1
|
Corrales-Guerrero S, Cui T, Castro-Aceituno V, Yang L, Nair S, Feng H, Venere M, Yoon S, DeWees T, Shen C, Williams TM. Inhibition of RRM2 radiosensitizes glioblastoma and uncovers synthetic lethality in combination with targeting CHK1. Cancer Lett 2023; 570:216308. [PMID: 37482342 DOI: 10.1016/j.canlet.2023.216308] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/29/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
Glioblastoma (GBM) is an aggressive malignant primary brain tumor. Radioresistance largely contributes to poor clinical outcomes in GBM patients. We targeted ribonucleotide reductase subunit 2 (RRM2) with triapine to radiosensitize GBM. We found RRM2 is associated with increasing tumor grade, is overexpressed in GBM over lower grade gliomas and normal tissue, and is associated with worse survival. We found silencing or inhibition of RRM2 by siRNA or triapine sensitized GBM cells to ionizing radiation (IR) and delayed resolution of IR-induced γ-H2AX nuclear foci. In vivo, triapine and IR reduced tumor growth and increased mouse survival. Intriguingly, triapine led to RRM2 upregulation and CHK1 activation, suggesting a CHK1-dependent RRM2 upregulation following RRM2 inhibition. Consistently, silencing or inhibition of CHK1 with rabusertib abolished the triapine-induced RRM2 upregulation. Accordingly, combining rabusertib and triapine resulted in synthetic lethality in GBM cells. Collectively, our results suggest RRM2 is a promising therapeutic target for GBM, and targeting RRM2 with triapine sensitizes GBM cells to radiation and independently induces synthetic lethality of GBM cells with CHK1 inhibition. Our findings suggest combining triapine with radiation or rabusertib may improve therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Sergio Corrales-Guerrero
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Tiantian Cui
- Department of Radiation Oncology, City of Hope, Duarte, CA, USA
| | | | - Linlin Yang
- Department of Radiation Oncology, City of Hope, Duarte, CA, USA
| | - Sindhu Nair
- Department of Radiation Oncology, City of Hope, Duarte, CA, USA
| | - Haihua Feng
- Department of Radiation Oncology, City of Hope, Duarte, CA, USA
| | - Monica Venere
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Stephanie Yoon
- Department of Radiation Oncology, City of Hope, Duarte, CA, USA
| | - Todd DeWees
- Division of Biostatistics, City of Hope, Duarte, CA, USA
| | - Changxian Shen
- Department of Radiation Oncology, City of Hope, Duarte, CA, USA
| | | |
Collapse
|
2
|
Lai J, Yang S, Lin Z, Huang W, Li X, Li R, Tan J, Wang W. Update on Chemoresistance Mechanisms to First-Line Chemotherapy for Gallbladder Cancer and Potential Reversal Strategies. Am J Clin Oncol 2023; 46:131-141. [PMID: 36867653 PMCID: PMC10030176 DOI: 10.1097/coc.0000000000000989] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
OBJECTIVE Gallbladder cancer (GBC) mortality remains high and chemoresistance is increasing. This review consolidates what is known about the mechanisms of chemoresistance to inform and accelerate the development of novel GBC-specific chemotherapies. METHODS Studies related to GBC-related chemoresistance were systematically screened in PubMed using the advanced search function. Search terms included GBC, chemotherapy, and signaling pathway. RESULTS Analysis of existing studies showed that GBC has poor sensitivity to cisplatin, gemcitabine (GEM), and 5-fluorouracil. DNA damage repair-related proteins, including CHK1, V-SCR, and H2AX, are involved in tumor adaptation to drugs. GBC-specific chemoresistance is often accompanied by changes in the apoptosis and autophagy-related molecules, BCL-2, CRT, and GBCDRlnc1. CD44 + and CD133 + GBC cells are less resistant to GEM, indicating that tumor stem cells are also involved in chemoresistance. In addition, glucose metabolism, fat synthesis, and glutathione metabolism can influence the development of drug resistance. Finally, chemosensitizers such as lovastatin, tamoxifen, chloroquine, and verapamil are able improve the therapeutic effect of cisplatin or GEM in GBC. CONCLUSIONS This review summarizes recent experimental and clinical studies of the molecular mechanisms of chemoresistance, including autophagy, DNA damage, tumor stem cells, mitochondrial function, and metabolism, in GBC. Information on potential chemosensitizers is also discussed. The proposed strategies to reverse chemoresistance should inform the clinical use of chemosensitizers and gene-based targeted therapy for this disease.
Collapse
Affiliation(s)
- Jinbao Lai
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| | - Songlin Yang
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| | - Zhuying Lin
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| | - Wenwen Huang
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| | - Xiao Li
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| | - Ruhong Li
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| | - Jing Tan
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| | - Wenju Wang
- Yan’an Affiliated Hospital of Kunming Medical University
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province
- Kunming Key Laboratory of Biotherapy, Kunming, Yunnan, China
| |
Collapse
|
3
|
Sturm N, Schuhbaur JS, Hüttner F, Perkhofer L, Ettrich TJ. Gallbladder Cancer: Current Multimodality Treatment Concepts and Future Directions. Cancers (Basel) 2022; 14:5580. [PMID: 36428670 PMCID: PMC9688543 DOI: 10.3390/cancers14225580] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Gallbladder cancer (GBC) is the most common primary tumor site of biliary tract cancer (BTC), accounting for 0.6% of newly diagnosed cancers and 0.9% of cancer-related deaths. Risk factors, including female sex, age, ethnic background, and chronic inflammation of the gallbladder, have been identified. Surgery is the only curative option for early-stage GBC, but only 10% of patients are primary eligible for curative treatment. After neoadjuvant treatment, up to one-third of locally advanced GBC patients could benefit from secondary surgical treatment. After surgery, only a high-risk subset of patients benefits from adjuvant treatment. For advanced-stage GBC, palliative chemotherapy with gemcitabine and cisplatin is the current standard of care in line with other BTCs. After the failure of gemcitabine and cisplatin, data for second-line treatment in non-resectable GBC is poor, and the only recommended chemotherapy regimen is FOLFOX (5-FU/folinic acid and oxaliplatin). Recent advances with the PD-L1 inhibitor durvalumab open the therapy landscape for immune checkpoint inhibition in GBC. Meanwhile, targeted therapy approaches are a cornerstone of GBC therapy based on molecular profiling and new evidence of molecular differences between different BTC forms and might further improve the prognosis of GBC patients.
Collapse
Affiliation(s)
- Niklas Sturm
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany
| | | | - Felix Hüttner
- Department of General and Visceral Surgery, Ulm University Hospital, 89081 Ulm, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany
| | - Thomas Jens Ettrich
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany
| |
Collapse
|
4
|
Tsui M, Min W, Ng S, Dobbs K, Notarangelo LD, Dror Y, Grunebaum E. The Use of Induced Pluripotent Stem Cells to Study the Effects of Adenosine Deaminase Deficiency on Human Neutrophil Development. Front Immunol 2021; 12:748519. [PMID: 34777360 PMCID: PMC8582638 DOI: 10.3389/fimmu.2021.748519] [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: 07/28/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Inherited defects that abrogate the function of the adenosine deaminase (ADA) enzyme and consequently lead to the accumulation of toxic purine metabolites cause profound lymphopenia and severe combined immune deficiency. Additionally, neutropenia and impaired neutrophil function have been reported among ADA-deficient patients. However, due to the rarity of the disorder, the neutrophil developmental abnormalities and the mechanisms contributing to them have not been characterized. Induced pluripotent stem cells (iPSC) generated from two unrelated ADA-deficient patients and from healthy controls were differentiated through embryoid bodies into neutrophils. ADA deficiency led to a significant reduction in the number of all early multipotent hematopoietic progenitors. At later stages of differentiation, ADA deficiency impeded the formation of granulocyte colonies in methylcellulose cultures, leading to a significant decrease in the number of neutrophils generated from ADA-deficient iPSCs. The viability and apoptosis of ADA-deficient neutrophils isolated from methylcellulose cultures were unaffected, suggesting that the abnormal purine homeostasis in this condition interferes with differentiation or proliferation. Additionally, there was a significant increase in the percentage of hyperlobular ADA-deficient neutrophils, and these neutrophils demonstrated significantly reduced ability to phagocytize fluorescent microspheres. Supplementing iPSCs and methylcellulose cultures with exogenous ADA, which can correct adenosine metabolism, reversed all abnormalities, cementing the critical role of ADA in neutrophil development. Moreover, chemical inhibition of the ribonucleotide reductase (RNR) enzyme, using hydroxyurea or a combination of nicotinamide and trichostatin A in iPSCs from healthy controls, led to abnormal neutrophil differentiation similar to that observed in ADA deficiency, implicating RNR inhibition as a potential mechanism for the neutrophil abnormalities. In conclusion, the findings presented here demonstrate the important role of ADA in the development and function of neutrophils while clarifying the mechanisms responsible for the neutrophil abnormalities in ADA-deficient patients.
Collapse
Affiliation(s)
- Michael Tsui
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,The Institute of Medical Sciences, The University of Toronto, Toronto, ON, Canada
| | - Weixian Min
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Stephanie Ng
- The Institute of Medical Sciences, The University of Toronto, Toronto, ON, Canada.,Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yigal Dror
- The Institute of Medical Sciences, The University of Toronto, Toronto, ON, Canada.,Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Marrow Failure and Myelodysplasia Program, Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Eyal Grunebaum
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,The Institute of Medical Sciences, The University of Toronto, Toronto, ON, Canada.,Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
5
|
Lavanya M, Haribabu J, Ramaiah K, Suresh Yadav C, Kumar Chitumalla R, Jang J, Karvembu R, Varada Reddy A, Jagadeesh M. 2′-Thiophenecarboxaldehyde derived thiosemicarbazone metal complexes of copper(II), palladium(II) and zinc(II) ions: Synthesis, spectroscopic characterization, anticancer activity and DNA binding studies. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Babak MV, Ahn D. Modulation of Intracellular Copper Levels as the Mechanism of Action of Anticancer Copper Complexes: Clinical Relevance. Biomedicines 2021; 9:biomedicines9080852. [PMID: 34440056 PMCID: PMC8389626 DOI: 10.3390/biomedicines9080852] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
Copper (Cu) is a vital element required for cellular growth and development; however, even slight changes in its homeostasis might lead to severe toxicity and deleterious medical conditions. Cancer patients are typically associated with higher Cu content in serum and tumor tissues, indicating increased demand of cancer cells for this micronutrient. Cu is known to readily cycle between the +1 and +2 oxidation state in biological systems. The mechanism of action of Cu complexes is typically based on their redox activity and induction of reactive oxygen species (ROS), leading to deadly oxidative stress. However, there are a number of other biomolecular mechanisms beyond ROS generation that contribute to the activity of anticancer Cu drug candidates. In this review, we discuss how interfering with intracellular Cu balance via either diet modification or addition of inorganic Cu supplements or Cu-modulating compounds affects tumor development, progression, and sensitivity to treatment modalities. We aim to provide the rationale for the use of Cu-depleting and Cu-overloading conditions to generate the best possible patient outcome with minimal toxicity. We also discuss the advantages of the use of pre-formed Cu complexes, such as Cu-(bis)thiosemicarbazones or Cu-N-heterocyclic thiosemicarbazones, in comparison with the in situ formed Cu complexes with metal-binding ligands. In this review, we summarize available clinical and mechanistic data on clinically relevant anticancer drug candidates, including Cu supplements, Cu chelators, Cu ionophores, and Cu complexes.
Collapse
|
7
|
Abstract
Cancer cells accumulate iron to supplement their aberrant growth and metabolism. Depleting cells of iron by iron chelators has been shown to be selectively cytotoxic to cancer cells in vitro and in vivo. Iron chelators are effective at combating a range of cancers including those which are difficult to treat such as androgen insensitive prostate cancer and cancer stem cells. This review will evaluate the impact of iron chelation on cancer cell survival and the underlying mechanisms of action. A plethora of studies have shown iron chelators can reverse some of the major hallmarks and enabling characteristics of cancer. Iron chelators inhibit signalling pathways that drive proliferation, migration and metastasis as well as return tumour suppressive signalling. In addition to this, iron chelators stimulate apoptotic and ER stress signalling pathways inducing cell death even in cells lacking a functional p53 gene. Iron chelators can sensitise cancer cells to PARP inhibitors through mimicking BRCAness; a feature of cancers trademark genomic instability. Iron chelators target cancer cell metabolism, attenuating oxidative phosphorylation and glycolysis. Moreover, iron chelators may reverse the major characteristics of oncogenic transformation. Iron chelation therefore represent a promising selective mode of cancer therapy.
Collapse
|
8
|
Shakya B, Yadav PN. Thiosemicarbazones as Potent Anticancer Agents and their Modes of Action. Mini Rev Med Chem 2020; 20:638-661. [DOI: 10.2174/1389557519666191029130310] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/17/2019] [Accepted: 10/03/2019] [Indexed: 11/22/2022]
Abstract
:Thiosemicarbazones (TSCs) are a class of Schiff bases usually obtained by the condensation of thiosemicarbazide with a suitable aldehyde or ketone. TSCs have been the focus of chemists and biologists due to their wide range of pharmacological effects. One of the promising areas in which these excellent metal chelators are being developed is their use against cancer. TSCs have a wide clinical antitumor spectrum with efficacy in various tumor types such as leukemia, pancreatic cancer, breast cancer, non-small cell lung cancer, cervical cancer, prostate cancer and bladder cancer. To obtain better activity, different series of TSCs have been developed by modifying the heteroaromatic system in their molecules. These compounds possessed significant antineoplastic activity when the carbonyl attachment of the side chain was located at a position α to the ring nitrogen atom, whereas attachment of the side chain β or γ to the heterocyclic N atom resulted in inactive antitumor agents. In addition, replacement of the heterocyclic ring N with C also resulted in a biologically inactive compound suggesting that a conjugated N,N,S-tridentate donor set is essential for the biological activities of thiosemicarbazones. Several possible mechanisms have been implemented for the anticancer activity of thiosemicarbazones.
Collapse
Affiliation(s)
- Bhushan Shakya
- Amrit Campus, Tribhuvan University, Thamel, Kathmandu, Nepal
| | - Paras Nath Yadav
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| |
Collapse
|
9
|
Brown RAM, Richardson KL, Kabir TD, Trinder D, Ganss R, Leedman PJ. Altered Iron Metabolism and Impact in Cancer Biology, Metastasis, and Immunology. Front Oncol 2020; 10:476. [PMID: 32328462 PMCID: PMC7160331 DOI: 10.3389/fonc.2020.00476] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Iron is an essential nutrient that plays a complex role in cancer biology. Iron metabolism must be tightly controlled within cells. Whilst fundamental to many cellular processes and required for cell survival, excess labile iron is toxic to cells. Increased iron metabolism is associated with malignant transformation, cancer progression, drug resistance and immune evasion. Depleting intracellular iron stores, either with the use of iron chelating agents or mimicking endogenous regulation mechanisms, such as microRNAs, present attractive therapeutic opportunities, some of which are currently under clinical investigation. Alternatively, iron overload can result in a form of regulated cell death, ferroptosis, which can be activated in cancer cells presenting an alternative anti-cancer strategy. This review focuses on alterations in iron metabolism that enable cancer cells to meet metabolic demands required during different stages of tumorigenesis in relation to metastasis and immune response. The strength of current evidence is considered, gaps in knowledge are highlighted and controversies relating to the role of iron and therapeutic targeting potential are discussed. The key question we address within this review is whether iron modulation represents a useful approach for treating metastatic disease and whether it could be employed in combination with existing targeted drugs and immune-based therapies to enhance their efficacy.
Collapse
Affiliation(s)
- Rikki A. M. Brown
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| | - Kirsty L. Richardson
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Tasnuva D. Kabir
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Debbie Trinder
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| | - Ruth Ganss
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Peter J. Leedman
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
10
|
Miklos W, Heffeter P, Pirker C, Hager S, Kowol CR, van Schoonhoven S, Stojanovic M, Keppler BK, Berger W. Loss of phosphodiesterase 4D mediates acquired triapine resistance via Epac-Rap1-Integrin signaling. Oncotarget 2016; 7:84556-84574. [PMID: 27602951 PMCID: PMC5356681 DOI: 10.18632/oncotarget.11821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/24/2016] [Indexed: 12/27/2022] Open
Abstract
Triapine, an anticancer thiosemicarbazone, is currently under clinical investigation. Whereas promising results were obtained in hematological diseases, trials in solid tumors widely failed. To understand mechanisms causing triapine insensitivity, we have analysed genomic alterations in a triapine-resistant SW480 subline (SW480/tria). Only one distinct genomic loss was observed specifically in SW480/tria cells affecting the phosphodiesterase 4D (PDE4D) gene locus. Accordingly, pharmacological inhibition of PDE4D resulted in significant triapine resistance in SW480 cells. Hence, we concluded that enhanced cyclic AMP levels might confer protection against triapine. Indeed, hyperactivation of both major downstream pathways, namely the protein kinase A (PKA)-cAMP response element-binding protein (Creb) and the exchange protein activated by cAMP (Epac)-Ras-related protein 1 (Rap1) signaling axes, was observed in SW480/tria cells. Unexpectedly, inhibition of PKA did not re-sensitize SW480/tria cells against triapine. In contrast, Epac activation resulted in distinct triapine resistance in SW480 cells. Conversely, knock-down of Epac expression and pharmacological inhibition of Rap1 re-sensitized SW480/tria cells against triapine. Rap1 is a well-known regulator of integrins. Accordingly, SW480/tria cells displayed enhanced plasma membrane expression of several integrin subunits, enhanced adhesion especially to RGD-containing matrix components, and bolstered activation/expression of the integrin downstream effectors Src and RhoA/Rac. Accordingly, integrin and Src inhibition resulted in potent triapine re-sensitization especially of SW480/tria cells. In summary, we describe for the first time integrin activation based on cAMP-Epac-Rap1 signaling as acquired drug resistance mechanism. combinations of triapine with inhibitors of several steps in this resistance cascade might be feasible strategies to overcome triapine insensitivity of solid tumors.
Collapse
Affiliation(s)
- Walter Miklos
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Hager
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Institute of Inorganic Chemistry, University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| | - Sushilla van Schoonhoven
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Mirjana Stojanovic
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry, University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| |
Collapse
|
11
|
Redox cycling metals: Pedaling their roles in metabolism and their use in the development of novel therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:727-48. [PMID: 26844773 DOI: 10.1016/j.bbamcr.2016.01.026] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
Essential metals, such as iron and copper, play a critical role in a plethora of cellular processes including cell growth and proliferation. However, concomitantly, excess of these metal ions in the body can have deleterious effects due to their ability to generate cytotoxic reactive oxygen species (ROS). Thus, the human body has evolved a very well-orchestrated metabolic system that keeps tight control on the levels of these metal ions. Considering their very high proliferation rate, cancer cells require a high abundance of these metals compared to their normal counterparts. Interestingly, new anti-cancer agents that take advantage of the sensitivity of cancer cells to metal sequestration and their susceptibility to ROS have been developed. These ligands can avidly bind metal ions to form redox active metal complexes, which lead to generation of cytotoxic ROS. Furthermore, these agents also act as potent metastasis suppressors due to their ability to up-regulate the metastasis suppressor gene, N-myc downstream regulated gene 1. This review discusses the importance of iron and copper in the metabolism and progression of cancer, how they can be exploited to target tumors and the clinical translation of novel anti-cancer chemotherapeutics.
Collapse
|
12
|
Müller BG, De Aretxabala X, González Domingo M. A review of recent data in the treatment of gallbladder cancer: what we know, what we do, and what should be done. Am Soc Clin Oncol Educ Book 2015:e165-70. [PMID: 24857099 DOI: 10.14694/edbook_am.2014.34.e165] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gallbladder cancer is now considered a distinct clinical entity, allowing for a separate analysis from that of other malignancies of the biliary tree. Symptoms related to a malignant tumor of the gallbladder include jaundice and abdominal pain, or a palpable abdominal mass that occurs in a late stage of the disease. The majority of patients with operable gallbladder cancer are diagnosed by cholecystectomy performed for presumed benign disease, mostly cholelithiasis, a clinical entity known as incidental gallbladder cancer. Given the poor prognosis if tumor invasion beyond the muscular layer and/or nodal metastasis is found, adjuvant treatments have been implemented, but few data are available to guide treatment decisions in this setting. For advanced disease, a multidisciplinary treatment approach including biliary drainage procedures and palliative support is needed in the management of this aggressive disease. Palliative chemotherapy with a combination of gemcitabine and cisplatin or oxaliplatin is the standard treatment based on the findings of two phase III trials that showed improved overall survival compared to single-agent chemotherapy and best supportive care. Several phase II studies have been reported investigating the role of targeted agents against EGFR, VEGF, HER2, and MEK. International collaboration to enhance our knowledge of gallbladder cancer should be encouraged.
Collapse
Affiliation(s)
- Bettina G Müller
- From the Department of Medical Oncology, Instituto Nacional del Cáncer, Santiago, Chile; the Department of Surgery, Clínica Alemana de Santiago, Santiago, Chile; the Department of Radiotherapy, Instituto Oncológico, Viña del Mar, Chile
| | - Xabier De Aretxabala
- From the Department of Medical Oncology, Instituto Nacional del Cáncer, Santiago, Chile; the Department of Surgery, Clínica Alemana de Santiago, Santiago, Chile; the Department of Radiotherapy, Instituto Oncológico, Viña del Mar, Chile
| | - Manuel González Domingo
- From the Department of Medical Oncology, Instituto Nacional del Cáncer, Santiago, Chile; the Department of Surgery, Clínica Alemana de Santiago, Santiago, Chile; the Department of Radiotherapy, Instituto Oncológico, Viña del Mar, Chile
| |
Collapse
|
13
|
de Oliveira JF, da Silva AL, Vendramini-Costa DB, da Cruz Amorim CA, Campos JF, Ribeiro AG, Olímpio de Moura R, Neves JL, Ruiz ALTG, Ernesto de Carvalho J, Alves de Lima MDC. Synthesis of thiophene-thiosemicarbazone derivatives and evaluation of their in vitro and in vivo antitumor activities. Eur J Med Chem 2015; 104:148-56. [DOI: 10.1016/j.ejmech.2015.09.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/26/2015] [Accepted: 09/29/2015] [Indexed: 01/08/2023]
|
14
|
Potůčková E, Roh J, Macháček M, Sahni S, Stariat J, Šesták V, Jansová H, Hašková P, Jirkovská A, Vávrová K, Kovaříková P, Kalinowski DS, Richardson DR, Šimůnek T. In Vitro Characterization of the Pharmacological Properties of the Anti-Cancer Chelator, Bp4eT, and Its Phase I Metabolites. PLoS One 2015; 10:e0139929. [PMID: 26460540 PMCID: PMC4604124 DOI: 10.1371/journal.pone.0139929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/19/2015] [Indexed: 12/01/2022] Open
Abstract
Cancer cells have a high iron requirement and many experimental studies, as well as clinical trials, have demonstrated that iron chelators are potential anti-cancer agents. The ligand, 2-benzoylpyridine 4-ethyl-3-thiosemicarbazone (Bp4eT), demonstrates both potent anti-neoplastic and anti-retroviral properties. In this study, Bp4eT and its recently identified amidrazone and semicarbazone metabolites were examined and compared with respect to their anti-proliferative activity towards cancer cells (HL-60 human promyelocytic leukemia, MCF-7 human breast adenocarcinoma, HCT116 human colon carcinoma and A549 human lung adenocarcinoma), non-cancerous cells (H9c2 neonatal rat-derived cardiomyoblasts and 3T3 mouse embryo fibroblasts) and their interaction with intracellular iron pools. Bp4eT was demonstrated to be a highly potent and selective anti-neoplastic agent that induces S phase cell cycle arrest, mitochondrial depolarization and apoptosis in MCF-7 cells. Both semicarbazone and amidrazone metabolites showed at least a 300-fold decrease in cytotoxic activity than Bp4eT towards both cancer and normal cell lines. The metabolites also lost the ability to: (1) promote the redox cycling of iron; (2) bind and mobilize iron from labile intracellular pools; and (3) prevent 59Fe uptake from 59Fe-labeled transferrin by MCF-7 cells. Hence, this study demonstrates that the highly active ligand, Bp4eT, is metabolized to non-toxic and pharmacologically inactive analogs, which most likely contribute to its favorable pharmacological profile. These findings are important for the further development of this drug candidate and contribute to the understanding of the structure-activity relationships of these agents.
Collapse
Affiliation(s)
- Eliška Potůčková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Jaroslav Roh
- Department of Inorganic and Organic Chemistry, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Miloslav Macháček
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Sumit Sahni
- Molecular Pharmacology and Pathology Program, Bosch Institute and Department of Pathology, University of Sydney, Sydney, Australia
| | - Ján Stariat
- Department of Pharmaceutical Chemistry and Drug Analysis, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Vít Šesták
- Department of Pharmaceutical Chemistry and Drug Analysis, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Hana Jansová
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Pavlína Hašková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Anna Jirkovská
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Kateřina Vávrová
- Department of Inorganic and Organic Chemistry, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Petra Kovaříková
- Department of Pharmaceutical Chemistry and Drug Analysis, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
| | - Danuta S. Kalinowski
- Molecular Pharmacology and Pathology Program, Bosch Institute and Department of Pathology, University of Sydney, Sydney, Australia
| | - Des R. Richardson
- Molecular Pharmacology and Pathology Program, Bosch Institute and Department of Pathology, University of Sydney, Sydney, Australia
- * E-mail: (TS); (DRR)
| | - Tomáš Šimůnek
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Hradec Králové, Czech Republic
- * E-mail: (TS); (DRR)
| |
Collapse
|
15
|
Abstract
OPINION STATEMENT A paradigm shift towards molecular-based, personalized cancer therapeutics has occurred in recent years and a number of targeted drugs have emerged. Various targeted therapies like erlotinib, trastuzumab, and cetuximab have been approved in lung, breast, and colon cancers, respectively. Numerous clinical trials involving targeted drugs in biliary tract cancers are currently in progress, though none have been approved for this disease. Biliary tract cancers are divided into separate entities both anatomically and in terms of pathogenesis but are grouped together in most trials given their rarity. Combination chemotherapy involving cisplatin and gemcitabine is the current standard of care in the metastatic setting. In this review, we will discuss the various molecular pathways implicated in biliary tract cancers and potential therapeutic targets.
Collapse
Affiliation(s)
- Amartej Merla
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY 10461 USA
| | - Kenneth G. Liu
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY 10461 USA
| | - Lakshmi Rajdev
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY 10461 USA
| |
Collapse
|
16
|
Gallbladder Cancer in the 21st Century. JOURNAL OF ONCOLOGY 2015; 2015:967472. [PMID: 26421012 PMCID: PMC4569807 DOI: 10.1155/2015/967472] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 08/07/2015] [Accepted: 08/12/2015] [Indexed: 02/07/2023]
Abstract
Gallbladder cancer (GBC) is an uncommon disease in the majority of the world despite being the most common and aggressive malignancy of the biliary tree. Early diagnosis is essential for improved prognosis; however, indolent and nonspecific clinical presentations with a paucity of pathognomonic/predictive radiological features often preclude accurate identification of GBC at an early stage. As such, GBC remains a highly lethal disease, with only 10% of all patients presenting at a stage amenable to surgical resection. Among this select population, continued improvements in survival during the 21st century are attributable to aggressive radical surgery with improved surgical techniques. This paper reviews the current available literature of the 21st century on PubMed and Medline to provide a detailed summary of the epidemiology and risk factors, pathogenesis, clinical presentation, radiology, pathology, management, and prognosis of GBC.
Collapse
|
17
|
Miklos W, Pelivan K, Kowol CR, Pirker C, Dornetshuber-Fleiss R, Spitzwieser M, Englinger B, van Schoonhoven S, Cichna-Markl M, Koellensperger G, Keppler BK, Berger W, Heffeter P. Triapine-mediated ABCB1 induction via PKC induces widespread therapy unresponsiveness but is not underlying acquired triapine resistance. Cancer Lett 2015; 361:112-20. [PMID: 25749419 DOI: 10.1016/j.canlet.2015.02.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/26/2015] [Accepted: 02/26/2015] [Indexed: 10/23/2022]
Abstract
Although triapine is promising for treatment of advanced leukemia, it failed against solid tumors due to widely unknown reasons. To address this issue, a new triapine-resistant cell line (SW480/tria) was generated by drug selection and investigated in this study. Notably, SW480/tria cells displayed broad cross-resistance against several known ABCB1 substrates due to high ABCB1 levels (induced by promoter hypomethylation). However, ABCB1 inhibition did not re-sensitize SW480/tria cells to triapine and subsequent analysis revealed that triapine is only a weak ABCB1 substrate without significant interaction with the ABCB1 transport function. Interestingly, in chemo-naive, parental SW480 cells short-time (24 h) treatment with triapine stimulated ABCB1 expression. These effects were based on activation of protein kinase C (PKC), a known response to cellular stress. In accordance, SW480/tria cells were characterized by elevated levels of PKC. Together, this led to the conclusion that increased ABCB1 expression is not the major mechanism of triapine resistance in SW480/tria cells. In contrast, increased ABCB1 expression was found to be a consequence of triapine stress-induced PKC activation. These data are especially of importance when considering the choice of chemotherapeutics for combination with triapine.
Collapse
Affiliation(s)
- W Miklos
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - K Pelivan
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", Vienna, Austria
| | - C R Kowol
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", Vienna, Austria
| | - C Pirker
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - R Dornetshuber-Fleiss
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria; Department of Pharmacology and Toxicology, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria
| | - M Spitzwieser
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090 Vienna, Austria
| | - B Englinger
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - S van Schoonhoven
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - M Cichna-Markl
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090 Vienna, Austria
| | - G Koellensperger
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090 Vienna, Austria
| | - B K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", Vienna, Austria
| | - W Berger
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", Vienna, Austria
| | - P Heffeter
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", Vienna, Austria.
| |
Collapse
|
18
|
Serda M, Kalinowski DS, Rasko N, Potůčková E, Mrozek-Wilczkiewicz A, Musiol R, Małecki JG, Sajewicz M, Ratuszna A, Muchowicz A, Gołąb J, Šimůnek T, Richardson DR, Polanski J. Exploring the anti-cancer activity of novel thiosemicarbazones generated through the combination of retro-fragments: dissection of critical structure-activity relationships. PLoS One 2014; 9:e110291. [PMID: 25329549 PMCID: PMC4199632 DOI: 10.1371/journal.pone.0110291] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/10/2014] [Indexed: 01/01/2023] Open
Abstract
Thiosemicarbazones (TSCs) are an interesting class of ligands that show a diverse range of biological activity, including anti-fungal, anti-viral and anti-cancer effects. Our previous studies have demonstrated the potent in vivo anti-tumor activity of novel TSCs and their ability to overcome resistance to clinically used chemotherapeutics. In the current study, 35 novel TSCs of 6 different classes were designed using a combination of retro-fragments that appear in other TSCs. Additionally, di-substitution at the terminal N4 atom, which was previously identified to be critical for potent anti-cancer activity, was preserved through the incorporation of an N4-based piperazine or morpholine ring. The anti-proliferative activity of the novel TSCs were examined in a variety of cancer and normal cell-types. In particular, compounds 1d and 3c demonstrated the greatest promise as anti-cancer agents with potent and selective anti-proliferative activity. Structure-activity relationship studies revealed that the chelators that utilized “soft” donor atoms, such as nitrogen and sulfur, resulted in potent anti-cancer activity. Indeed, the N,N,S donor atom set was crucial for the formation of redox active iron complexes that were able to mediate the oxidation of ascorbate. This further highlights the important role of reactive oxygen species generation in mediating potent anti-cancer activity. Significantly, this study identified the potent and selective anti-cancer activity of 1d and 3c that warrants further examination.
Collapse
Affiliation(s)
- Maciej Serda
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
| | - Danuta S. Kalinowski
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Nathalie Rasko
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Eliška Potůčková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Anna Mrozek-Wilczkiewicz
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
- A. Chełkowski Institute of Physics and Silesian Interdisciplinary Centre for Education and Research, University of Silesia, Katowice, Silesia, Poland
| | - Robert Musiol
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
| | - Jan G. Małecki
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
| | | | - Alicja Ratuszna
- A. Chełkowski Institute of Physics and Silesian Interdisciplinary Centre for Education and Research, University of Silesia, Katowice, Silesia, Poland
| | - Angelika Muchowicz
- Department of Immunology, Medical University of Warsaw, Warsaw, Mazovia, Poland
| | - Jakub Gołąb
- Department of Immunology, Medical University of Warsaw, Warsaw, Mazovia, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Mazovia, Poland
| | - Tomáš Šimůnek
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Des R. Richardson
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
- * E-mail: (JP); (DRR)
| | - Jaroslaw Polanski
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
- * E-mail: (JP); (DRR)
| |
Collapse
|
19
|
Mrozek-Wilczkiewicz A, Serda M, Musiol R, Malecki G, Szurko A, Muchowicz A, Golab J, Ratuszna A, Polanski J. Iron chelators in photodynamic therapy revisited: synergistic effect by novel highly active thiosemicarbazones. ACS Med Chem Lett 2014; 5:336-9. [PMID: 24900837 DOI: 10.1021/ml400422a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/23/2014] [Indexed: 11/28/2022] Open
Abstract
In photodynamic therapy (PDT), a noninvasive anticancer treatment, visible light, is used as a magic bullet selectively destroying cancer cells by a photosensitizer that is nontoxic in the dark. Protoporphyrin IX (PpIX) is a natural photosensitizer synthesized in the cell, which is also a chelating agent that if bonded to Fe(2+) forms heme, a central component of hemoglobin. Therefore, xenobiotic iron chelators can disturb iron homeostasis, increasing the accumulation of PpIX, obstructing the last step of heme biosynthesis, and enhancing PDT efficiency. However, the attempts to use this promising idea have not proved to be hugely successful. Herein, we revisited this issue by analyzing the application of iron chelators highly toxic in the dark, which should have higher Fe(2+) affinity than the nontoxic chelators used so far. We have designed and prepared thiosemicarbazones (TSC) with the highest dark cellular cytotoxicity among TSCs ever reported. We demonstrate that compound 2 exerts powerful PDT enhancement when used in combination with 5-aminolevulinic acid (ALA), a precursor of PpIX.
Collapse
Affiliation(s)
- Anna Mrozek-Wilczkiewicz
- Institute
of Chemistry, University of Silesia, Szkolna 9, PL-40-006 Katowice, Poland
- Institute
of Physics, University of Silesia, Uniwersytecka 4, PL-40-007 Katowice, Poland
| | - Maciej Serda
- Institute
of Chemistry, University of Silesia, Szkolna 9, PL-40-006 Katowice, Poland
| | - Robert Musiol
- Institute
of Chemistry, University of Silesia, Szkolna 9, PL-40-006 Katowice, Poland
| | - Grzegorz Malecki
- Institute
of Chemistry, University of Silesia, Szkolna 9, PL-40-006 Katowice, Poland
| | - Agnieszka Szurko
- Institute
of Physics, University of Silesia, Uniwersytecka 4, PL-40-007 Katowice, Poland
| | - Angelika Muchowicz
- Center
of Biostructure Research, Medical University of Warsaw, Banacha 1a, PL-02-097 Warsaw, Poland
| | - Jakub Golab
- Center
of Biostructure Research, Medical University of Warsaw, Banacha 1a, PL-02-097 Warsaw, Poland
| | - Alicja Ratuszna
- Institute
of Physics, University of Silesia, Uniwersytecka 4, PL-40-007 Katowice, Poland
| | - Jaroslaw Polanski
- Institute
of Chemistry, University of Silesia, Szkolna 9, PL-40-006 Katowice, Poland
| |
Collapse
|
20
|
Trondl R, Flocke LS, Kowol CR, Heffeter P, Jungwirth U, Mair GE, Steinborn R, Enyedy ÉA, Jakupec MA, Berger W, Keppler BK. Triapine and a more potent dimethyl derivative induce endoplasmic reticulum stress in cancer cells. Mol Pharmacol 2014; 85:451-9. [PMID: 24378333 DOI: 10.1124/mol.113.090605] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Triapine (3-AP; 3-aminopyridine-2-carboxaldehyde thiosemicarbazone), a ribonucleotide reductase inhibitor, has been extensively evaluated in clinical trials in the last decade. This study addresses the role of endoplasmic reticulum (ER) stress in the anticancer activity of 3-AP and the derivative N(4),N(4)-dimethyl-triapine (3-AP-Me), differing from 3-AP only by dimethylation of the terminal nitrogen. Treatment of colon cancer cells with 3-AP or 3-AP-Me activated all three ER stress pathways (PERK, IRE1a, ATF6) by phosphorylation of eIF2α and upregulation of gene expression of activating transcription factors ATF4 and ATF6. In particular, 3-AP-Me led to an upregulation of the alternatively spliced mRNA variant XBP1 (16-fold). Moreover, 3-AP and 3-AP-Me activated the cellular stress kinases c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases, and inhibition of JNK activity antagonized the cytotoxic effect of both compounds. Subsequent to induction of the unfolded protein response, a significant upregulation of proapoptotic proteins was detected, including the transcription factor CHOP and Bim, an essential factor for ER stress-related apoptosis. In correlation with the higher degree of ER stress after 3-AP-Me treatment, also a more potent depolarization of mitochondrial membranes was found. These data suggest that 3-AP and 3-AP-Me induce apoptosis via ER stress. This was further corroborated by showing that inhibition of protein biosynthesis with cycloheximide prior to 3-AP and 3-AP-Me treatment leads to a significant reduction of the antiproliferative properties of both compounds. Taken together, this study demonstrates that induction of ER stress contributes to the mode of action of 3-AP and that terminal dimethylation leads to an even more pronounced manifestation of this effect.
Collapse
Affiliation(s)
- Robert Trondl
- Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria (R.T., L.S.F., C.R.K., M.A.J., B.K.K.); Research Platform "Translational Cancer Therapy Research" (R.T., C.R.K., P.H., U.J., M.A.J., W.B., B.K.K.), Institute of Cancer Research (P.H., U.J., W.B.), and Comprehensive Cancer Centre (P.H., U.J., W.B.), Medical University of Vienna, Vienna, Austria; Genomics Core Facility, VetCore, University of Veterinary Medicine, Vienna, Austria (G.E.M., R.S.); Department of Inorganic and Analytical Chemistry, University of Szeged, Szeged, Hungary (E.A.E.); and Hungarian Academy of Science-USZ Bioinorganic Chemistry Research Group, Szeged, Hungary (E.A.E.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
A phase I study of prolonged infusion of triapine in combination with fixed dose rate gemcitabine in patients with advanced solid tumors. Invest New Drugs 2012; 31:685-95. [PMID: 22847785 DOI: 10.1007/s10637-012-9863-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/18/2012] [Indexed: 12/21/2022]
Abstract
PURPOSE Prolonged exposure of cancer cells to triapine, an inhibitor of ribonucleotide reductase, followed by gemcitabine enhances gemcitabine activity in vitro. Fixed-dose-rate gemcitabine (FDR-G) has improved efficacy compared to standard-dose. We conducted a phase I trial to determine the maximum tolerated dose (MTD), safety, pharmacokinetics (PK), pharmacodynamics (PD), and preliminary efficacy of prolonged triapine infusion followed by FDR-G. EXPERIMENTAL DESIGN Triapine was given as a 24-hour infusion, immediately followed by FDR-G (1000 mg/m(2) over 100-minute). Initially, this combination was administered days 1 and 8 of a 21-day cycle (Arm A, triapine starting dose 120 mg); but because of myelosuppression, it was changed to days 1 and 15 of a 28-day cycle (Arm B, starting dose of triapine 75 mg). Triapine steady-state concentrations (Css) and circulating ribonucleotide reductase M2-subunit (RRM2) were measured. RESULTS Thirty-six patients were enrolled. The MTD was determined to be triapine 90 mg (24-hour infusion) immediately followed by gemcitabine 1000 mg/m(2) (100-minute infusion), every 2 weeks of a 4-week cycle. DLTs included grade 4 thrombocytopenia, leukopenia and neutropenia. The treatment was well tolerated with fatigue, nausea/vomiting, fever, transaminitis, and cytopenias being the most common toxicities. Among 30 evaluable patients, 1 had a partial response and 15 had stable disease. Triapine PK was similar, although more variable, compared to previous studies using doses normalized to body-surface-area. Steady decline in circulating levels of RRM2 may correlate with outcome. CONCLUSIONS This combination was well tolerated and showed evidence of preliminary activity in this heavily pretreated patient population, including prior gemcitabine failure.
Collapse
|