1
|
Pérez-de-Oliveira ME, Wagner VP, Bingle CD, Vargas PA, Bingle L. Disruption of oncogenic pathways in mucoepidermoid carcinoma: CREB inhibitor 666.15 as a potential therapeutic agent. Oral Oncol 2024; 159:107029. [PMID: 39332274 DOI: 10.1016/j.oraloncology.2024.107029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/07/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVES Mucoepidermoid carcinoma (MEC) is the most common malignant salivary gland tumour with around 50 % of cases carrying the CRTC1-MAML2 translocation. The CREB pathway has been associated with the transforming activity of this translocation. The aim of this study was to determine the effects of CREB inhibition on MEC cell behaviour in vitro. MATERIAL AND METHODS Two translocation-positive (UM-HMC-2 and H292) and one translocation-negative (H253) MEC cell lines were treated with 666.15, a CREB inhibitor. Drug IC50 doses were determined for each cell line. Clonogenic and spheroid assays were used to assess survival, including percentage of cancer stem cells, and transwell and scratch assays evaluated invasive and migratory capacities, respectively. Immunofluorescence staining was used to determine E-cadherin expression. RESULTS CREB inhibition significantly reduced the number of surviving colonies and spheroids and delayed cell invasion in all cell lines, but this was more significant in the fusion positive, UM-HMC-2 cells. The expression of E-cadherin was significantly higher in treated UM-HMC-2 and H292 cells. CONCLUSION CREB inhibition with 666.15 impaired key MEC oncogenic behaviours associated with metastasis and drug resistance, including cell invasion and survival.
Collapse
Affiliation(s)
- Maria Eduarda Pérez-de-Oliveira
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Vivian Petersen Wagner
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom; Department of Oral Medicine, School of Dentistry, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Colin D Bingle
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil
| | - Lynne Bingle
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
2
|
Maslah N, Rety S, Bonnamy M, Aguinaga L, Huynh T, Parietti V, Giraudier S, Fenaux P, Cassinat B. Niclosamide combined to Azacitidine to target TP53-mutated MDS/AML cells. Leukemia 2024; 38:1630-1633. [PMID: 38777834 PMCID: PMC11216995 DOI: 10.1038/s41375-024-02281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Nabih Maslah
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France.
- APHP, Hopital Saint-Louis, Laboratoire de Biologie Cellulaire, Universite Paris Cite, Paris, France.
| | - Salome Rety
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
| | - Melina Bonnamy
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
| | - Lorea Aguinaga
- APHP, Hopital Saint-Louis, Service d'Hematologie Seniors, Universite Paris Cite, Paris, France
| | - Tony Huynh
- APHP, Hopital Saint-Louis, Service d'Hematologie Seniors, Universite Paris Cite, Paris, France
| | - Veronique Parietti
- Université Paris Cité, INSERM/CNRS, US53/UAR2030, Institut de Recherche Saint-Louis, Paris, France
| | - Stephane Giraudier
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
- APHP, Hopital Saint-Louis, Laboratoire de Biologie Cellulaire, Universite Paris Cite, Paris, France
| | - Pierre Fenaux
- APHP, Hopital Saint-Louis, Service d'Hematologie Seniors, Universite Paris Cite, Paris, France.
| | - Bruno Cassinat
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
- APHP, Hopital Saint-Louis, Laboratoire de Biologie Cellulaire, Universite Paris Cite, Paris, France
| |
Collapse
|
3
|
Murphy LA, Winters AC. Emerging and Future Targeted Therapies for Pediatric Acute Myeloid Leukemia: Targeting the Leukemia Stem Cells. Biomedicines 2023; 11:3248. [PMID: 38137469 PMCID: PMC10741170 DOI: 10.3390/biomedicines11123248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Acute myeloid leukemia (AML) is a rare subtype of acute leukemia in the pediatric and adolescent population but causes disproportionate morbidity and mortality in this age group. Standard chemotherapeutic regimens for AML have changed very little in the past 3-4 decades, but the addition of targeted agents in recent years has led to improved survival in select subsets of patients as well as a better biological understanding of the disease. Currently, one key paradigm of bench-to-bedside practice in the context of adult AML is the focus on leukemia stem cell (LSC)-targeted therapies. Here, we review current and emerging immunotherapies and other targeted agents that are in clinical use for pediatric AML through the lens of what is known (and not known) about their LSC-targeting capability. Based on a growing understanding of pediatric LSC biology, we also briefly discuss potential future agents on the horizon.
Collapse
Affiliation(s)
- Lindsey A. Murphy
- Department of Pediatrics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Amanda C. Winters
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Syamprasad NP, Madje N, Bachannagari J, Jannu AK, Jain S, Tene K, Shantanu PA, Naidu V, Chella N. Niclosamide nanocrystal for enhanced in-vivo efficacy against gastrointestinal stromal tumor via regulating EGFR/STAT-3/DR-4 axis. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
5
|
Wang Z, Ren J, Du J, Wang H, Liu J, Wang G. Niclosamide as a Promising Therapeutic Player in Human Cancer and Other Diseases. Int J Mol Sci 2022; 23:16116. [PMID: 36555754 PMCID: PMC9782559 DOI: 10.3390/ijms232416116] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/03/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Niclosamide is an FDA-approved anthelmintic drug for the treatment of parasitic infections. However, over the past few years, increasing evidence has shown that niclosamide could treat diseases beyond parasitic diseases, which include metabolic diseases, immune system diseases, bacterial and viral infections, asthma, arterial constriction, myopia, and cancer. Therefore, we systematically reviewed the pharmacological activities and therapeutic prospects of niclosamide in human disease and cancer and summarized the related molecular mechanisms and signaling pathways, indicating that niclosamide is a promising therapeutic player in various human diseases, including cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Guiling Wang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China
| |
Collapse
|
6
|
Jiang H, Li AM, Ye J. The magic bullet: Niclosamide. Front Oncol 2022; 12:1004978. [PMID: 36479072 PMCID: PMC9720275 DOI: 10.3389/fonc.2022.1004978] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/12/2022] [Indexed: 08/27/2023] Open
Abstract
The term 'magic bullet' is a scientific concept proposed by the German Nobel laureate Paul Ehrlich in 1907, describing a medicine that could specifically and efficiently target a disease without harming the body. Oncologists have been looking for a magic bullet for cancer therapy ever since. However, the current therapies for cancers-including chemotherapy, radiation therapy, hormone therapy, and targeted therapy-pose either pan-cytotoxicity or only single-target efficacy, precluding their ability to function as a magic bullet. Intriguingly, niclosamide, an FDA-approved drug for treating tapeworm infections with an excellent safety profile, displays broad anti-cancer activity in a variety of contexts. In particular, niclosamide inhibits multiple oncogenic pathways such as Wnt/β-catenin, Ras, Stat3, Notch, E2F-Myc, NF-κB, and mTOR and activates tumor suppressor signaling pathways such as p53, PP2A, and AMPK. Moreover, niclosamide potentially improves immunotherapy by modulating pathways such as PD-1/PDL-1. We recently discovered that niclosamide ethanolamine (NEN) reprograms cellular metabolism through its uncoupler function, consequently remodeling the cellular epigenetic landscape to promote differentiation. Inspired by the promising results from the pre-clinical studies, several clinical trials are ongoing to assess the therapeutic effect of niclosamide in cancer patients. This current review summarizes the functions, mechanism of action, and potential applications of niclosamide in cancer therapy as a magic bullet.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Albert M. Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
7
|
Ren J, Wang B, Wu Q, Wang G. Combination of niclosamide and current therapies to overcome resistance for cancer: New frontiers for an old drug. Biomed Pharmacother 2022; 155:113789. [DOI: 10.1016/j.biopha.2022.113789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/02/2022] Open
|
8
|
Lam SK, Yan S, Lam JSM, Feng Y, Khan M, Chen C, Ko FCF, Ho JCM. Disturbance of the Warburg effect by dichloroacetate and niclosamide suppresses the growth of different sub-types of malignant pleural mesothelioma in vitro and in vivo. Front Pharmacol 2022; 13:1020343. [PMID: 36304150 PMCID: PMC9592830 DOI: 10.3389/fphar.2022.1020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Inhalation of asbestos fibers is the most common cause of malignant pleural mesothelioma (MPM). In 2004, the United States Food and Drug Administration approved a combination of cisplatin with pemetrexed to treat unresectable MPM. Nonetheless novel treatment is urgently needed. The objective of this study is to report the combination effect of dichloroacetate (DCA) or niclosamide (Nic) Nic in MPM. Materials and methods: The effect of a combination of DCA and Nic was studied using a panel of MPM cell lines (H28, MSTO-211H, H226, H2052, and H2452). Cell viability was monitored by MTT assay. Glycolysis, oxidative phosphorylation, glucose, glycogen, pyruvate, lactate, citrate, succinate and ATP levels were determined by corresponding ELISA. Apoptosis, mitochondrial transmembrane potential, cell cycle analysis, hydrogen peroxide and superoxide were investigated by flow cytometry. Cell migration and colony formation were investigated by transwell migration and colony formation assays respectively. The in vivo effect was confirmed using 211H and H226 nude mice xenograft models. Results and conclusion: Cell viability was reduced. Disturbance of glycolysis and/or oxidative phosphorylation resulted in downregulation of glycogen, citrate and succinate. DCA and/or Nic increased apoptosis, mitochondrial transmembrane depolarization, G2/M arrest and reactive oxygen species. Moreover, DCA and/or Nic suppressed cell migration and colony formation. Furthermore, a better initial tumor suppressive effect was induced by the DCA/Nic combination compared with either drug alone in both 211H and H226 xenograft models. In H226 xenografts, DCA/Nic increased median survival of mice compared with single treatment. Single drug and/or a combination disturbed the Warburg effect and activated apoptosis, and inhibition of migration and proliferation in vivo. In conclusion, dichloroacetate and/or niclosamide showed a tumor suppressive effect in MPM in vitro and in vivo, partially mediated by disturbance of glycolysis/oxidative phosphorylation, apoptosis, ROS production, G2/M arrest, and suppression of migration and proliferation.
Collapse
|
9
|
Expressions of TGF-β1 and VEGF in patients with acute myeloid leukemia and associations with prognosis. REV ROMANA MED LAB 2022. [DOI: 10.2478/rrlm-2022-0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Background: To study the expressions of transforming growth factor-β1 (TGF-β1) and vascular endothelial growth factor (VEGF) in patients with acute myeloid leukemia (AML) and their values for prognosis.
Methods: A total of 120 AML patients treated from January 2015 to December 2018 were selected. Bone marrow mononuclear cells were isolated. The expressions of TGF-β1 and VEGF were detected by RT-PCR, and their associations with clinical characteristics were analyzed. The overall survival (OS) and disease-free survival (DFS) were assessed using the Kaplan-Meier method. The risk factors for prognosis were analyzed through the Cox proportional hazards model.
Results: The AML group had significantly lower relative expression of TGF-β1 and higher relative expression of VEGF than those of the control group (P<0.05). TGF-β1 and VEGF levels were significantly correlated with white blood cell count, hemoglobin, platelets, and peripheral blood juvenile cells (P<0.05). TGF-β1 level was higher and VEGF level was lower in the patients with complete response than those in the patients with partial response and no response (P<0.05). Both OS and DFS of the patients with high TGF-β1 expression were better than those of the patients with low TGF-β1 expression, while they were also superior among the patients with low VEGF expression (P<0.05). Platelets, TGF-β1 and VEGF were independent influencing factors for OS, and white blood cells, TGF-β1 and VEGF were independent influencing factors for DFS (P<0.05).
Conclusions: AML patients have decreased expression of TGF-β1 and increased expression of VEGF, and such changes are closely associated with the prognosis of AML.
Collapse
|
10
|
Ahmed MB, Alghamdi AAA, Islam SU, Lee JS, Lee YS. cAMP Signaling in Cancer: A PKA-CREB and EPAC-Centric Approach. Cells 2022; 11:cells11132020. [PMID: 35805104 PMCID: PMC9266045 DOI: 10.3390/cells11132020] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is one of the most common causes of death globally. Despite extensive research and considerable advances in cancer therapy, the fundamentals of the disease remain unclear. Understanding the key signaling mechanisms that cause cancer cell malignancy may help to uncover new pharmaco-targets. Cyclic adenosine monophosphate (cAMP) regulates various biological functions, including those in malignant cells. Understanding intracellular second messenger pathways is crucial for identifying downstream proteins involved in cancer growth and development. cAMP regulates cell signaling and a variety of physiological and pathological activities. There may be an impact on gene transcription from protein kinase A (PKA) as well as its downstream effectors, such as cAMP response element-binding protein (CREB). The position of CREB downstream of numerous growth signaling pathways implies its oncogenic potential in tumor cells. Tumor growth is associated with increased CREB expression and activation. PKA can be used as both an onco-drug target and a biomarker to find, identify, and stage tumors. Exploring cAMP effectors and their downstream pathways in cancer has become easier using exchange protein directly activated by cAMP (EPAC) modulators. This signaling system may inhibit or accelerate tumor growth depending on the tumor and its environment. As cAMP and its effectors are critical for cancer development, targeting them may be a useful cancer treatment strategy. Moreover, by reviewing the material from a distinct viewpoint, this review aims to give a knowledge of the impact of the cAMP signaling pathway and the related effectors on cancer incidence and development. These innovative insights seek to encourage the development of novel treatment techniques and new approaches.
Collapse
Affiliation(s)
- Muhammad Bilal Ahmed
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | | | - Salman Ul Islam
- Department of Pharmacy, Cecos University, Peshawar, Street 1, Sector F 5 Phase 6 Hayatabad, Peshawar 25000, Pakistan;
| | - Joon-Seok Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | - Young-Sup Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
- Correspondence: ; Tel.: +82-53-950-6353; Fax: +82-53-943-2762
| |
Collapse
|
11
|
Yan Y, Ding X, Han C, Gao J, Liu Z, Liu Y, Wang K. Involvement of TMEM16A/ANO1 upregulation in the oncogenesis of colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166370. [DOI: 10.1016/j.bbadis.2022.166370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022]
|
12
|
Pei X, Zheng F, Li Y, Lin Z, Han X, Feng Y, Tian Z, Ren D, Cao K, Li C. Niclosamide Ethanolamine Salt Alleviates Idiopathic Pulmonary Fibrosis by Modulating the PI3K-mTORC1 Pathway. Cells 2022; 11:cells11030346. [PMID: 35159160 PMCID: PMC8834116 DOI: 10.3390/cells11030346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/05/2022] [Accepted: 01/15/2022] [Indexed: 12/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial pneumonia characterized by chronic progressive fibrosis, ultimately leading to respiratory failure and early mortality. Although not fully explored, the major causative factors in IPF pathogenesis are dysregulated fibroblast proliferation and excessive accumulation of extracellular matrix (ECM) deposited by myofibroblasts differentiated from pulmonary fibroblasts. More signalling pathways, including the PI3K-Akt-mTOR and autophagy pathways, are involved in IPF pathogenesis. Niclosamide ethanolamine salt (NEN) is a highly effective multitarget small-molecule inhibitor reported in antitumor studies. Here, we reported that in an IPF animal model treated with NEN for 14 days, attractive relief of pulmonary function and hydroxyproline content were observed. To further explore, the therapeutic effect of NEN in IPF and pathological changes in bleomycin-challenged mouse lung sections were assessed. Additionally, the effects of NEN on abnormal proliferation and ECM production in IPF cell models established with TGF-β1-stimulated A549 cells or DHLF-IPF cells were studied. In nonclinical studies, NEN ameliorated lung function and histopathological changes in bleomycin-challenged mice, and the lung hydroxyproline content was significantly diminished with NEN treatment. In vitro, NEN inhibited PI3K-mTORC1 signalling and arrested the cell cycle to prevent uncontrolled fibroblast proliferation. Additionally, NEN inhibited TGF-β1-induced epithelial–mesenchymal transition (EMT) and ECM accumulation via the mTORC1-4EBP1 axis. Furthermore, NEN-activated noncanonical autophagy resensitized fibroblasts to apoptosis. The above findings demonstrated the potential antifibrotic effect of NEN mediated via modulation of the PI3K-mTORC1 and autophagy pathways. These data provide strong evidence for a therapeutic role for NEN in IPF.
Collapse
Affiliation(s)
- Xiaolin Pei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300350, China; (X.P.); (F.Z.); (Y.L.); (Z.L.); (X.H.); (Y.F.)
| | - Fangxu Zheng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300350, China; (X.P.); (F.Z.); (Y.L.); (Z.L.); (X.H.); (Y.F.)
| | - Yin Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300350, China; (X.P.); (F.Z.); (Y.L.); (Z.L.); (X.H.); (Y.F.)
| | - Zhoujun Lin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300350, China; (X.P.); (F.Z.); (Y.L.); (Z.L.); (X.H.); (Y.F.)
| | - Xiao Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300350, China; (X.P.); (F.Z.); (Y.L.); (Z.L.); (X.H.); (Y.F.)
| | - Ya Feng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300350, China; (X.P.); (F.Z.); (Y.L.); (Z.L.); (X.H.); (Y.F.)
| | - Zhenhuan Tian
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, China;
| | - Dunqiang Ren
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China;
| | - Ke Cao
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
- Correspondence: (K.C.); (C.L.)
| | - Chenggang Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300350, China; (X.P.); (F.Z.); (Y.L.); (Z.L.); (X.H.); (Y.F.)
- Correspondence: (K.C.); (C.L.)
| |
Collapse
|
13
|
Lohiya G, Katti DS. A Synergistic Combination of Niclosamide and Doxorubicin as an Efficacious Therapy for All Clinical Subtypes of Breast Cancer. Cancers (Basel) 2021; 13:cancers13133299. [PMID: 34209317 PMCID: PMC8268129 DOI: 10.3390/cancers13133299] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Chemotherapy is the gold standard treatment option for metastatic cancers. However, the efficacy of chemotherapy is limited due to the development of resistance. The aberrantly expressed Wnt/β-catenin signaling pathway acts as one of the major cancer drivers that also causes the development of resistance. Therefore, in this study, we explored the combinatorial approach of downregulating the Wnt/β-catenin pathway along with using a chemotherapeutic agent as a strategy to overcome drug resistance and improve cancer therapy. We evaluated the combinatorial efficacy of Niclosamide (an antihelminthic repurposed as a Wnt signaling inhibitor) and Doxorubicin (first-line treatment for multiple cancers in the clinic) against breast cancer. The combination showed synergistically enhanced death of all three clinical subtypes of breast cancer cells in both the sequential and concurrent treatment regimens and holds the potential to be developed as an efficient therapeutic option for breast cancer irrespective of its clinical subtype. Abstract Drug resistance is one of the major hurdles in the success of cancer chemotherapy. Notably, aberrantly expressed Wnt/β-catenin signaling plays a major role in the initiation and maintenance of oncogenesis along with development of chemoresistance. Therefore, the combinatorial approach of targeting Wnt/β-catenin pathway along with using a chemotherapeutic agent seems to be a promising strategy to improve cancer therapy. In the present study, we evaluated the combination of niclosamide (Nic), an FDA-approved antihelminthic drug repurposed as a Wnt signaling inhibitor, and doxorubicin (Dox), a conventional anticancer agent, in all clinical subtypes of breast cancer viz. triple negative breast cancer, HER2 positive breast cancer, and hormone receptor positive breast cancer. The results demonstrated that the combination induced apoptosis and caused synergistically enhanced death of all breast cancer cell types at multiple combinatorial concentrations using both the sequential and concurrent treatment regimens. Mechanistically, downregulation of Wnt/β-catenin signaling and cell cycle arrest at G0/G1 phase by Nic and increase in reactive oxygen species by both Nic and Dox along with the inherent cytotoxicity of Dox mediated the synergism between the two drugs in both the treatment regimens. Overall, the combination of Nic and Dox holds promise to be developed as an efficient therapeutic option for breast cancer irrespective of its clinical subtype.
Collapse
Affiliation(s)
- Garima Lohiya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India;
- Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Dhirendra S. Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India;
- Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Correspondence:
| |
Collapse
|
14
|
Fajardo-Orduña GR, Ledesma-Martínez E, Aguiñiga-Sánchez I, Mora-García MDL, Weiss-Steider B, Santiago-Osorio E. Inhibitors of Chemoresistance Pathways in Combination with Ara-C to Overcome Multidrug Resistance in AML. A Mini Review. Int J Mol Sci 2021; 22:ijms22094955. [PMID: 34066940 PMCID: PMC8124548 DOI: 10.3390/ijms22094955] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML), the most common type of leukemia in older adults, is a heterogeneous disease that originates from the clonal expansion of undifferentiated hematopoietic progenitor cells. These cells present a remarkable variety of genes and proteins with altered expression and function. Despite significant advances in understanding the molecular panorama of AML and the development of therapies that target mutations, survival has not improved significantly, and the therapy standard is still based on highly toxic chemotherapy, which includes cytarabine (Ara-C) and allogeneic hematopoietic cell transplantation. Approximately 60% of AML patients respond favorably to these treatments and go into complete remission; however, most eventually relapse, develop refractory disease or chemoresistance, and do not survive for more than five years. Therefore, drug resistance that initially occurs in leukemic cells (primary resistance) or that develops during or after treatment (acquired resistance) has become the main obstacle to AML treatment. In this work, the main molecules responsible for generating chemoresistance to Ara-C in AML are discussed, as well as some of the newer strategies to overcome it, such as the inclusion of molecules that can induce synergistic cytotoxicity with Ara-C (MNKI-8e, emodin, metformin and niclosamide), subtoxic concentrations of chemotherapy (PD0332991), and potently antineoplastic treatments that do not damage nonmalignant cells (heteronemin or hydroxyurea + azidothymidine).
Collapse
Affiliation(s)
- Guadalupe Rosario Fajardo-Orduña
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico; (G.R.F.-O.); (E.L.-M.); (I.A.-S.); (B.W.-S.)
| | - Edgar Ledesma-Martínez
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico; (G.R.F.-O.); (E.L.-M.); (I.A.-S.); (B.W.-S.)
| | - Itzen Aguiñiga-Sánchez
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico; (G.R.F.-O.); (E.L.-M.); (I.A.-S.); (B.W.-S.)
- Department of Biomedical Sciences, School of Medicine, Faculty of High Studies Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico
| | - María de Lourdes Mora-García
- Immunobiology Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico;
| | - Benny Weiss-Steider
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico; (G.R.F.-O.); (E.L.-M.); (I.A.-S.); (B.W.-S.)
| | - Edelmiro Santiago-Osorio
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico; (G.R.F.-O.); (E.L.-M.); (I.A.-S.); (B.W.-S.)
- Correspondence: ; Tel.: +52-55-57-73-41-08
| |
Collapse
|
15
|
Nathan S, Zhang H, Andreoli M, Leopold PL, Crystal RG. CREB-dependent LPA-induced signaling initiates a pro-fibrotic feedback loop between small airway basal cells and fibroblasts. Respir Res 2021; 22:97. [PMID: 33794877 PMCID: PMC8015171 DOI: 10.1186/s12931-021-01677-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/04/2021] [Indexed: 01/20/2023] Open
Abstract
Background Lysophosphatidic acid (LPA), generated extracellularly by the action of autotaxin and phospholipase A2, functions through LPA receptors (LPARs) or sphingosine-1-phosphate receptors (S1PRs) to induce pro-fibrotic signaling in the lower respiratory tract of patients with idiopathic pulmonary fibrosis (IPF). We hypothesized that LPA induces changes in small airway epithelial (SAE) basal cells (BC) that create cross-talk between the BC and normal human lung fibroblasts (NHLF), enhancing myofibroblast formation. Methods To assess LPA-induced signaling, BC were treated with LPA for 2.5 min and cell lysates were analyzed by phosphokinase array and Western blot. To assess transcriptional changes, BC were treated with LPA for 3 h and harvested for collection and analysis of RNA by quantitative polymerase chain reaction (qPCR). To assess signaling protein production and function, BC were washed thoroughly after LPA treatment and incubated for 24 h before collection for protein analysis by ELISA or functional analysis by transfer of conditioned medium to NHLF cultures. Transcription, protein production, and proliferation of NHLF were assessed. Results LPA treatment induced signaling by cAMP response element-binding protein (CREB), extracellular signal-related kinases 1 and 2 (Erk1/2), and epithelial growth factor receptor (EGFR) resulting in elevated expression of connective tissue growth factor (CTGF), endothelin-1 (EDN1/ET-1 protein), and platelet derived growth factor B (PDGFB) at the mRNA and protein levels. The conditioned medium from LPA-treated BC induced NHLF proliferation and increased NHLF expression of collagen I (COL1A1), smooth muscle actin (ACTA2), and autotaxin (ENPP2) at the mRNA and protein levels. Increased autotaxin secretion from NHLF correlated with increased LPA in the NHLF culture medium. Inhibition of CREB signaling blocked LPA-induced changes in BC transcription and translation as well as the pro-fibrotic effects of the conditioned medium on NHLF. Conclusion Inhibition of CREB signaling may represent a novel target for alleviating the LPA-induced pro-fibrotic feedback loop between SAE BC and NHLF. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01677-0.
Collapse
Affiliation(s)
- Shyam Nathan
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Haijun Zhang
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Mirko Andreoli
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Philip L Leopold
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA.
| |
Collapse
|
16
|
Chung I, Zhou K, Barrows C, Banyard J, Wilson A, Rummel N, Mizokami A, Basu S, Sengupta P, Shaikh B, Sengupta S, Bielenberg DR, Zetter BR. Unbiased Phenotype-Based Screen Identifies Therapeutic Agents Selective for Metastatic Prostate Cancer. Front Oncol 2021; 10:594141. [PMID: 33738243 PMCID: PMC7962607 DOI: 10.3389/fonc.2020.594141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023] Open
Abstract
In American men, prostate cancer is the second leading cause of cancer-related death. Dissemination of prostate cancer cells to distant organs significantly worsens patients' prognosis, and currently there are no effective treatment options that can cure advanced-stage prostate cancer. In an effort to identify compounds selective for metastatic prostate cancer cells over benign prostate cancer cells or normal prostate epithelial cells, we applied a phenotype-based in vitro drug screening method utilizing multiple prostate cancer cell lines to test 1,120 different compounds from a commercial drug library. Top drug candidates were then examined in multiple mouse xenograft models including subcutaneous tumor growth, experimental lung metastasis, and experimental bone metastasis assays. A subset of compounds including fenbendazole, fluspirilene, clofazimine, niclosamide, and suloctidil showed preferential cytotoxicity and apoptosis towards metastatic prostate cancer cells in vitro and in vivo. The bioavailability of the most discerning agents, especially fenbendazole and albendazole, was improved by formulating as micelles or nanoparticles. The enhanced forms of fenbendazole and albendazole significantly prolonged survival in mice bearing metastases, and albendazole-treated mice displayed significantly longer median survival times than paclitaxel-treated mice. Importantly, these drugs effectively targeted taxane-resistant tumors and bone metastases - two common clinical conditions in patients with aggressive prostate cancer. In summary, we find that metastatic prostate tumor cells differ from benign prostate tumor cells in their sensitivity to certain drug classes. Taken together, our results strongly suggest that albendazole, an anthelmintic medication, may represent a potential adjuvant or neoadjuvant to standard therapy in the treatment of disseminated prostate cancer.
Collapse
Affiliation(s)
- Ivy Chung
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Kun Zhou
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Courtney Barrows
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
| | - Jacqueline Banyard
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Arianne Wilson
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
| | - Nathan Rummel
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Washington, DC, United States
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Sudipta Basu
- Laboratory for Nanomedicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Poulomi Sengupta
- Laboratory for Nanomedicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Badaruddin Shaikh
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Washington, DC, United States
| | - Shiladitya Sengupta
- Laboratory for Nanomedicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Diane R. Bielenberg
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Bruce R. Zetter
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
Zhang H, Kong Q, Wang J, Jiang Y, Hua H. Complex roles of cAMP-PKA-CREB signaling in cancer. Exp Hematol Oncol 2020; 9:32. [PMID: 33292604 PMCID: PMC7684908 DOI: 10.1186/s40164-020-00191-1] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is the first discovered second messenger, which plays pivotal roles in cell signaling, and regulates many physiological and pathological processes. cAMP can regulate the transcription of various target genes, mainly through protein kinase A (PKA) and its downstream effectors such as cAMP-responsive element binding protein (CREB). In addition, PKA can phosphorylate many kinases such as Raf, GSK3 and FAK. Aberrant cAMP-PKA signaling is involved in various types of human tumors. Especially, cAMP signaling may have both tumor-suppressive and tumor-promoting roles depending on the tumor types and context. cAMP-PKA signaling can regulate cancer cell growth, migration, invasion and metabolism. This review highlights the important roles of cAMP-PKA-CREB signaling in tumorigenesis. The potential strategies to target this pathway for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Hongying Zhang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qingbin Kong
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
18
|
Steven A, Friedrich M, Jank P, Heimer N, Budczies J, Denkert C, Seliger B. What turns CREB on? And off? And why does it matter? Cell Mol Life Sci 2020; 77:4049-4067. [PMID: 32347317 PMCID: PMC7532970 DOI: 10.1007/s00018-020-03525-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/21/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
Altered expression and function of the transcription factor cyclic AMP response-binding protein (CREB) has been identified to play an important role in cancer and is associated with the overall survival and therapy response of tumor patients. This review focuses on the expression and activation of CREB under physiologic conditions and in tumors of distinct origin as well as the underlying mechanisms of CREB regulation by diverse stimuli and inhibitors. In addition, the clinical relevance of CREB is summarized, including its use as a prognostic and/or predictive marker as well as a therapeutic target.
Collapse
Affiliation(s)
- André Steven
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Michael Friedrich
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Paul Jank
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Nadine Heimer
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Jan Budczies
- Institute of Pathology, University Clinic Heidelberg, 69120, Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
| |
Collapse
|
19
|
Valli D, Gruszka AM, Alcalay M. Has Drug Repurposing Fulfilled its Promise in Acute Myeloid Leukaemia? J Clin Med 2020; 9:E1892. [PMID: 32560371 PMCID: PMC7356362 DOI: 10.3390/jcm9061892] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Drug repurposing is a method of drug discovery that consists of finding a new therapeutic context for an old drug. Compound identification arises from screening of large libraries of active compounds, through interrogating databases of cell line gene expression response upon treatment or by merging several types of information concerning disease-drug relationships. Although, there is a general consensus on the potential and advantages of this drug discovery modality, at the practical level to-date no non-anti-cancer repurposed compounds have been introduced into standard acute myeloid leukaemia (AML) management, albeit that preclinical validation yielded several candidates. The review presents the state-of-the-art drug repurposing approach in AML and poses the question of what has to be done in order to take a full advantage of it, both at the stage of screening design and later when progressing from the preclinical to the clinical phases of drug development. We argue that improvements are needed to model and read-out systems as well as to screening technologies, but also to more funding and trust in drug repurposing strategies.
Collapse
Affiliation(s)
- Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
| | - Alicja M. Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20 122 Milan, Italy
| |
Collapse
|
20
|
Illiano M, Conte M, Salzillo A, Ragone A, Spina A, Nebbioso A, Altucci L, Sapio L, Naviglio S. The KDM Inhibitor GSKJ4 Triggers CREB Downregulation via a Protein Kinase A and Proteasome-Dependent Mechanism in Human Acute Myeloid Leukemia Cells. Front Oncol 2020; 10:799. [PMID: 32582541 PMCID: PMC7289982 DOI: 10.3389/fonc.2020.00799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/23/2020] [Indexed: 01/02/2023] Open
Abstract
Acute myeloid leukemia (AML) is a progressive hematopoietic-derived cancer arising from stepwise genetic mutations of the myeloid lineage. cAMP response element-binding protein (CREB) is a nuclear transcription factor, which plays a key role in the multistep process of leukemogenesis, thus emerging as an attractive potential drug target for AML treatment. Since epigenetic dysregulations, such as DNA methylation, histone modifications, as well as chromatin remodeling, are a frequent occurrence in AML, an increasing and selective number of epi-drugs are emerging as encouraging therapeutic agents. Here, we demonstrate that the histone lysine demethylases (KDMs) JMJD3/UTX inhibitor GSKJ4 results in both proliferation decrease and CREB protein downregulation in AML cells. We found that GSKJ4 clearly decreases CREB protein, but not CREB mRNA levels. By cycloheximide assay, we provide evidence that GSKJ4 reduces CREB protein stability; moreover, proteasome inhibition largely counteracts the GSKJ4-induced CREB downregulation. Very interestingly, a rapid CREB phosphorylation at the Ser133 residue precedes CREB protein decrease in response to GSKJ4 treatment. In addition, protein kinase A (PKA) inhibition, but not extracellular signal-regulated kinase (ERK)1/2 inhibition, almost completely prevents both GSKJ4-induced p-Ser133-CREB phosphorylation and CREB protein downregulation. Overall, our study enforces the evidence regarding CREB as a potential druggable target, identifies the small epigenetic molecule GSKJ4 as an “inhibitor” of CREB, and encourages the design of future GSKJ4-based studies for the development of innovative approaches for AML therapy.
Collapse
Affiliation(s)
- Michela Illiano
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alessia Salzillo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Angela Ragone
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annamaria Spina
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luigi Sapio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Silvio Naviglio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
21
|
Fu Q, Jin X, Zhang Z, Lv H. Preparation and in vitro antitumor effects on MDA-MB-231 cells of niclosamide nanocrystals stabilized by poloxamer188 and PBS. Int J Pharm 2020; 584:119432. [DOI: 10.1016/j.ijpharm.2020.119432] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 01/17/2023]
|
22
|
Wojcicki AV, Kadapakkam M, Frymoyer A, Lacayo N, Chae HD, Sakamoto KM. Repurposing Drugs for Acute Myeloid Leukemia: A Worthy Cause or a Futile Pursuit? Cancers (Basel) 2020; 12:cancers12020441. [PMID: 32069925 PMCID: PMC7072462 DOI: 10.3390/cancers12020441] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clinically and genetically heterogenous malignancy of myeloid progenitor cells that affects patients of all ages. Despite decades of research and improvement in overall outcomes, standard therapy remains ineffective for certain subtypes of AML. Current treatment is intensive and leads to a number of secondary effects with varying results by patient population. Due to the high cost of discovery and an unmet need for new targeted therapies that are well tolerated, alternative drug development strategies have become increasingly attractive. Repurposing existing drugs is one approach to identify new therapies with fewer financial and regulatory hurdles. In this review, we provide an overview of previously U.S. Food and Drug Administration (FDA) approved non-chemotherapy drugs under investigation for the treatment of AML.
Collapse
Affiliation(s)
- Anna V. Wojcicki
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.V.W.); (M.K.); (N.L.); (H.-D.C.)
| | - Meena Kadapakkam
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.V.W.); (M.K.); (N.L.); (H.-D.C.)
| | - Adam Frymoyer
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Norman Lacayo
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.V.W.); (M.K.); (N.L.); (H.-D.C.)
| | - Hee-Don Chae
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.V.W.); (M.K.); (N.L.); (H.-D.C.)
| | - Kathleen M. Sakamoto
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.V.W.); (M.K.); (N.L.); (H.-D.C.)
- Correspondence: ; Tel.: +650-725-7126
| |
Collapse
|
23
|
Chae HD, Cox N, Capolicchio S, Lee JW, Horikoshi N, Kam S, Ng AA, Edwards J, Butler TL, Chan J, Lee Y, Potter G, Capece MC, Liu CW, Wakatsuki S, Smith M, Sakamoto KM. SAR optimization studies on modified salicylamides as a potential treatment for acute myeloid leukemia through inhibition of the CREB pathway. Bioorg Med Chem Lett 2019; 29:2307-2315. [PMID: 31253529 DOI: 10.1016/j.bmcl.2019.06.023] [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: 04/23/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 10/26/2022]
Abstract
Disruption of cyclic adenosine monophosphate response element binding protein (CREB) provides a potential new strategy to address acute leukemia, a disease associated with poor prognosis, and for which conventional treatment options often carry a significant risk of morbidity and mortality. We describe the structure-activity relationships (SAR) for a series of XX-650-23 derived from naphthol AS-E phosphate that disrupts binding and activation of CREB by the CREB-binding protein (CBP). Through the development of this series, we identified several salicylamides that are potent inhibitors of acute leukemia cell viability through inhibition of CREB-CBP interaction. Among them, a biphenyl salicylamide, compound 71, was identified as a potent inhibitor of CREB-CBP interaction with improved physicochemical properties relative to previously described derivatives of naphthol AS-E phosphate.
Collapse
Affiliation(s)
- Hee-Don Chae
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nick Cox
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA; Presently at Novo Nordisk Research Center Seattle, Inc., USA
| | | | - Jae Wook Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Naoki Horikoshi
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Sharon Kam
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew A Ng
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Jeffrey Edwards
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tae-León Butler
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Justin Chan
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yvonne Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Garrett Potter
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Mark C Capece
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Corey W Liu
- Macromolecular Structure Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Soichi Wakatsuki
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA; BioSciences Division, SLAC National Accelerator Laboratory, Menlo Park, CA, USA
| | - Mark Smith
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA.
| | - Kathleen M Sakamoto
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|