma cells resulted in decreased Mdm2, increased p53 and p21 and reduced expression of β-catenin and AKT. These effects were mediated partially by p53 and caspase activity. Finally, we discovered that CK1α inactivation enhanced the cytotoxic effect of both bortezomib and lenalidomide. Overall, our study supports a role for CK1α as a potential therapeutic target in MM in combination with proteasome inhibitors and/or immunomodulatory drugs.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Marilena Carrino
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Martina Manzoni
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Ketty Gianesin
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Sara Canovas Nunes
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Matteo Costacurta
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Paolo Macaccaro
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Elisa Taiana
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Anna Cabrelle
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Gregorio Barilà
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Annalisa Martines
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS- Padova, Italy
| | - Renato Zambello
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Laura Bonaldi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS- Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
25
|
Lenalidomide increases human dendritic cell maturation in multiple myeloma patients targeting monocyte differentiation and modulating mesenchymal stromal cell inhibitory properties. Oncotarget 2017; 8:53053-53067. [PMID: 28881793 PMCID: PMC5581092 DOI: 10.18632/oncotarget.18085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/10/2017] [Indexed: 11/25/2022] Open
Abstract
The use of Lenalidomide (LEN), to reverse tumor-mediated immune suppression and amplify multiple myeloma-specific immunity is currently being explored. Particularly, LEN effects on dendritic cells (DCs) are still unclear. In this study, we investigated the potential effect of LEN on DC differentiation and activity. DCs were differentiated either from CD14+ cells obtained from patients with multiple myeloma or from a human monocytic cell line. LEN, at the concentration range reached in vivo, significantly increased the median intensity expression of HLA-DR, CD86 and CD209 by DCs derived from both bone marrow and peripheral myeloma monocytes and enhanced the production of Interleukin-8, C-C motif chemokine ligand (CCL) 2, CCL5 and tumor necrosis factor-α. Consistently, LEN pre-treated DCs showed an increased ability to stimulate autologous CD3+ cell proliferation. LEN effect on dendritic differentiation was associated with the degradation of the Cereblon-related factors Ikaros and Aiolos. Moreover, we showed that LEN also blunted mesenchymal stromal cell inhibitory effect on dendritic differentiation, inhibiting Casein Kinase-1α levels. Finally, in vitro data were confirmed in ex vivo cultures obtained from relapsed myeloma patients treated with LEN, showing a significant increase of DC differentiation from peripheral blood monocytes. In conclusion, LEN increased the expression of mature dendritic markers both directly and indirectly and enhanced DC ability to stimulate T cell proliferation and to release chemokines. This suggests a new possible mechanism by which LEN could exert its anti-myeloma activity.
Collapse
|
26
|
Cee VJ, Chavez F, Herberich B, Lanman BA, Pettus LH, Reed AB, Wu B, Wurz RP, Andrews KL, Chen J, Hickman D, Laszlo J, Lee MR, Guerrero N, Mattson BK, Nguyen Y, Mohr C, Rex K, Sastri CE, Wang P, Wu Q, Wu T, Xu Y, Zhou Y, Winston JT, Lipford JR, Tasker AS, Wang HL. Discovery and Optimization of Macrocyclic Quinoxaline-pyrrolo-dihydropiperidinones as Potent Pim-1/2 Kinase Inhibitors. ACS Med Chem Lett 2016; 7:408-12. [PMID: 27096050 DOI: 10.1021/acsmedchemlett.5b00403] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
The identification of Pim-1/2 kinase overexpression in B-cell malignancies suggests that Pim kinase inhibitors will have utility in the treatment of lymphoma, leukemia, and multiple myeloma. Starting from a moderately potent quinoxaline-dihydropyrrolopiperidinone lead, we recognized the potential for macrocyclization and developed a series of 13-membered macrocycles. The structure-activity relationships of the macrocyclic linker were systematically explored, leading to the identification of 9c as a potent, subnanomolar inhibitor of Pim-1 and -2. This molecule also potently inhibited Pim kinase activity in KMS-12-BM, a multiple myeloma cell line with relatively high endogenous levels of Pim-1/2, both in vitro (pBAD IC50 = 25 nM) and in vivo (pBAD EC50 = 30 nM, unbound), and a 100 mg/kg daily dose was found to completely arrest the growth of KMS-12-BM xenografts in mice.
Collapse
Affiliation(s)
- Victor J. Cee
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Frank Chavez
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Bradley Herberich
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Brian A. Lanman
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Liping H. Pettus
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Anthony B. Reed
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Bin Wu
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Ryan P. Wurz
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Kristin L. Andrews
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Jie Chen
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Dean Hickman
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Jimmy Laszlo
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Matthew R. Lee
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Nadia Guerrero
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Bethany K. Mattson
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Yen Nguyen
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Christopher Mohr
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Karen Rex
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Christine E. Sastri
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Paul Wang
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Qiong Wu
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Tian Wu
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Yang Xu
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Yihong Zhou
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Jeffrey T. Winston
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - J. Russell Lipford
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Andrew S. Tasker
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Hui-Ling Wang
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| |
Collapse
|
27
|
Abstract
INTRODUCTION The conventional term 'casein kinase' (CK) denotes three classes of kinases - CK1, CK2 and Golgi-CK (G-CK)/Fam20C (family with sequence similarity 20, member C) - sharing the ability to phoshorylate casein in vitro, but otherwise unrelated to each other. All CKs have been reported to be implicated in human diseases, and reviews individually dealing with the druggability of CK1 and CK2 are available. Our aim is to provide a comparative analysis of the three classes of CKs as therapeutic targets. AREAS COVERED CK2 is the CK for which implication in neoplasia is best documented, with the survival of cancer cells often relying on its overexpression. An ample variety of cell-permeable CK2 inhibitors have been developed, with a couple of these now in clinical trials. Isoform-specific CK1 inhibitors that are expected to play a beneficial role in oncology and neurodegeneration have been also developed. In contrast, the pathogenic potential of G-CK/Fam20C is caused by its loss of function. Activators of Fam20C, notably sphingolipids and their analogs, may prove beneficial in this respect. EXPERT OPINION Optimization of CK2 and CK1 inhibitors will prove useful to develop new therapeutic strategies for treating cancer and neurodegenerative disorders, while the design of potent activators of G-CK/Fam20C will provide a new tool in the fields of bio-mineralization and hypophosphatemic diseases.
Collapse
Affiliation(s)
- Giorgio Cozza
- a 1 University of Padova, Department of Biomedical Sciences , Via Ugo Bassi 58B, 35131 Padova, Italy
| | - Lorenzo A Pinna
- a 1 University of Padova, Department of Biomedical Sciences , Via Ugo Bassi 58B, 35131 Padova, Italy .,b 2 University of Padova, Department of Biomedical Sciences and CNR Institute of Neurosciences , Padova, Italy ;
| |
Collapse
|
28
|
Cheong JK, Zhang F, Chua PJ, Bay BH, Thorburn A, Virshup DM. Casein kinase 1α-dependent feedback loop controls autophagy in RAS-driven cancers. J Clin Invest 2015; 125:1401-18. [PMID: 25798617 DOI: 10.1172/jci78018] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/28/2015] [Indexed: 12/17/2022] Open
Abstract
Activating mutations in the RAS oncogene are common in cancer but are difficult to therapeutically target. RAS activation promotes autophagy, a highly regulated catabolic process that metabolically buffers cells in response to diverse stresses. Here we report that casein kinase 1α (CK1α), a ubiquitously expressed serine/threonine kinase, is a key negative regulator of oncogenic RAS-induced autophagy. Depletion or pharmacologic inhibition of CK1α enhanced autophagic flux in oncogenic RAS-driven human fibroblasts and multiple cancer cell lines. FOXO3A, a master longevity mediator that transcriptionally regulates diverse autophagy genes, was a critical target of CK1α, as depletion of CK1α reduced levels of phosphorylated FOXO3A and increased expression of FOXO3A-responsive genes. Oncogenic RAS increased CK1α protein abundance via activation of the PI3K/AKT/mTOR pathway. In turn, elevated levels of CK1α increased phosphorylation of nuclear FOXO3A, thereby inhibiting transactivation of genes critical for RAS-induced autophagy. In both RAS-driven cancer cells and murine xenograft models, pharmacologic CK1α inactivation synergized with lysosomotropic agents to inhibit growth and promote tumor cell death. Together, our results identify a kinase feedback loop that influences RAS-dependent autophagy and suggest that targeting CK1α-regulated autophagy offers a potential therapeutic opportunity to treat oncogenic RAS-driven cancers.
Collapse
|
29
|
Winkler BS, Oltmer F, Richter J, Bischof J, Xu P, Burster T, Leithäuser F, Knippschild U. CK1δ in lymphoma: gene expression and mutation analyses and validation of CK1δ kinase activity for therapeutic application. Front Cell Dev Biol 2015; 3:9. [PMID: 25750912 PMCID: PMC4335261 DOI: 10.3389/fcell.2015.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/31/2015] [Indexed: 01/04/2023] Open
Abstract
The prognosis of lymphoid neoplasms has improved considerably during the last decades. However, treatment response for some lymphoid neoplasms is still poor, indicating the need for new therapeutic approaches. One promising new strategy is the inhibition of kinases regulating key signal transduction pathways, which are of central importance in tumorigenesis. Kinases of the CK1 family may represent an attractive drug target since CK1 expression and/or activity are associated with the pathogenesis of malignant diseases. Over the last years efforts were taken to develop highly potent and selective CK1-specific inhibitor compounds and their therapeutic potential has now to be proved in pre-clinical trials. Therefore, we analyzed expression and mutational status of CK1δ in several cell lines representing established lymphoma entities, and also measured the mRNA expression level in primary lymphoma tissue as well as in non-neoplastic blood cells. For a selection of lymphoma cell lines we furthermore determined CK1δ kinase activity and demonstrated therapeutic potential of CK1-specific inhibitors as a putative therapeutic option in the treatment of lymphoid neoplasms.
Collapse
Affiliation(s)
| | - Franziska Oltmer
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital Ulm, Germany
| | - Julia Richter
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital Ulm, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital Ulm, Germany
| | - Pengfei Xu
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital Ulm, Germany
| | - Timo Burster
- Department of Neurosurgery, Ulm University Hospital Ulm, Germany
| | | | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital Ulm, Germany
| |
Collapse
|