1
|
Wu X, Zhou Z, Li K, Liu S. Nanomaterials-Induced Redox Imbalance: Challenged and Opportunities for Nanomaterials in Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308632. [PMID: 38380505 PMCID: PMC11040387 DOI: 10.1002/advs.202308632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/24/2024] [Indexed: 02/22/2024]
Abstract
Cancer cells typically display redox imbalance compared with normal cells due to increased metabolic rate, accumulated mitochondrial dysfunction, elevated cell signaling, and accelerated peroxisomal activities. This redox imbalance may regulate gene expression, alter protein stability, and modulate existing cellular programs, resulting in inefficient treatment modalities. Therapeutic strategies targeting intra- or extracellular redox states of cancer cells at varying state of progression may trigger programmed cell death if exceeded a certain threshold, enabling therapeutic selectivity and overcoming cancer resistance to radiotherapy and chemotherapy. Nanotechnology provides new opportunities for modulating redox state in cancer cells due to their excellent designability and high reactivity. Various nanomaterials are widely researched to enhance highly reactive substances (free radicals) production, disrupt the endogenous antioxidant defense systems, or both. Here, the physiological features of redox imbalance in cancer cells are described and the challenges in modulating redox state in cancer cells are illustrated. Then, nanomaterials that regulate redox imbalance are classified and elaborated upon based on their ability to target redox regulations. Finally, the future perspectives in this field are proposed. It is hoped this review provides guidance for the design of nanomaterials-based approaches involving modulating intra- or extracellular redox states for cancer therapy, especially for cancers resistant to radiotherapy or chemotherapy, etc.
Collapse
Affiliation(s)
- Xumeng Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150006China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
| | - Ziqi Zhou
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| | - Kai Li
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| | - Shaoqin Liu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150006China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| |
Collapse
|
2
|
Zhang C, Kuo JCT, Huang Y, Hu Y, Deng L, Yung BC, Zhao X, Zhang Z, Pan J, Ma Y, Lee RJ. Optimized Liposomal Delivery of Bortezomib for Advancing Treatment of Multiple Myeloma. Pharmaceutics 2023; 15:2674. [PMID: 38140015 PMCID: PMC10747406 DOI: 10.3390/pharmaceutics15122674] [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: 10/09/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Bortezomib (BTZ), a boronic acid-derived proteasome inhibitor, is commonly employed in treating multiple myeloma (MM). However, the applications of BTZ are limited due to its poor stability and low bioavailability. Herein, we develop an optimized liposomal formulation of BTZ (L-BTZ) by employing a remote-loading strategy. This formulation uses Tiron, a divalent anionic catechol derivative, as the internal complexing agent. Compared to earlier BTZ-related formulations, this alternative formulation showed significantly greater stability due to the Tiron-BTZ complex's higher pH stability and negative charges, compared to the meglumine-BTZ complex. Significantly, the plasma AUC of L-BTZ was found to be 30 times greater than that of free BTZ, suggesting an extended blood circulation duration. In subsequent therapeutic evaluations using two murine xenograft tumor models of MM, the NCI-H929 and OPM2 models showed tumor growth inhibition (TGI) values of 37% and 57%, respectively. In contrast, free BTZ demonstrated TGI values of 17% and 11% in these models. Further, L-BTZ presented enhanced antitumor efficacy in the Hepa1-6 HCC syngeneic model, indicating its potential broader applicability as an antineoplastic agent. These findings suggest that the optimized L-BTZ formulation offers a significant advancement in BTZ delivery, holding substantial promise for clinical investigation in not merely MM, but other cancer types.
Collapse
Affiliation(s)
- Chi Zhang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Y.H.); (Z.Z.)
| | - Jimmy Chun-Tien Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Y.H.); (Z.Z.)
| | - Yirui Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Y.H.); (Z.Z.)
| | - Yingwen Hu
- The Whiteoak Group, Inc., Rockville, MD 20855, USA; (Y.H.); (L.D.); (B.C.Y.); (X.Z.)
| | - Lan Deng
- The Whiteoak Group, Inc., Rockville, MD 20855, USA; (Y.H.); (L.D.); (B.C.Y.); (X.Z.)
| | - Bryant C. Yung
- The Whiteoak Group, Inc., Rockville, MD 20855, USA; (Y.H.); (L.D.); (B.C.Y.); (X.Z.)
| | - Xiaobin Zhao
- The Whiteoak Group, Inc., Rockville, MD 20855, USA; (Y.H.); (L.D.); (B.C.Y.); (X.Z.)
| | - Zhongkun Zhang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Y.H.); (Z.Z.)
| | - Junjie Pan
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA;
| | - Yifan Ma
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA;
| | - Robert J. Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Y.H.); (Z.Z.)
| |
Collapse
|
3
|
Ng NS, Newbery M, Touffu A, Maksour S, Chung J, Carroll L, Zaw T, Wu Y, Ooi L. Edaravone and mitochondrial transfer as potential therapeutics for vanishing white matter disease astrocyte dysfunction. CNS Neurosci Ther 2023. [PMID: 36971196 PMCID: PMC10401142 DOI: 10.1111/cns.14190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 02/23/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
INTRODUCTION Previous research has suggested that vanishing white matter disease (VWMD) astrocytes fail to fully differentiate and respond differently to cellular stresses compared to healthy astrocytes. However, few studies have investigated potential VWMD therapeutics in monoculture patient-derived cell-based models. METHODS To investigate the impact of alterations in astrocyte expression and function in VWMD, astrocytes were differentiated from patient and control induced pluripotent stem cells and analyzed by proteomics, pathway analysis, and functional assays, in the absence and presence of stressors or potential therapeutics. RESULTS Vanishing white matter disease astrocytes demonstrated significantly reduced expression of astrocyte markers and markers of inflammatory activation or cellular stress relative to control astrocytes. These alterations were identified both in the presence and absence of polyinosinic:polycytidylic acid stimuli, which is used to simulate viral infections. Pathway analysis highlighted differential signaling in multiple pathways in VWMD astrocytes, including eukaryotic initiation factor 2 (EIF2) signaling, oxidative stress, oxidative phosphorylation (OXPHOS), mitochondrial function, the unfolded protein response (UPR), phagosome regulation, autophagy, ER stress, tricarboxylic acid cycle (TCA) cycle, glycolysis, tRNA signaling, and senescence pathways. Since oxidative stress and mitochondrial function were two of the key pathways affected, we investigated whether two independent therapeutic strategies could ameliorate astrocyte dysfunction: edaravone treatment and mitochondrial transfer. Edaravone treatment reduced differential VWMD protein expression of the UPR, phagosome regulation, ubiquitination, autophagy, ER stress, senescence, and TCA cycle pathways. Meanwhile, mitochondrial transfer decreased VWMD differential expression of the UPR, glycolysis, calcium transport, phagosome formation, and ER stress pathways, while further modulating EIF2 signaling, tRNA signaling, TCA cycle, and OXPHOS pathways. Mitochondrial transfer also increased the gene and protein expression of the astrocyte marker, glial fibrillary acidic protein (GFAP) in VWMD astrocytes. CONCLUSION This study provides further insight into the etiology of VWMD astrocytic failure and suggests edaravone and mitochondrial transfer as potential candidate VWMD therapeutics that can ameliorate disease pathways in astrocytes related to oxidative stress, mitochondrial dysfunction, and proteostasis.
Collapse
|
4
|
Allegra A, Petrarca C, Di Gioacchino M, Casciaro M, Musolino C, Gangemi S. Modulation of Cellular Redox Parameters for Improving Therapeutic Responses in Multiple Myeloma. Antioxidants (Basel) 2022; 11:antiox11030455. [PMID: 35326105 PMCID: PMC8944660 DOI: 10.3390/antiox11030455] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 01/25/2023] Open
Abstract
Raised oxidative stress and abnormal redox status are typical features of multiple myeloma cells, and the identification of the intimate mechanisms that regulate the relationships between neoplastic cells and redox homeostasis may reveal possible new anti-myeloma therapeutic targets to increase the effectiveness of anti-myeloma drugs synergistically or to eradicate drug-resistant clones while reducing toxicity toward normal cells. An alteration of the oxidative state is not only responsible for the onset of multiple myeloma and its progression, but it also appears essential for the therapeutic response and for developing any chemoresistance. Our review aimed to evaluate the literature’s current data on the effects of oxidative stress on the response to drugs generally employed in the therapy of multiple myeloma, such as proteasome inhibitors, immunomodulators, and autologous transplantation. In the second part of the review, we analyzed the possibility of using other substances, often of natural origin, to modulate the oxidative stress to interfere with the progression of myelomatous disease.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
- Correspondence: (A.A.); (M.D.G.)
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- Correspondence: (A.A.); (M.D.G.)
| | - Marco Casciaro
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| |
Collapse
|
5
|
Retzlaff D, Dörfler J, Kutschan S, Freuding M, Büntzel J, Hübner J. The Vitamin E Isoform α-Tocopherol is Not Effective as a Complementary Treatment in Cancer Treatment: A Systematic Review. Nutr Cancer 2021; 74:2313-2336. [PMID: 34918607 DOI: 10.1080/01635581.2021.2014905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The term vitamin E describes tocopherols and tocotrienols, whose chemical variations result in different biological activities including antioxidants. Neuroprotective effects of alpha-tocopherol against different toxins are assumed, therefore, it is discussed as a possible protective factor for adverse effects in cancer treatment. In July 2020, a systematic search was conducted searching five databases (Embase, Cochrane, PsychInfo, Cinahl, Medline) to find studies concerning the impact of α-tocopherol application and its potential harm on cancer patients. From 7546 search results, 22 publications referring to 20 studies with 1941 patients were included. Included patients were diagnosed with various cancer types and stages. Outcome variables were overall survival of cancer, symptom management of mucositis and chemotherapy-induced peripheral neuropathy (CIPN). The studies had different methodological qualities (mainly acceptable) and reported heterogeneous results: some reported significant improvement of mucositis and CIPN while others did not find changes concerning these endpoints. Due to heterogeneous results and methodical limitations of the included studies, a clear statement regarding the effectiveness of α-tocopherol as complementary treatment for cancer patients is not possible. Despite findings regarding reduction of oral side effects, usage of α-tocopherol during therapy must be discouraged because of potential negative influence on survival rates.
Collapse
Affiliation(s)
- Dorothee Retzlaff
- Klinik für Innere Medizin II, Hematology and Internal Oncology, Universitätsklinikum Jena, Jena, Germany
| | - Jennifer Dörfler
- Klinik für Innere Medizin II, Hematology and Internal Oncology, Universitätsklinikum Jena, Jena, Germany
| | - Sabine Kutschan
- Klinik für Innere Medizin II, Hematology and Internal Oncology, Universitätsklinikum Jena, Jena, Germany
| | - Maren Freuding
- Klinik für Innere Medizin II, Hematology and Internal Oncology, Universitätsklinikum Jena, Jena, Germany
| | - Jens Büntzel
- Department of Otorhinolaryngology Südharzklinikum Nordhausen, Germany
| | - Jutta Hübner
- Klinik für Innere Medizin II, Hematology and Internal Oncology, Universitätsklinikum Jena, Jena, Germany
| |
Collapse
|
6
|
Sacco A, Federico C, Todoerti K, Ziccheddu B, Palermo V, Giacomini A, Ravelli C, Maccarinelli F, Bianchi G, Belotti A, Ribolla R, Favasuli V, Revenko AS, Macleod AR, Willis B, Cai H, Hauser J, Rooney C, Willis SE, Martin PL, Staniszewska A, Ambrose H, Hanson L, Cattaneo C, Tucci A, Rossi G, Ronca R, Neri A, Mitola S, Bolli N, Presta M, Moschetta M, Ross S, Roccaro AM. Specific targeting of the KRAS mutational landscape in myeloma as a tool to unveil the elicited antitumor activity. Blood 2021; 138:1705-1720. [PMID: 34077955 PMCID: PMC9710471 DOI: 10.1182/blood.2020010572] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/07/2021] [Indexed: 12/14/2022] Open
Abstract
Alterations in KRAS have been identified as the most recurring somatic variants in the multiple myeloma (MM) mutational landscape. Combining DNA and RNA sequencing, we studied 756 patients and observed KRAS as the most frequently mutated gene in patients at diagnosis; in addition, we demonstrated the persistence or de novo occurrence of the KRAS aberration at disease relapse. Small-molecule inhibitors targeting KRAS have been developed; however, they are selective for tumors carrying the KRASG12C mutation. Therefore, there is still a need to develop novel therapeutic approaches to target the KRAS mutational events found in other tumor types, including MM. We used AZD4785, a potent and selective antisense oligonucleotide that selectively targets and downregulates all KRAS isoforms, as a tool to dissect the functional sequelae secondary to KRAS silencing in MM within the context of the bone marrow niche and demonstrated its ability to significantly silence KRAS, leading to inhibition of MM tumor growth, both in vitro and in vivo, and confirming KRAS as a driver and therapeutic target in MM.
Collapse
Affiliation(s)
- Antonio Sacco
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Cinzia Federico
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Katia Todoerti
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Bachisio Ziccheddu
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Valentina Palermo
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giada Bianchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Angelo Belotti
- Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | | | - Vanessa Favasuli
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | | | | | | | - Joana Hauser
- Oncology R &D, AstraZeneca, Cambridge, United Kingdom; and
| | - Claire Rooney
- Oncology R &D, AstraZeneca, Cambridge, United Kingdom; and
| | | | | | | | - Helen Ambrose
- Oncology R &D, AstraZeneca, Cambridge, United Kingdom; and
| | - Lyndsey Hanson
- Oncology R &D, AstraZeneca, Cambridge, United Kingdom; and
| | | | | | - Giuseppe Rossi
- Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonino Neri
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Niccolò Bolli
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Sarah Ross
- Oncology R &D, AstraZeneca, Cambridge, United Kingdom; and
| | - Aldo M. Roccaro
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| |
Collapse
|
7
|
Hoppe C, Freuding M, Büntzel J, Münstedt K, Hübner J. Clinical efficacy and safety of oral and intravenous vitamin C use in patients with malignant diseases. J Cancer Res Clin Oncol 2021; 147:3025-3042. [PMID: 34402972 PMCID: PMC8397678 DOI: 10.1007/s00432-021-03759-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/07/2021] [Indexed: 11/27/2022]
Abstract
Background Vitamin C, also called ascorbic acid, is a water-soluble antioxidant and free radical scavenger. It is required in the body for numerous metabolic functions and is involved in the development of proteins and connective tissue. Methods In April 2020, a systematic search was carried out on five electronic databases (Medline, Embase, Cochrane, Cinahl, PsycINFO) to find studies on the use, efficacy and safety of a complementary therapy with vitamin C in oncological patients. Results Out of the initial 23,195 search results, 21 studies with 1961 patients were included in this review. Five of the included studies (n = 417) were randomized controlled trials (RCTs). The remaining 16 studies belonged to a lower class of evidence. The patients who were treated with vitamin C suffered from various malignant diseases, some in an advanced and palliative stage. Vitamin C was applied intravenously or orally. It was either the only treatment or was combined with chemo- or radiotherapy. Endpoints included the development of the disease-related symptoms, quality of life, mortality, progression-free survival and safety of vitamin C. The studies were of moderate quality and showed either no effect of vitamin C or a positive trend, although this has rarely been statistically proven in group comparisons. No or only slight side effects with both oral and intravenous administration of vitamin C were reported. Conclusion Oral intake of vitamin C does not appear to have any effect in patients with malignancies. Data are heterogeneous for intravenous administration. There are no RCTs with statistical group comparisons.
Collapse
Affiliation(s)
- Catalina Hoppe
- Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany.
| | - Maren Freuding
- Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Jens Büntzel
- Klinik für HNO-Erkrankungen, Kopf-Hals-Chirurgie, Interdisziplinäre Palliativstation, Südharz Klinikum Nordhausen, Dr.-Robert-Koch-Straße 39, 99734, Nordhausen, Germany
| | - Karsten Münstedt
- Gynäkologie und Geburtshilfe, Ortenau Klinikum Offenburg-Kehl, Ebertplatz 12, 77654, Offenburg, Germany
| | - Jutta Hübner
- Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| |
Collapse
|
8
|
Poluboyarinov PA, Elistratov DG, Moiseeva IJ. Antitumor Activity of Selenium and Search Parameters for Its New Potentially Active Derivatives. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1068162020060254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
9
|
Hackman GL, Collins M, Lu X, Lodi A, DiGiovanni J, Tiziani S. Predicting and Quantifying Antagonistic Effects of Natural Compounds Given with Chemotherapeutic Agents: Applications for High-Throughput Screening. Cancers (Basel) 2020; 12:cancers12123714. [PMID: 33322034 PMCID: PMC7763027 DOI: 10.3390/cancers12123714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 01/12/2023] Open
Abstract
Natural products have been used for centuries to treat various human ailments. In recent decades, multi-drug combinations that utilize natural products to synergistically enhance the therapeutic effects of cancer drugs have been identified and have shown success in improving treatment outcomes. While drug synergy research is a burgeoning field, there are disagreements on the definitions and mathematical parameters that prevent the standardization and proper usage of the terms synergy, antagonism, and additivity. This contributes to the relatively small amount of data on the antagonistic effects of natural products on cancer drugs that can diminish their therapeutic efficacy and prevent cancer regression. The ability of natural products to potentially degrade or reverse the molecular activity of cancer therapeutics represents an important but highly under-emphasized area of research that is often overlooked in both pre-clinical and clinical studies. This review aims to evaluate the body of work surrounding the antagonistic interactions between natural products and cancer therapeutics and highlight applications for high-throughput screening (HTS) and deep learning techniques for the identification of natural products that antagonize cancer drug efficacy.
Collapse
Affiliation(s)
- G. Lavender Hackman
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (G.L.H.); (M.C.); (X.L.); (A.L.)
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA;
| | - Meghan Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (G.L.H.); (M.C.); (X.L.); (A.L.)
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA;
| | - Xiyuan Lu
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (G.L.H.); (M.C.); (X.L.); (A.L.)
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA;
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (G.L.H.); (M.C.); (X.L.); (A.L.)
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA;
| | - John DiGiovanni
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA;
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (G.L.H.); (M.C.); (X.L.); (A.L.)
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA;
- Department of Oncology, Dell Medical School, LiveSTRONG Cancer Institutes, The University of Texas at Austin, Austin, TX 78723, USA
- Correspondence: ; Tel.: +1-512-495-4706
| |
Collapse
|
10
|
Boretti A, Banik BK. Intravenous vitamin C for reduction of cytokines storm in acute respiratory distress syndrome. PHARMANUTRITION 2020; 12:100190. [PMID: 32322486 PMCID: PMC7172861 DOI: 10.1016/j.phanu.2020.100190] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022]
Abstract
The recent outbreak of Covid19 has required urgent treatments for numerous patients. No suitable vaccines or antivirals are available for Covid19. The efficiency against Covid19 of WHO therapies of choice, that are two antivirals developed for other pathologies, is controversial. Therefore, alternative approaches are required. Intravenous (IV) Vitamin C (Vit-C) has emerged as one of the other alternatives for this purpose. Here we review the effects of IV Vit-C on the immune system response, the antiviral properties of IV Vit-C, and finally the antioxidant properties of IV Vit-C to specifically address the cytokines' storm characteristic of the Acute Respiratory Distress Syndrome (ARDS) that occur in the later cycle of the Covid19 infectious disease.
Collapse
Affiliation(s)
- Alberto Boretti
- Prince Mohammad Bin Fahd University, P.O. Box 1664, Al Khobar, 31952, Saudi Arabia
| | - Bimal Krishna Banik
- Prince Mohammad Bin Fahd University, P.O. Box 1664, Al Khobar, 31952, Saudi Arabia
| |
Collapse
|
11
|
From Seabed to Bedside: A Review on Promising Marine Anticancer Compounds. Biomolecules 2020; 10:biom10020248. [PMID: 32041255 PMCID: PMC7072248 DOI: 10.3390/biom10020248] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023] Open
Abstract
The marine environment represents an outstanding source of antitumoral compounds and, at the same time, remains highly unexplored. Organisms living in the sea synthesize a wide variety of chemicals used as defense mechanisms. Interestingly, a large number of these compounds exert excellent antitumoral properties and have been developed as promising anticancer drugs that have later been approved or are currently under validation in clinical trials. However, due to the high need for these compounds, new methodologies ensuring its sustainable supply are required. Also, optimization of marine bioactives is an important step for their success in the clinical setting. Such optimization involves chemical modifications to improve their half-life in circulation, potency and tumor selectivity. In this review, we outline the most promising marine bioactives that have been investigated in cancer models and/or tested in patients as anticancer agents. Moreover, we describe the current state of development of anticancer marine compounds and discuss their therapeutic limitations as well as different strategies used to overcome these limitations. The search for new marine antitumoral agents together with novel identification and chemical engineering approaches open the door for novel, more specific and efficient therapeutic agents for cancer treatment.
Collapse
|
12
|
Metabolic Remodelling: An Accomplice for New Therapeutic Strategies to Fight Lung Cancer. Antioxidants (Basel) 2019; 8:antiox8120603. [PMID: 31795465 PMCID: PMC6943435 DOI: 10.3390/antiox8120603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Metabolic remodelling is a hallmark of cancer, however little has been unravelled in its role in chemoresistance, which is a major hurdle to cancer control. Lung cancer is a leading cause of death by cancer, mainly due to the diagnosis at an advanced stage and to the development of resistance to therapy. Targeted therapeutic agents combined with comprehensive drugs are commonly used to treat lung cancer. However, resistance mechanisms are difficult to avoid. In this review, we will address some of those therapeutic regimens, resistance mechanisms that are eventually developed by lung cancer cells, metabolic alterations that have already been described in lung cancer and putative new therapeutic strategies, and the integration of conventional drugs and genetic and metabolic-targeted therapies. The oxidative stress is pivotal in this whole network. A better understanding of cancer cell metabolism and molecular adaptations underlying resistance mechanisms will provide clues to design new therapeutic strategies, including the combination of chemotherapeutic and targeted agents, considering metabolic intervenients. As cancer cells undergo a constant metabolic adaptive drift, therapeutic regimens must constantly adapt.
Collapse
|
13
|
Marampon F, Codenotti S, Megiorni F, Del Fattore A, Camero S, Gravina GL, Festuccia C, Musio D, De Felice F, Nardone V, Santoro AN, Dominici C, Fanzani A, Pirtoli L, Fioravanti A, Tombolini V, Cheleschi S, Tini P. NRF2 orchestrates the redox regulation induced by radiation therapy, sustaining embryonal and alveolar rhabdomyosarcoma cells radioresistance. J Cancer Res Clin Oncol 2019; 145:881-893. [PMID: 30701326 DOI: 10.1007/s00432-019-02851-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/23/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Tumor cells generally exhibit higher levels of reactive oxygen species (ROS), however, when stressed, tumor cells can undergo a process of 'Redox Resetting' to acquire a new redox balance with stronger antioxidant systems that enable cancer cells to become resistant to radiation therapy (RT). Here, we describe how RT affects the oxidant/antioxidant balance in human embryonal (RD) and alveolar (RH30) rhabdomyosarcoma (RMS) cell lines, investigating on the molecular mechanisms involved. METHODS Radiations were delivered using an x-6 MV photon linear accelerator and their effects were assessed by vitality and clonogenic assays. The expression of specific antioxidant-enzymes, such as Superoxide Dismutases (SODs), Catalase (CAT) and Glutathione Peroxidases 4 (GPx4), miRNAs (miR-22, -126, -210, -375, -146a, -34a) and the transcription factor NRF2 was analyzed by quantitative polymerase chain reaction (q-PCR) and western blotting. RNA interference experiments were performed to evaluate the role of NRF2. RESULTS Doses of RT higher than 2 Gy significantly affected RMS clonogenic ability by increasing ROS production. RMS rapidly and efficiently brought back ROS levels by up-regulating the gene expression of antioxidant enzymes, miRNAs as well as of NRF2. Silencing of NRF2 restrained the RMS ability to counteract RT-induced ROS accumulation, antioxidant enzyme and miRNA expression and was able to increase the abundance of γ-H2AX, a biomarker of DNA damage, in RT-treated cells. CONCLUSIONS Taken together, our data suggest the strategic role of oxidant/antioxidant balance in restraining the therapeutic efficiency of RT in RMS treatment and identify NRF2 as a new potential molecular target whose inhibition might represent a novel radiosensitizing therapeutic strategy for RMS clinical management.
Collapse
Affiliation(s)
- Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy.
| | - Silvia Codenotti
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Andrea Del Fattore
- Multi-Factorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, IRCCS, Viale di San Paolo 15, 00146, Rome, Italy
| | - Simona Camero
- Department of Pediatrics, "Sapienza" University of Rome, Rome, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Daniela Musio
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Francesca De Felice
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | | | | | - Carlo Dominici
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Alessandro Fanzani
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luigi Pirtoli
- Azienda Ospedaliera Universitaria Senese, Siena, Italy.,Unit of Radiation Oncology, Azienda Ospedaliera Universitaria Senese, Siena, Italy.,Istituto Toscano Tumori, Florence, Italy.,Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA.,Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
| | | | - Vincenzo Tombolini
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Sara Cheleschi
- Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
| | - Paolo Tini
- Unit of Radiation Oncology, Azienda Ospedaliera Universitaria Senese, Siena, Italy.,Istituto Toscano Tumori, Florence, Italy.,Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA.,Sbarro Health Research Organization, Temple University, Philadelphia, PA, USA
| |
Collapse
|
14
|
Dom M, Offner F, Vanden Berghe W, Van Ostade X. Proteomic characterization of Withaferin A-targeted protein networks for the treatment of monoclonal myeloma gammopathies. J Proteomics 2018; 179:17-29. [PMID: 29448055 DOI: 10.1016/j.jprot.2018.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 01/09/2018] [Accepted: 02/05/2018] [Indexed: 02/09/2023]
Abstract
Withaferin A (WA), a natural steroid lactone from the plant Withania somnifera, is often studied because of its antitumor properties. Although many in vitro and in vivo studies have been performed, the identification of Withaferin A protein targets and its mechanism of antitumor action remain incomplete. We used quantitative chemoproteomics and differential protein expression analysis to characterize the WA antitumor effects on a multiple myeloma cell model. Identified relevant targets were further validated by Ingenuity Pathway Analysis and Western blot and indicate that WA targets protein networks that are specific for monoclonal gammopathy of undetermined significance (MGUS) and other closely related disorders, such as multiple myeloma (MM) and Waldenström macroglobulinemia (WM). By blocking the PSMB10 proteasome subunit, downregulation of ANXA4, potential association with HDAC6 and upregulation of HMOX1, WA puts a massive blockage on both proteotoxic and oxidative stress responses pathways, leaving cancer cells defenseless against WA induced stresses. These results indicate that WA mediated apoptosis is preceded by simultaneous targeting of cellular stress response pathways like proteasome degradation, autophagy and unfolded protein stress response and thus suggests that WA can be used as an effective treatment for MGUS and other closely related disorders. SIGNIFICANCE Multifunctional antitumor compounds are of great potential since they reduce the risk of multidrug resistance in chemotherapy. Unfortunately, characterization of all protein targets of a multifunctional compound is lacking. Therefore, we optimized an SILAC quantitative chemoproteomics workflow to identify the potential protein targets of Withaferin A (WA), a natural multifunctional compound with promising antitumor properties. To further understand the antitumor mechanisms of WA, we performed a differential protein expression analysis and combined the altered expression data with chemoproteome WA target data in the highly curated Ingenuity Pathway database. We provide a first global overview on how WA kills multiple myeloma cancer cells and serve as a starting point for further in depth experiments. Furthermore, the combined approach can be used for other types of cancer and/or other promising multifunctional compounds, thereby increasing the potential development of new antitumor therapies.
Collapse
Affiliation(s)
- Martin Dom
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Belgium
| | - Fritz Offner
- Hematology, Department Internal Medicine, Ghent University, Ghent, Belgium
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Belgium
| | - Xaveer Van Ostade
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Belgium.
| |
Collapse
|
15
|
Liu Y, Li Q, Zhou L, Xie N, Nice EC, Zhang H, Huang C, Lei Y. Cancer drug resistance: redox resetting renders a way. Oncotarget 2016; 7:42740-42761. [PMID: 27057637 PMCID: PMC5173169 DOI: 10.18632/oncotarget.8600] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/28/2016] [Indexed: 02/05/2023] Open
Abstract
Disruption of redox homeostasis is a crucial factor in the development of drug resistance, which is a major problem facing current cancer treatment. Compared with normal cells, tumor cells generally exhibit higher levels of reactive oxygen species (ROS), which can promote tumor progression and development. Upon drug treatment, some tumor cells can undergo a process of 'Redox Resetting' to acquire a new redox balance with higher levels of ROS accumulation and stronger antioxidant systems. Evidence has accumulated showing that the 'Redox Resetting' enables cancer cells to become resistant to anticancer drugs by multiple mechanisms, including increased rates of drug efflux, altered drug metabolism and drug targets, activated prosurvival pathways and inefficient induction of cell death. In this article, we provide insight into the role of 'Redox Resetting' on the emergence of drug resistance that may contribute to pharmacological modulation of resistance.
Collapse
Affiliation(s)
- Yuan Liu
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
- Department of Neurology, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan, P. R. China
| | - Qifu Li
- Department of Neurology, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan, P. R. China
| | - Li Zhou
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
| | - Na Xie
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Haiyuan Zhang
- Department of Neurology, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan, P. R. China
| | - Canhua Huang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
16
|
Vasuthasawat A, Yoo EM, Trinh KR, Lichtenstein A, Timmerman JM, Morrison SL. Targeted immunotherapy using anti-CD138-interferon α fusion proteins and bortezomib results in synergistic protection against multiple myeloma. MAbs 2016; 8:1386-1397. [PMID: 27362935 DOI: 10.1080/19420862.2016.1207030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although recent advances have substantially improved the management of multiple myeloma, it remains an incurable malignancy. We now demonstrate that anti-CD138 molecules genetically fused to type I interferons (IFN) synergize with the approved therapeutic bortezomib in arresting the proliferation of human multiple myeloma cell lines both in vitro and in vivo. The anti-CD138-IFNα14 fusion protein was active in inducing increased expression of signal transducer and activator of transcription 1 (STAT1) and its phosphorylation while the cell death pathway induced by bortezomib included generation of reactive oxygen species. Interferon regulatory factor 4 (IRF4), an important survival factor for myeloma cells, was down regulated following combination treatment. Induction of cell death appeared to be caspase-independent because treatment with inhibitors of caspase activation did not decrease the level of cell death. The observed caspase-independent synergistic cell death involved mitochondrial membrane depolarization, and poly(ADP-ribose) polymerase-1 (PARP-1) cleavage, and resulted in enhanced induction of apoptosis. Importantly, using 2 different in vivo xenograft models, we found that combination therapy of anti-CD138-IFNα14 and bortezomib was able to cure animals with established tumors (7 of 8 using OCI-My5 or 8 of 8 using NCI-H929). Thus, the combination of anti-CD138-IFNα with bortezomib shows great promise as a novel therapeutic approach for the treatment of multiple myeloma, a malignancy for which there are currently no cures.
Collapse
Affiliation(s)
- Alex Vasuthasawat
- a Department of Microbiology, Immunology and Molecular Genetics , University of California Los Angeles , Los Angeles , CA , USA.,b Molecular Biology Institute, UCLA , Los Angeles , CA , USA
| | - Esther M Yoo
- a Department of Microbiology, Immunology and Molecular Genetics , University of California Los Angeles , Los Angeles , CA , USA.,b Molecular Biology Institute, UCLA , Los Angeles , CA , USA
| | - Kham R Trinh
- a Department of Microbiology, Immunology and Molecular Genetics , University of California Los Angeles , Los Angeles , CA , USA.,b Molecular Biology Institute, UCLA , Los Angeles , CA , USA
| | - Alan Lichtenstein
- c Greater Los Angeles Veterans Administration Healthcare Center , Los Angeles , CA , USA.,d Jonsson Comprehensive Cancer Center , Los Angeles , CA , USA.,e Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, UCLA , Los Angeles , CA , USA
| | - John M Timmerman
- d Jonsson Comprehensive Cancer Center , Los Angeles , CA , USA.,e Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, UCLA , Los Angeles , CA , USA
| | - Sherie L Morrison
- a Department of Microbiology, Immunology and Molecular Genetics , University of California Los Angeles , Los Angeles , CA , USA.,b Molecular Biology Institute, UCLA , Los Angeles , CA , USA
| |
Collapse
|
17
|
Lipchick BC, Fink EE, Nikiforov MA. Oxidative stress and proteasome inhibitors in multiple myeloma. Pharmacol Res 2016; 105:210-5. [PMID: 26827824 PMCID: PMC5044866 DOI: 10.1016/j.phrs.2016.01.029] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 01/20/2016] [Accepted: 01/22/2016] [Indexed: 11/23/2022]
Abstract
Multiple myeloma is a form of plasma cell neoplasm that accounts for approximately 10% of all hematological malignancies. Recently, several novel drugs have been discovered that almost doubled the overall survival of multiple myeloma patients. One of these drugs, the first-in-class proteasome inhibitor bortezomib (Velcade) has demonstrated remarkable response rates in multiple myeloma patients, and yet, currently this disease remains incurable. The major factor undermining the success of multiple myeloma treatment is a rapidly emerging resistance to the available therapy. Thus, the development of stand-alone or adjuvant anti-myeloma agents becomes of paramount importance. Overproduction of intracellular reactive oxygen species (ROS) often accompanies malignant transformation due to oncogene activation and/or enhanced metabolism in tumor cells. As a result, these cells possess higher levels of ROS and lower levels of antioxidant molecules compared to their normal counterparts. Unbalanced production of ROS leads to oxidative stress which, if left unchecked, could be toxic for the cell. In multiple myeloma cells where high rates of immunoglobulin synthesis is an additional factor contributing to overproduction of ROS, further induction of oxidative stress can be an effective strategy to cope with this disease. Here we will review the available data on the role of oxidative stress in the cytotoxicity of proteasome inhibitors and the use of ROS-inducing compounds as anti-myeloma agents.
Collapse
Affiliation(s)
- Brittany C Lipchick
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | - Emily E Fink
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Mikhail A Nikiforov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
18
|
Branković B, Stanojević G, Nestorović M, Veljković A, Stojanović I, Petrović D, Pavlović D, Kocić G, Đinđić B, Krivokapić Z. TROSATIVE STRESS PARAMETERS IN COLON CANCER TUMOR, ADJACENT AND HEALTHY TISSUE. ACTA MEDICA MEDIANAE 2016. [DOI: 10.5633/amm.2016.0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
19
|
Božić A, Marinković A, Bjelogrlić S, Todorović TR, Cvijetić IN, Novaković I, Muller CD, Filipović NR. Quinoline based mono- and bis-(thio)carbohydrazones: synthesis, anticancer activity in 2D and 3D cancer and cancer stem cell models. RSC Adv 2016. [DOI: 10.1039/c6ra23940d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Study of antitumor activity of mono- and bis-quinoline based (thio)carbohydrazones on THP-1 and AsPC-1 cancer stem cells, revealed that thiocarbohydrazones had superior pro-apoptotic activity than carbohydrazones with multi-target profile activities.
Collapse
Affiliation(s)
- Aleksandra Božić
- Faculty of Technology and Metallurgy
- University of Belgrade
- Belgrade
- Serbia
| | | | | | | | - Ilija N. Cvijetić
- Innovation Center of the Faculty of Chemistry
- University of Belgrade
- Belgrade
- Serbia
| | - Irena Novaković
- Institute of Chemistry, Technology and Metallurgy
- University of Belgrade
- Belgrade
- Serbia
| | - Christian D. Muller
- Institut Pluridisciplinaire Hubert Curien
- UMR 7178
- CNRS
- Université de Strasbourg
- 67401 Illkirch
| | | |
Collapse
|
20
|
Mutant p53 (p53-R248Q) functions as an oncogene in promoting endometrial cancer by up-regulating REGγ. Cancer Lett 2015; 360:269-79. [DOI: 10.1016/j.canlet.2015.02.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 11/17/2022]
|
21
|
Brnjic S, Mazurkiewicz M, Fryknäs M, Sun C, Zhang X, Larsson R, D'Arcy P, Linder S. Induction of tumor cell apoptosis by a proteasome deubiquitinase inhibitor is associated with oxidative stress. Antioxid Redox Signal 2014; 21:2271-85. [PMID: 24011031 PMCID: PMC4241954 DOI: 10.1089/ars.2013.5322] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AIMS b-AP15 is a recently described inhibitor of the USP14/UCHL5 deubiquitinases (DUBs) of the 19S proteasome. Exposure to b-AP15 results in blocking of proteasome function and accumulation of polyubiquitinated protein substrates in cells. This novel mechanism of proteasome inhibition may potentially be exploited for cancer therapy, in particular for treatment of malignancies resistant to currently used proteasome inhibitors. The aim of the present study was to characterize the cellular response to b-AP15-mediated proteasome DUB inhibition. RESULTS We report that b-AP15 elicits a similar, but yet distinct, cellular response as the clinically used proteasome inhibitor bortezomib. b-AP15 induces a rapid apoptotic response, associated with enhanced induction of oxidative stress and rapid activation of Jun-N-terminal kinase 1/2 (JNK)/activating protein-1 signaling. Scavenging of reactive oxygen species and pharmacological inhibition of JNK reduced b-AP15-induced apoptosis. We further report that endoplasmic reticulum (ER) stress is induced by b-AP15 and is involved in apoptosis induction. In contrast to bortezomib, ER stress is associated with induction of α-subunit of eukaryotic initiation factor 2 phosphorylation. INNOVATION The findings establish that different modes of proteasome inhibition result in distinct cellular responses, a finding of potential therapeutic importance. CONCLUSION Our data show that enhanced oxidative stress and ER stress are major determinants of the strong apoptotic response elicited by the 19S DUB inhibitor b-AP15.
Collapse
Affiliation(s)
- Slavica Brnjic
- 1 Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institute , Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hübner J, Hanf V. Commonly used methods of complementary medicine in the treatment of breast cancer. ACTA ACUST UNITED AC 2014; 8:341-7. [PMID: 24415987 DOI: 10.1159/000355705] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Many patients with cancer look for information on complementary or alternative medicine (CAM) and use various CAM methods. Women with breast cancer are amongst the most avid users. Patients in Europe prefer drug-bound CAM methods, which are prone to side effects and drug interactions. In order to reduce these risks, communication between the patient and the physician on CAM is indispensible. Yet, most patients do not discuss CAM in general and complementary drug therapy in particular with their oncologists and most oncologists themselves are not overly familiar with the topic. This article gives an overview on the most often used CAM methods with regard to breast cancer. The current state of the scientific evidence, the benefits and risks are summarized.
Collapse
Affiliation(s)
- Jutta Hübner
- Dr. Senckenberg Chronomedical Institute, J.W. Goethe University, Frankfurt, Germany
| | - Volker Hanf
- Frauenklinik Nathanstift, Klinikum Fürth, Germany
| |
Collapse
|
23
|
Bergadà L, Yeramian A, Sorolla A, Matias-Guiu X, Dolcet X. Antioxidants impair anti-tumoral effects of Vorinostat, but not anti-neoplastic effects of Vorinostat and caspase-8 downregulation. PLoS One 2014; 9:e92764. [PMID: 24651472 PMCID: PMC3961419 DOI: 10.1371/journal.pone.0092764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 02/26/2014] [Indexed: 11/18/2022] Open
Abstract
We have recently demonstrated that histone deacetylase inhibitor, Vorinostat, applied as a single therapy or in combination with caspase-8 downregulation exhibits high anti-tumoral activity on endometrial carcinoma cell lines. In the present study, we have assessed the signalling processes underlying anti-tumoral effects of Vorinostat. Increasing evidence suggests that reactive oxygen species are responsible for histone deacetylase inhibitor-induced cell killing. We have found that Vorinostat induces formation of reactive oxygen species and DNA damage. To investigate the role of oxidative stress as anti-neoplastic mechanism, we have evaluated the effects of different antioxidants (Bha, Nac and Tiron) on endometrial carcinoma cell line Ishikawa treated with Vorinostat. We show that Bha, Nac and Tiron markedly inhibited the cytotoxic effects of Vorinostat, increasing cell viability in vitro. We found that all three antioxidants did not inhibited accumulation of acetyl Histone H4, so that antioxidants did not inhibit Vorinostat activity. Finally, we have evaluated the effects of antioxidants on anti-tumoral activity of Vorinostat as monotherapy or in combination with caspase-8 downregulation in vivo. Interestingly, antioxidants blocked the reduction of tumour growth caused by Vorinostat, but they were unable to inhibit anti-tumoral activity of Vorinostat plus caspase-8 inhibition.
Collapse
Affiliation(s)
- Laura Bergadà
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Andree Yeramian
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Annabel Sorolla
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
| | - Xavier Dolcet
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida, Lleida, Spain
- * E-mail:
| |
Collapse
|
24
|
Biskup E, Kamstrup MR, Manfé V, Gniadecki R. Proteasome inhibition as a novel mechanism of the proapoptotic activity of γ-secretase inhibitor I in cutaneous T-cell lymphoma. Br J Dermatol 2013; 168:504-12. [PMID: 23445313 DOI: 10.1111/bjd.12071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND We have previously discovered that Notch1 is expressed on malignant T cells in cutaneous T-cell lymphoma (CTCL), and is required for survival of CTCL cell lines. Notch can be inhibited by γ-secretase inhibitors (GSIs), which differ widely in their ability to induce apoptosis in CTCL. OBJECTIVES To investigate whether GSI-I, in addition to inhibiting Notch, induces apoptosis in CTCL by proteasome inhibition, as GSI-I is very potent and has structural similarity to the proteasome inhibitor MG-132. METHODS Cell lines derived from CTCL (MyLa, SeAx, JK, Mac1 and Mac2a) were treated with GSI-I and two other proteasome inhibitors (MG-132 and bortezomib). The effects on cell viability, apoptosis and proteasome activity were measured, as was the impact on the prosurvival, nuclear factor κB (NF-κB) pathway. RESULTS In CTCL, GSI-I had proteasome-blocking activity with a potency comparable to the proteasome inhibitors MG-132 and bortezomib. Proteasome inhibition was the main mechanism responsible for GSI-I-induced cell death, as tiron, a compound known to reverse the effect of MG-132, restored proteasome activity and largely abrogated the cytotoxic effect of GSI-I. Although inactivation of NF-κB is an important mechanism of action for proteasome inhibitors, we demonstrated an apparent activation of NF-κB. Furthermore, we showed that while the tumour suppressor protein p53 was induced during proteasome inhibition, it was dispensable for CTCL apoptosis, as both SeAx cells, which harbour p53 mutations that attenuate the apoptotic capacity, and HuT-78 cells, which have a deleted p53 gene, demonstrated potent apoptotic response. CONCLUSIONS GSI-I represents an interesting drug with a dual mechanism of action comprising inhibition of both Notch and the proteasome.
Collapse
Affiliation(s)
- E Biskup
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Bispebjerg Bakke 23, Copenhagen-2400, Denmark
| | | | | | | |
Collapse
|
25
|
Thangaraju S, Subramani E, Chakravarty B, Chaudhury K. Therapeutic targeting of the TNF superfamily: A promising treatment for advanced endometrial adenocarcinoma. Gynecol Oncol 2012; 127:426-32. [DOI: 10.1016/j.ygyno.2012.07.125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/26/2012] [Accepted: 07/29/2012] [Indexed: 12/18/2022]
|
26
|
Abstract
Targeting tumor cells is an important strategy to improve the selectivity of cancer therapies. With the advanced studies in cancer biology, we know that cancer cells are usually under increased oxidative stress. The high level of reactive oxygen species in cancer cells has been exploited for developing novel therapeutic strategies to preferentially kill cancer cells. Our group, amongst others, have used boronic acids/esters as triggers for developing ROS-activated anticancer prodrugs that target cancer cells. The selectivity was achieved by combining a specific reaction between boronates and H2O2, with the efficient masking of drug toxicity in the prodrug via boronates. Prodrugs activated via ferrocene-mediated oxidation have also been developed to improve the selectivity of anticancer drugs. We describe how the strategies of ROS-activation can be used for further development of new ROS-targeting prodrugs, eventually leading to novel approaches and/or combined technology for more efficient and selective treatment of cancers.
Collapse
|
27
|
Sardina JL, López-Ruano G, Sánchez-Sánchez B, Llanillo M, Hernández-Hernández A. Reactive oxygen species: are they important for haematopoiesis? Crit Rev Oncol Hematol 2011; 81:257-74. [PMID: 21507675 DOI: 10.1016/j.critrevonc.2011.03.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 03/15/2011] [Accepted: 03/22/2011] [Indexed: 02/07/2023] Open
Abstract
The production of reactive oxygen species (ROS) has traditionally been related to deleterious effects for cells. However, it is now widely accepted that ROS can play an important role in regulating cellular signalling and gene expression. NADPH oxidase ROS production seems to be especially important in this regard. Some lines of evidence suggest that ROS may be important modulators of cell differentiation, including haematopoietic differentiation, in both physiologic and pathologic conditions. Here we shall review how ROS can regulate cell signalling and gene expression. We shall also focus on the importance of ROS for haematopoietic stem cell (HSC) biology and for haematopoietic differentiation. We shall review the involvement of ROS and NADPH oxidases in cancer, and in particular what is known about the relationship between ROS and haematological malignancies. Finally, we shall discuss the use of ROS as cancer therapeutic targets.
Collapse
Affiliation(s)
- José L Sardina
- Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca, Spain
| | | | | | | | | |
Collapse
|
28
|
Acharya A, Das I, Chandhok D, Saha T. Redox regulation in cancer: a double-edged sword with therapeutic potential. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2010; 3:23-34. [PMID: 20716925 PMCID: PMC2835886 DOI: 10.4161/oxim.3.1.10095] [Citation(s) in RCA: 339] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Oxidative stress, implicated in the etiology of cancer, results from an imbalance in the production of reactive oxygen species (ROS) and cell’s own antioxidant defenses. ROS deregulate the redox homeostasis and promote tumor formation by initiating an aberrant induction of signaling networks that cause tumorigenesis. Ultraviolet (UV) exposures, γ-radiation and other environmental carcinogens generate ROS in the cells, which can exert apoptosis in the tumors, thereby killing the malignant cells or induce the progression of the cancer growth by blocking cellular defense system. Cancer stem cells take the advantage of the aberrant redox system and spontaneously proliferate. Oxidative stress and gene-environment interactions play a significant role in the development of breast, prostate, pancreatic and colon cancer. Prolonged lifetime exposure to estrogen is associated with several kinds of DNA damage. Oxidative stress and estrogen receptor-associated proliferative changes are suggested to play important roles in estrogen-induced breast carcinogenesis. BRCA1, a tumor suppressor against hormone responsive cancers such as breast and prostate cancer, plays a significant role in inhibiting ROS and estrogen mediated DNA damage; thereby regulate the redox homeostasis of the cells. Several transcription factors and tumor suppressors are involved during stress response such as Nrf2, NFκB and BRCA1. A promising strategy for targeting redox status of the cells is to use readily available natural substances from vegetables, fruits, herbs and spices. Many of the phytochemicals have already been identified to have chemopreventive potential, capable of intervening in carcinogenesis.
Collapse
Affiliation(s)
- Asha Acharya
- Lombardi Comprehensive Cancer Center, Pre Clinical Science, Washington DC, USA.
| | | | | | | |
Collapse
|
29
|
Abstract
Elevated rates of reactive oxygen species (ROS) have been detected in almost all cancers, where they promote many aspects of tumour development and progression. However, tumour cells also express increased levels of antioxidant proteins to detoxify from ROS, suggesting that a delicate balance of intracellular ROS levels is required for cancer cell function. Further, the radical generated, the location of its generation, as well as the local concentration is important for the cellular functions of ROS in cancer. A challenge for novel therapeutic strategies will be the fine tuning of intracellular ROS signalling to effectively deprive cells from ROS-induced tumour promoting events, towards tipping the balance to ROS-induced apoptotic signalling. Alternatively, therapeutic antioxidants may prevent early events in tumour development, where ROS are important. However, to effectively target cancer cells specific ROS-sensing signalling pathways that mediate the diverse stress-regulated cellular functions need to be identified. This review discusses the generation of ROS within tumour cells, their detoxification, their cellular effects, as well as the major signalling cascades they utilize, but also provides an outlook on their modulation in therapeutics.
Collapse
Affiliation(s)
- Geou-Yarh Liou
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville FL 32224, USA
| | | |
Collapse
|
30
|
Jagannath S, Kyle RA, Palumbo A, Siegel DS, Cunningham S, Berenson J. The Current Status and Future of Multiple Myeloma in the Clinic. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2010; 10:28-43. [DOI: 10.3816/clml.2010.n.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Monge M, Doll A, Colas E, Gil-Moreno A, Castellvi J, Garcia A, Colome N, Perez-Benavente A, Pedrola N, Lopez-Lopez R, Dolcet X, Ramon y Cajal S, Xercavins J, Matias-Guiu X, Canals F, Reventos J, Abal M. Subtractive proteomic approach to the endometrial carcinoma invasion front. J Proteome Res 2010; 8:4676-84. [PMID: 19691290 DOI: 10.1021/pr900390t] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Tumor invasion defines the transition between tissue-restricted carcinomas, related to good outcome as optimal surgery becomes possible, and metastatic tumors associated with poor prognosis and a dramatic decrease in survival. In endometrial cancer, myometrial infiltration represents a determinant parameter highly valuable in prognosis. To date, the identification of proteins involved in endometrial carcinoma invasion has been essentially conducted by immunohistochemical methods, without a global perception on the invasive front. Laser microdissection presents nowadays limitations to the profound spatiotemporal regulation from both the tumor and the surrounding stroma occurring at the invasive front. In this work, we attempted an alternative proteomic approach to characterize specific components of the tumor invasive front or its reactive stroma, by comparing the invasive area of an endometrial carcinoma with the noninvasive superficial tumor area and normal tissue from the same patients. This strategy led us to identify proteins involved in cellular morphology, assembly and movement, differentially expressed at the invasive front, as well as pathways like cell-to-cell signaling and interaction and a modulated response to oxidative stress as events related to endometrial carcinoma invasion. In conclusion, we could identify new players of myometrial infiltration by applying a subtractive proteomic approach to the endometrial carcinoma invasion front.
Collapse
Affiliation(s)
- Marta Monge
- Biomedical Research Unit, Research Institute Vall d'Hebron University Hospital, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Jagannath S, Kyle RA, Palumbo A, Siegel DS, Cunningham S, Berenson J. The Current Status and Future of Multiple Myeloma in the Clinic. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2009. [DOI: 10.3816/clm.2010.n.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Shah JJ, Orlowski RZ. Proteasome inhibitors in the treatment of multiple myeloma. Leukemia 2009; 23:1964-79. [PMID: 19741722 PMCID: PMC4737506 DOI: 10.1038/leu.2009.173] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 06/16/2009] [Accepted: 06/29/2009] [Indexed: 01/09/2023]
Abstract
Targeting intracellular protein turnover by inhibiting the ubiquitin-proteasome pathway as a strategy for cancer therapy is a new addition to our chemotherapeutic armamentarium, and has seen its greatest successes against multiple myeloma. The first-in-class proteasome inhibitor, bortezomib, was initially approved for treatment of patients in the relapsed/refractory setting as a single agent, and was recently shown to induce even greater benefits as part of rationally designed combinations that overcome chemoresistance. Modulation of proteasome function is also a rational approach to achieve chemosensitization to other antimyeloma agents, and bortezomib has now been incorporated into the front-line setting. Bortezomib-based induction regimens are able to achieve higher overall response rates and response qualities than was the case with prior standards of care, and unlike these older approaches, maintain efficacy in patients with clinically and molecularly defined high-risk disease. Second-generation proteasome inhibitors with novel properties, such as NPI-0052 and carfilzomib, are entering the clinical arena, and showing evidence of antimyeloma activity. In this spotlight review, we provide an overview of the current state of the art use of bortezomib and other proteasome inhibitors against multiple myeloma, and highlight areas for future study that will further optimize our ability to benefit patients with this disease.
Collapse
Affiliation(s)
- Jatin J. Shah
- The University of Texas M. D. Anderson Cancer Center, Department of Lymphoma & Myeloma, Houston, TX
| | - Robert Z. Orlowski
- The University of Texas M. D. Anderson Cancer Center, Department of Lymphoma & Myeloma, Houston, TX
- The University of Texas M. D. Anderson Cancer Center, Department of Experimental Therapeutics, Division of Cancer Medicine, Houston, TX
| |
Collapse
|
34
|
Han J, Ma I, Hendzel MJ, Allalunis-Turner J. The cytotoxicity of gamma-secretase inhibitor I to breast cancer cells is mediated by proteasome inhibition, not by gamma-secretase inhibition. Breast Cancer Res 2009; 11:R57. [PMID: 19660128 PMCID: PMC2750119 DOI: 10.1186/bcr2347] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 07/02/2009] [Accepted: 08/06/2009] [Indexed: 02/06/2023] Open
Abstract
Introduction Notch is a family of transmembrane protein receptors whose activation requires proteolytic cleavage by γ-secretase. Since aberrant Notch signaling can induce mammary carcinomas in transgenic mice and high expression levels of Notch receptors and ligands correlates with overall poor clinical outcomes, inhibiting γ-secretase with small molecules may be a promising approach for breast cancer treatment. Consistent with this hypothesis, two recent papers reported that γ-secretase inhibitor I (GSI I), Z-LLNle-CHO, is toxic to breast cancer cells both in vitro and in vivo. In this study, we compared the activity and cytotoxicity of Z-LLNle-CHO to that of two highly specific GSIs, DAPT and L-685,458 and three structurally unrelated proteasome inhibitors, MG132, lactacystin, and bortezomib in order to study the mechanism underlying the cytotoxicity of Z-LLNle-CHO in breast cancer cells. Methods Three estrogen receptor (ER) positive cell lines, MCF-7, BT474, and T47D, and three ER negative cell lines, SKBR3, MDA-MB-231, and MDA-MB-468, were used in this study. Both SKBR3 and BT474 cells also overexpress HER2/neu. Cytotoxicity was measured by using an MTS cell viability/proliferation assay. Inhibition of γ-secretase activity was measured by both immunoblotting and immunofluorescent microscopy in order to detect active Notch1 intracellular domain. Proteasome inhibition was determined by using a cell-based proteasome activity assay kit, by immunoblotting to detect accumulation of polyubiquitylated protein, and by immunofluorescent microscopy to detect redistribution of cellular ubiquitin. Results We found that blocking γ-secretase activity by DAPT and L-685,458 had no effect on the survival and proliferation of a panel of six breast cancer cell lines while Z-LLNle-CHO could cause cell death even at concentrations that inhibited γ-secretase activity less efficiently. Furthermore, we observed that Z-LLNle-CHO could inhibit proteasome activity and the relative cellular sensitivity of these six breast cancer cell lines to Z-LLNle-CHO was the same as observed for three proteasome inhibitors. Finally, we found that the cell killing effect of Z-LLNle-CHO could be reversed by a chemical that restored the proteasome activity. Conclusions We conclude that the cytotoxicity of Z-LLNle-CHO in breast cancer cells is mediated by proteasome inhibition, not by γ-secretase inhibition.
Collapse
Affiliation(s)
- Jianxun Han
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta, Canada T6G 1Z2.
| | | | | | | |
Collapse
|
35
|
Byun JY, Kim MJ, Eum DY, Yoon CH, Seo WD, Park KH, Hyun JW, Lee YS, Lee JS, Yoon MY, Lee SJ. Reactive oxygen species-dependent activation of Bax and poly(ADP-ribose) polymerase-1 is required for mitochondrial cell death induced by triterpenoid pristimerin in human cervical cancer cells. Mol Pharmacol 2009; 76:734-44. [PMID: 19574249 DOI: 10.1124/mol.109.056259] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Naturally occurring triterpenoid compounds have long been used as anti-inflammatory, antimalarial, and insecticidal agents. It has become evident that some of the natural or synthetic triterpenoids have promising clinical potential as both a therapeutic and chemopreventive agent for cancer. However, the molecular basis for the antitumor activity of triterpenoid has yet to be defined. In this study, we show that pristimerin, a natural triterpenoid, induces mitochondrial cell death in human cervical cancer cells and that reactive oxygen species (ROS)-dependent activation of both Bax and poly(ADP-ribose) polymerase-1 (PARP-1) is critically required for the mitochondrial dysfunction. We also showed that c-Jun N-terminal kinase (JNK) is involved in ROS-dependent Bax activation. Treatment of pristimerin induced an increase in intracellular ROS, JNK activation, conformational change, and mitochondrial redistribution of Bax, mitochondrial membrane potential loss, and cell death. The PARP-1 was also found to be activated by pristimerin treatment. An antioxidant, N-acetyl-l-cysteine (NAC), inhibited pristimerin-induced JNK activation, Bax relocalization, and PARP-1 activation, as well as mitochondrial cell death. Moreover, inhibition of JNK clearly suppressed conformational change and mitochondrial translocation of Bax and subsequent mitochondrial cell death but did not affect PARP-1 activation. Inhibition of PARP-1 with 1,5-dihydroxyisoquinoline (DIQ) or with small interfering RNA of PARP-1 significantly attenuated pristimerin-induced mitochondrial membrane potential loss and cell death but did not affect JNK activation and Bax relocalization. These results indicate that the natural triterpenoid pristimerin induces mitochondrial cell death through ROS-dependent activation of both Bax and PARP-1 in human cervical cancer cells and that JNK is involved in ROS-dependent Bax activation.
Collapse
Affiliation(s)
- Joo-Yun Byun
- Department of Chemistry, Hanyang University, Seoul 133-791, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov 2009; 8:579-91. [PMID: 19478820 DOI: 10.1038/nrd2803] [Citation(s) in RCA: 4024] [Impact Index Per Article: 268.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Increased generation of reactive oxygen species (ROS) and an altered redox status have long been observed in cancer cells, and recent studies suggest that this biochemical property of cancer cells can be exploited for therapeutic benefits. Cancer cells in advanced stage tumours frequently exhibit multiple genetic alterations and high oxidative stress, suggesting that it might be possible to preferentially eliminate these cells by pharmacological ROS insults. However, the upregulation of antioxidant capacity in adaptation to intrinsic oxidative stress in cancer cells can confer drug resistance. Abrogation of such drug-resistant mechanisms by redox modulation could have significant therapeutic implications. We argue that modulating the unique redox regulatory mechanisms of cancer cells might be an effective strategy to eliminate these cells.
Collapse
|
37
|
Chandra J. Oxidative stress by targeted agents promotes cytotoxicity in hematologic malignancies. Antioxid Redox Signal 2009; 11:1123-37. [PMID: 19018667 PMCID: PMC2842131 DOI: 10.1089/ars.2008.2302] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The past decade has seen an exponential increase in the number of cancer therapies with defined molecular targets. Interestingly, many of these new agents are also documented to raise levels of intracellular reactive oxygen species (ROS) in addition to inhibiting a biochemical target. In most cases, the exact link between the primary target of the drug and effects on cellular redox status is unknown. However, it is important to understand the role of oxidative stress in promoting cytotoxicity by these agents, because the design of multiregimen strategies could conceivably build on these redox alterations. Also, drug resistance mediated by antioxidant defenses could potentially be anticipated and circumvented with improved knowledge of the redox-related effects of these targeted agents. Given the large number of targeted chemotherapies, in this review, we focus on selected agents that have shown promise in hematologic malignancies: proteasome inhibitors, histone deacetylase inhibitors, Bcl-2-targeted agents, and a kinase inhibitor called adaphostin. Despite structural differences within classes of these compounds, a commonality of causing increased oxidative stress exists, which contributes to induction of cell death.
Collapse
Affiliation(s)
- Joya Chandra
- Department of Pediatrics Research, Unit 853, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA.
| |
Collapse
|
38
|
Perrone G, Hideshima T, Ikeda H, Okawa Y, Calabrese E, Gorgun G, Santo L, Cirstea D, Raje N, Chauhan D, Baccarani M, Cavo M, Anderson KC. Ascorbic acid inhibits antitumor activity of bortezomib in vivo. Leukemia 2009; 23:1679-86. [PMID: 19369963 DOI: 10.1038/leu.2009.83] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Earlier studies have shown that ascorbic acid (vitamin C) inhibits bortezomib-induced cytotoxicity against cancer cells in vitro. However, the clinical significance of vitamin C on bortezomib treatment is unclear. In this study, we examined whether daily oral intake of vitamin C inhibits antimultiple myeloma (MM) activities of bortezomib. Vitamin C, at orally achievable concentrations, inhibited in vitro MM cell cytotoxicity of bortezomib and blocked its inhibitory effect on 20S proteasome activity. Specifically, plasma collected from healthy volunteers taking 1 g/day vitamin C reduced bortezomib-induced MM cell death in vitro. This antagonistic effect of vitamin C against proteasome inhibitors is limited to the boronate class of inhibitors (bortezomib and MG262). In vivo activity of this combination treatment was then evaluated using our xenograft model of human MM in SCID (severe combined immune-deficient) mice. Bortezomib (0.1 mg/kg twice a week for 4 weeks) significantly inhibits in vivo MM cell growth, which was blocked by oral vitamin C (40 mg/kg/day). Therefore, our results for the first time show that vitamin C can significantly reduce the activity of bortezomib treatment in vivo; and importantly, suggest that patients receiving treatment with bortezomib should avoid taking vitamin C dietary supplements.
Collapse
Affiliation(s)
- G Perrone
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Green tea polyphenols block the anticancer effects of bortezomib and other boronic acid-based proteasome inhibitors. Blood 2009; 113:5927-37. [PMID: 19190249 DOI: 10.1182/blood-2008-07-171389] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The anticancer potency of green tea and its individual components is being intensely investigated, and some cancer patients already self-medicate with this "miracle herb" in hopes of augmenting the anticancer outcome of their chemotherapy. Bortezomib (BZM) is a proteasome inhibitor in clinical use for multiple myeloma. Here, we investigated whether the combination of these compounds would yield increased antitumor efficacy in multiple myeloma and glioblastoma cell lines in vitro and in vivo. Unexpectedly, we discovered that various green tea constituents, in particular (-)-epigallocatechin gallate (EGCG) and other polyphenols with 1,2-benzenediol moieties, effectively prevented tumor cell death induced by BZM in vitro and in vivo. This pronounced antagonistic function of EGCG was evident only with boronic acid-based proteasome inhibitors (BZM, MG-262, PS-IX), but not with several non-boronic acid proteasome inhibitors (MG-132, PS-I, nelfinavir). EGCG directly reacted with BZM and blocked its proteasome inhibitory function; as a consequence, BZM could not trigger endoplasmic reticulum stress or caspase-7 activation, and did not induce tumor cell death. Taken together, our results indicate that green tea polyphenols may have the potential to negate the therapeutic efficacy of BZM and suggest that consumption of green tea products may be contraindicated during cancer therapy with BZM.
Collapse
|
40
|
Ortega E, Marti RM, Yeramian A, Sorolla A, Dolcet X, Llobet D, Abal L, Santacana M, Pallares J, Llombart-Cussac A, Matias-Guiu X. Targeted therapies in gynecologic cancers and melanoma. Semin Diagn Pathol 2008; 25:262-73. [PMID: 19013892 DOI: 10.1053/j.semdp.2008.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The article reviews the main molecular pathology alterations of endometrial and ovarian carcinomas and melanoma. Several promising drugs targeting the genes most frequently altered in these tumors are under consideration. The most promising signaling pathways to be targeted for therapies in these tumors are the tyrosine kinase receptor (EGFR, HER2, c-KIT), the RAS/B-RAF/MAPK, the PI3K-mTOR, and apoptosis signaling pathways.
Collapse
Affiliation(s)
- Eugenia Ortega
- Department of Medical Oncology, Dermatology, and Pathology, Hospital Universitari Arnau de Vilanova, University of Lleida-IRB Lleida, Lleida, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|