1
|
Wang X, He J, Sun M, Wang S, Qu J, Shi H, Rao B. High-dose vitamin C as a metabolic treatment of cancer: a new dimension in the era of adjuvant and intensive therapy. Clin Transl Oncol 2025; 27:1366-1382. [PMID: 39259387 DOI: 10.1007/s12094-024-03553-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/04/2024] [Indexed: 09/13/2024]
Abstract
The anti-cancer mechanism of High-dose Vitamin C (HDVC) is mainly to participate in the Fenton reaction, hydroxylation reaction, and epigenetic modification, which leads to the energy crisis, metabolic collapse, and severe peroxidation stress that results in the proliferation inhibition or death of cancer cells. However, the mainstream view is that HDVC does not significantly improve cancer treatment outcomes. In clinical work and scientific research, we found that some drugs or therapies can significantly improve the anti-cancer effects of HDVC, such as PD-1 inhibitors that can increase the anti-cancer effects of cancerous HDVC by nearly three times. Here, the adjuvant and intensive therapy and synergistic mechanisms including HDVC combined application of chemoradiotherapies multi-vitamins, targeted drugs, immunotherapies, and oncolytic virus are discussed in detail. Adjuvant and intensive therapy of HDVC can significantly improve the therapeutic effect of HDVC in the metabolic treatment of cancer, but more clinical evidence is needed to support its clinical application.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jia He
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Minmin Sun
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shiwan Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jinxiu Qu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Hanping Shi
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China.
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China.
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| | - Benqiang Rao
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China.
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China.
| |
Collapse
|
2
|
Agathocleous M. The physiological functions of ascorbate in the development of cancer. Dis Model Mech 2025; 18:dmm052201. [PMID: 40213851 PMCID: PMC12010911 DOI: 10.1242/dmm.052201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025] Open
Abstract
The metabolite ascorbate (vitamin C) is synthesized endogenously in most animals or, in humans and some other species, obtained from the diet. Its role in cancer development is controversial. Addition of ascorbate to cultured cells or high-dose administration in animals can inhibit growth of many cancers, but most of these effects are caused by non-physiological biochemical activities. Few experiments have tested the physiological roles of ascorbate in cancer development by depleting it in physiological settings. Ascorbate depletion inhibits the activity of ten-eleven translocation (TET) enzymes in hematopoietic and leukemia cells and accelerates myeloid leukemia development. Many clinical trials have tested ascorbate supplementation in cancers and shown little or no evidence that it has a beneficial role. I propose that depletion experiments are needed to define the cancers in which ascorbate has a physiological role, establish its cellular and molecular targets, and provide a rationale for clinical trials.
Collapse
Affiliation(s)
- Michalis Agathocleous
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
3
|
Gencheva R, Coppo L, Arnér ESJ, Ren X. Selenium supplementation protects cancer cells from the oxidative stress and cytotoxicity induced by the combination of ascorbate and menadione sodium bisulfite. Free Radic Biol Med 2025; 233:317-329. [PMID: 40180024 DOI: 10.1016/j.freeradbiomed.2025.03.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
The combination of ascorbate (vitamin C) and menadione sodium bisulfite (MSB, vitamin K3), here called VC/VK3 (also named Apatone®, or M/A), has shown selective cytotoxicity in cancer cells and is under clinical investigation as a cancer therapy. However, the mechanisms of VC/VK3-induced cell death are not fully understood. In this in vitro study using human glioblastoma and non-transformed glial cell lines, we found that VC/VK3 caused higher toxicity in cancer cells in an H2O2- and iron-dependent manner, suggesting that ferroptosis may play a role in the cell death process. Furthermore, selenium supplementation significantly protected cancer cells from VC/VK3 treatment concomitantly with enhanced expression levels and enzymatic activity of antioxidant selenoproteins, including thioredoxin reductases (TXNRDs) and glutathione reductases (GPXs). We also found that VC/VK3 competes for electrons with thioredoxin (TXN), impairing peroxiredoxin 1 (PRDX1) in cells. Finally, chemically inhibiting TXNRDs or the glutathione-dependent antioxidant systems exaggerated the toxicity of VC/VK3. Overall, this study elucidated parts of the cell death mechanisms of VC/VK3 and identified combination strategies to overcome selenium-mediated resistance, advancing the translational potential of this prooxidant treatment.
Collapse
Affiliation(s)
- Radosveta Gencheva
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Lucia Coppo
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; Department of Selenoprotein Research, National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary
| | - Xiaoyuan Ren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; IC-MedTech Corporation, Las Vegas, NV, USA.
| |
Collapse
|
4
|
Pulliam CF, Fath MA, Sho S, Johnson ST, Wagner BA, Singhania M, Kalen AL, Bayanbold K, Solst SR, Allen BG, George BN, Caster JM, Buettner GR, Riley DP, Keene JL, Beardsley RA, Spitz DR. Pharmacological ascorbate combined with rucosopasem selectively radio-chemo-sensitizes NSCLC via generation of H 2O 2. Redox Biol 2025; 80:103505. [PMID: 39884000 PMCID: PMC11830350 DOI: 10.1016/j.redox.2025.103505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Differences in cancer and normal cell oxidative metabolism provide a unique therapeutic opportunity for developing combined modality approaches with redox-active small molecules as radio-chemosensitizers that are well-tolerated by normal tissues. Pentaazamacrocyclic Mn (II)-containing (MnPAM) superoxide dismutase (SOD) mimetics and pharmacological ascorbate given IV to achieve [mM] plasma levels (pharmacological ascorbate: P-AscH‾) have been shown to act individually as cancer cell radio- and chemosensitizers via the generation of H2O2in vivo. The current study shows that the combination of newly developed MnPAM dismutase mimetic, rucosopasem manganese (RUC) with P-AscH‾ radio-sensitizes non-small cell lung cancer cells (NSCLC) and increases steady state levels of intracellular H2O2 with no additional toxicity to normal human bronchial epithelial cells (HBECs). Conditional over expression of catalase (CAT) in H1299T CATc15 cells demonstrates that the combination of RUC and P-AscH‾ causes radio-sensitization through an H2O2-dependent mechanism. Interestingly, RUC combined with P-AscH‾ demonstrates more than additive cytotoxicity in both H1299T and A549 NSCLC cells, but conditional over-expression of ferritin heavy chain (FtH) protected only the H1299T, and not the A549, from this toxicity. Most importantly, the combination of RUC + P-AscH‾ was found to sensitize both H1299T and A549 cell types to radio-chemotherapy with cisplatin (CIS) + etoposide (ETOP). Finally, in H1299T NSCLC xenografts the combination of RUC + P-AscH‾ with CIS + ETOP and 12 × 2 Gy radiation significantly inhibits tumor growth and increased median overall over survival. These results support the hypothesis that selective MnPAM dismutase mimetic + P-AscH‾ enhances the efficacy of radio-chemotherapy in NSCLC through a mechanism governed by redox active metals and H2O2 production.
Collapse
Affiliation(s)
- C F Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA.
| | - M A Fath
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - S Sho
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - S T Johnson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - B A Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - M Singhania
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - A L Kalen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - K Bayanbold
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - S R Solst
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - B G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA.
| | - B N George
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - J M Caster
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - G R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA.
| | - D P Riley
- Galera Therapeutics, Malvern, PA, 19355, USA.
| | - J L Keene
- Galera Therapeutics, Malvern, PA, 19355, USA.
| | | | - D R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
5
|
Kim HS, Kwon SH, Choi OK, Lim T. High-dose ascorbic acid synergizes with anti-PD1 therapy in non-small cell lung cancer in vitro and in vivo models. Front Immunol 2025; 15:1512605. [PMID: 39896806 PMCID: PMC11783322 DOI: 10.3389/fimmu.2024.1512605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction Immune checkpoint inhibitors(ICIs) targeting programmed cell death protein 1 (PD1) confer significant survival benefits to patients with non-small cell lung cancer (NSCLC). However, there remains a substantial unmet need to identify therapeutic approaches to overcome resistance and provide benefits to these patients. High-dose ascorbic acid (AA) acts synergistically with many standard anticancer treatments. However, little is known about the effect of high-dose AA on improving the efficacy of anti-PD1 inhibitors in NSCLC. This study aimed to elucidate the effects of high-dose AA on anti-PD1 immunotherapy in NSCLC. Methods The combined effects of high-dose AA and anti-PD1 were investigated using a coculture model of H460 cells and CD8+ T cells and an LLC1 lung cancer syngeneic mouse model. To investigate the molecular mechanism, tumor tissues from mice were analyzed by comprehensive proteomic profiling using nano-LC-ESI-MS/MS. Results Pretreatment with a high dose of AA led to enhanced the sensitivity to the cytotoxicity of CD8+ T cells derived from healthy donor for H460 cells. Additionally, the combination of anti-PD1 and high-dose AA significantly increased CD8+ T cell cytotoxicity in H460 cells. The combination of anti-PD1 and high-dose AA showed dramatic antitumor effects in a syngeneic mouse model of lung cancer by significantly reducing tumor growth and increasing CD8+ T cell-dependent cytotoxicity and macrophage activity. Comprehensive protein analysis confirmed that high-dose AA in anti-PD1-treated tumor tissues enhanced the antitumor effects by regulating various immune-related mechanisms, including the B cell and T cell receptor signaling pathways, Fc gamma R-mediated phagocytosis, and natural killer (NK) cell-mediated cytotoxicity. Discussion Our results suggest that high-dose AA may be a promising adjuvant to potentiate the efficacy of anti-PD1 immunotherapy.
Collapse
Affiliation(s)
- Hak Su Kim
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Seung-hyun Kwon
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Ok Kyung Choi
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Taekyu Lim
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
- Division of Hematology-Oncology, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| |
Collapse
|
6
|
Papavassiliou KA, Sofianidi AA, Gogou VA, Papavassiliou AG. Leveraging the ROS-TME Axis for Cancer Treatment. Antioxidants (Basel) 2024; 13:1365. [PMID: 39594507 PMCID: PMC11591396 DOI: 10.3390/antiox13111365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
The discovery of reactive oxygen species (ROS) dates back to the early 20th century [...].
Collapse
Affiliation(s)
- Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (V.A.G.)
| | - Amalia A. Sofianidi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassiliki A. Gogou
- First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (V.A.G.)
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
7
|
Nazari A, Osati P, Seifollahy Fakhr S, Faghihkhorasani F, Ghanaatian M, Faghihkhorasani F, Rezaei-Tazangi F, Pazhouhesh Far N, Shourideh A, Ebrahimi N, Aref AR. New Emerging Therapeutic Strategies Based on Manipulation of the Redox Regulation Against Therapy Resistance in Cancer. Antioxid Redox Signal 2024. [PMID: 39506926 DOI: 10.1089/ars.2023.0491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Background: Resistance to standard therapeutic methods, including chemotherapy, immunotherapy, and targeted therapy, remains a critical challenge in effective cancer treatment. Redox homeostasis modification has emerged as a promising approach to address medication resistance. Objective: This review aims to explore the mechanisms of redox alterations and signaling pathways contributing to treatment resistance in cancer. Methods: In this study, a comprehensive review of the molecular mechanisms underlying drug resistance governed by redox signaling was conducted. Emphasis was placed on understanding how tumor cells manage increased reactive oxygen species (ROS) levels through upregulated antioxidant systems, enabling resistance across multiple therapeutic pathways. Results: Key mechanisms identified include alterations in drug efflux, target modifications, metabolic changes, enhanced DNA damage repair, stemness preservation, and tumor microenvironment remodeling. These pathways collectively facilitate tumor cells' adaptive response and resistance to various cancer treatments. Conclusion: Developing a detailed understanding of the interrelationships between these redox-regulated mechanisms and therapeutic resistance holds potential to improve treatment effectiveness, offering valuable insights for both fundamental and clinical cancer research. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Parisa Osati
- Department of Chemical Engineering, Fouman Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Siavash Seifollahy Fakhr
- Department of Biotechnology, Faculty of Applied Ecology, Agricultural Science and Biotechnology, Campus Hamar, Norway
| | - Ferdos Faghihkhorasani
- Department of Cardiology, Internal Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, Shaanxi Province, 710061, China
| | - Masoud Ghanaatian
- Master 1 Bio-Santé-Parcours Toulouse Graduate School of Cancer, Ageing and Rejuvenation (CARe), Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Fereshteh Faghihkhorasani
- General Physician in Medicine Program,General Doctorate Degree of Yazd Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Amir Shourideh
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA and Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Bodeker KL, Smith BJ, Berg DJ, Chandrasekharan C, Sharif S, Fei N, Vollstedt S, Brown H, Chandler M, Lorack A, McMichael S, Wulfekuhle J, Wagner BA, Buettner GR, Allen BG, Caster JM, Dion B, Kamgar M, Buatti JM, Cullen JJ. A randomized trial of pharmacological ascorbate, gemcitabine, and nab-paclitaxel for metastatic pancreatic cancer. Redox Biol 2024; 77:103375. [PMID: 39369582 PMCID: PMC11491967 DOI: 10.1016/j.redox.2024.103375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have poor 5-year survival. Pharmacological ascorbate (P-AscH-, high dose, intravenous, vitamin C) has shown promise as an adjunct to chemotherapy. We hypothesized adding P-AscH- to gemcitabine and nab-paclitaxel would increase survival in patients with metastatic PDAC. METHODS Patients diagnosed with stage IV pancreatic cancer randomized 1:1 to gemcitabine and nab-paclitaxel only (SOC, control) or to SOC with concomitant P-AscH-, 75 g three times weekly (ASC, investigational). The primary outcome was overall survival with secondary objectives of determining progression-free survival and adverse event incidence. Quality of life and patient reported outcomes for common oncologic symptoms were captured as an exploratory objective. Thirty-six participants were randomized; of this 34 received their assigned study treatment. All analyses were based on data frozen on December 11, 2023. RESULTS Intravenous P-AscH- increased serum ascorbate levels from micromolar to millimolar levels. P-AscH- added to the gemcitabine + nab-paclitaxel (ASC) increased overall survival to 16 months compared to 8.3 months with gemcitabine + nab-paclitaxel (SOC) (HR = 0.46; 90 % CI 0.23, 0.92; p = 0.030). Median progression free survival was 6.2 (ASC) vs. 3.9 months (SOC) (HR = 0.43; 90 % CI 0.20, 0.92; p = 0.029). Adding P-AscH- did not negatively impact quality of life or increase the frequency or severity of adverse events. CONCLUSIONS P-AscH- infusions of 75 g three times weekly in patients with metastatic pancreatic cancer prolongs overall and progression free survival without detriment to quality of life or added toxicity (ClinicalTrials.gov number NCT02905578).
Collapse
Affiliation(s)
- Kellie L Bodeker
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Brian J Smith
- College of Public Health, The University of Iowa, Iowa City, IA, USA
| | - Daniel J Berg
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Chandrikha Chandrasekharan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Saima Sharif
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Naomi Fei
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Sandy Vollstedt
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Heather Brown
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Meghan Chandler
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Amanda Lorack
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Stacy McMichael
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Jared Wulfekuhle
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Brett A Wagner
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Garry R Buettner
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Bryan G Allen
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joseph M Caster
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Barbara Dion
- Medical College of Wisconsin Division of Hematology and Oncology, Milwaukee, WI, USA
| | - Mandana Kamgar
- Medical College of Wisconsin Division of Hematology and Oncology, Milwaukee, WI, USA
| | - John M Buatti
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joseph J Cullen
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA; Department of Surgery, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
9
|
Vaishampayan P, Lee Y. Redox-active vitamin C suppresses human osteosarcoma growth by triggering intracellular ROS-iron-calcium signaling crosstalk and mitochondrial dysfunction. Redox Biol 2024; 75:103288. [PMID: 39083898 PMCID: PMC11342202 DOI: 10.1016/j.redox.2024.103288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Pharmacological vitamin C (VC) has gained attention for its pro-oxidant characteristics and selective ability to induce cancer cell death. However, defining its role in cancer has been challenging due to its complex redox properties. In this study, using a human osteosarcoma (OS) model, we show that the redox-active property of VC is critical for inducing non-apoptotic cancer cell death via intracellular reactive oxygen species (ROS)-iron-calcium crosstalk and mitochondrial dysfunction. In both 2D and 3D OS cell culture models, only the oxidizable form of VC demonstrated potent dose-dependent cytotoxicity, while non-oxidizable and oxidized VC derivatives had minimal effects. Live-cell imaging showed that only oxidizable VC caused a surge in cytotoxic ROS, dependent on iron rather than copper. Inhibitors of ferroptosis, a form of iron-dependent cell death, along with classical apoptosis inhibitors, were unable to completely counteract the cytotoxic effects induced by VC. Further pharmacological and genetic inhibition analyses showed that VC triggers calcium release through inositol 1,4,5-trisphosphate receptors (IP3Rs), leading to mitochondrial ROS production and eventual cell death. RNA sequencing revealed down-regulation of genes involved in the mitochondrial electron transport chain and oxidative phosphorylation upon pharmacological VC treatment. Consistently, high-dose VC reduced mitochondrial membrane potential, oxidative phosphorylation, and ATP levels, with ATP reconstitution rescuing VC-induced cytotoxicity. In vivo OS xenograft studies demonstrated reduced tumor growth with high-dose VC administration, concomitant with the altered expression of mitochondrial ATP synthase (MT-ATP). These findings emphasize VC's potential clinical utility in osteosarcoma treatment by inducing mitochondrial metabolic dysfunction through a vicious intracellular ROS-iron-calcium cycle.
Collapse
Affiliation(s)
- Prajakta Vaishampayan
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| | - Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA; Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, 99202, USA; Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
10
|
Zaher A, Petronek MS, Allen BG, Mapuskar KA. Balanced Duality: H 2O 2-Based Therapy in Cancer and Its Protective Effects on Non-Malignant Tissues. Int J Mol Sci 2024; 25:8885. [PMID: 39201571 PMCID: PMC11354297 DOI: 10.3390/ijms25168885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
Conventional cancer therapy strategies, although centered around killing tumor cells, often lead to severe side effects on surrounding normal tissues, thus compromising the chronic quality of life in cancer survivors. Hydrogen peroxide (H2O2) is a secondary signaling molecule that has an array of functions in both tumor and normal cells, including the promotion of cell survival pathways and immune cell modulation in the tumor microenvironment. H2O2 is a reactive oxygen species (ROS) crucial in cellular homeostasis and signaling (at concentrations maintained under nM levels), with increased steady-state levels in tumors relative to their normal tissue counterparts. Increased steady-state levels of H2O2 in tumor cells, make them vulnerable to oxidative stress and ultimately, cell death. Recently, H2O2-producing therapies-namely, pharmacological ascorbate and superoxide dismutase mimetics-have emerged as compelling complementary treatment strategies in cancer. Both pharmacological ascorbate and superoxide dismutase mimetics can generate excess H2O2 to overwhelm the impaired H2O2 removal capacity of cancer cells. This review presents an overview of H2O2 metabolism in the physiological and malignant states, in addition to discussing the anti-tumor and normal tissue-sparing mechanism(s) of, and clinical evidence for, two H2O2-based therapies, pharmacological ascorbate and superoxide dismutase mimetics.
Collapse
Affiliation(s)
| | | | | | - Kranti A. Mapuskar
- Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA; (A.Z.); (M.S.P.); (B.G.A.)
| |
Collapse
|
11
|
Glorieux C, Liu S, Trachootham D, Huang P. Targeting ROS in cancer: rationale and strategies. Nat Rev Drug Discov 2024; 23:583-606. [PMID: 38982305 DOI: 10.1038/s41573-024-00979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
Reactive oxygen species (ROS) in biological systems are transient but essential molecules that are generated and eliminated by a complex set of delicately balanced molecular machineries. Disruption of redox homeostasis has been associated with various human diseases, especially cancer, in which increased ROS levels are thought to have a major role in tumour development and progression. As such, modulation of cellular redox status by targeting ROS and their regulatory machineries is considered a promising therapeutic strategy for cancer treatment. Recently, there has been major progress in this field, including the discovery of novel redox signalling pathways that affect the metabolism of tumour cells as well as immune cells in the tumour microenvironment, and the intriguing ROS regulation of biomolecular phase separation. Progress has also been made in exploring redox regulation in cancer stem cells, the role of ROS in determining cell fate and new anticancer agents that target ROS. This Review discusses these research developments and their implications for cancer therapy and drug discovery, as well as emerging concepts, paradoxes and future perspectives.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shihua Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Metabolic Innovation Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
12
|
Chen G, Li MY, Yang JY, Zhou ZH. Will AMPK be a potential therapeutic target for hepatocellular carcinoma? Am J Cancer Res 2024; 14:3241-3258. [PMID: 39113872 PMCID: PMC11301289 DOI: 10.62347/yavk1315] [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: 03/06/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer is the disease that poses the greatest threat to human health today. Among them, hepatocellular carcinoma (HCC) is particularly prominent due to its high recurrence rate and extremely low five-year postoperative survival rate. In addition to surgical treatment, radiotherapy, chemotherapy, and immunotherapy are the main methods for treating HCC. Due to the natural drug resistance of chemoradiotherapy and targeted drugs, satisfactory results have not been achieved in terms of therapeutic efficacy and cost. AMP-Activated Protein Kinase (AMPK) is a serine/threonine protein kinase. It mainly coordinates the metabolism and transformation of energy between cells, which maintaining a balance between energy supply and demand. The processes of cell growth, proliferation, autophagy, and survival all involve various reaction of cells to energy changes. The regulatory role of AMPK in cellular energy metabolism plays an important role in the occurrence, development, treatment, and prognosis of HCC. Here, we reviewed the latest progress on the regulatory role of AMPK in the occurrence and development of HCC. Firstly, the molecular structure and activation mechanism of AMPK were introduced. Secondly, the emerging regulator related to AMPK and tumors were elaborated. Next, the multitasking roles of AMPK in the occurrence and development mechanism of HCC were discussed separately. Finally, the translational implications and the challenges of AMPK-targeted therapies for HCC treatment were elaborated. In summary, these pieces of information suggest that AMPK can serve as a promising specific therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Guo Chen
- Department of Oncology, Anhui Hospital, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineHefei, Anhui, China
| | - Ming-Yuan Li
- Department of Oncology, Anhui Hospital, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineHefei, Anhui, China
| | - Jing-Yi Yang
- Department of Oncology, Feixi Hospital of Traditional Chinese MedicineFeixi, Hefei, Anhui, China
| | - Zhen-Hua Zhou
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineShanghai, China
| |
Collapse
|
13
|
Talib WH, Ahmed Jum’AH DA, Attallah ZS, Jallad MS, Al Kury LT, Hadi RW, Mahmod AI. Role of vitamins A, C, D, E in cancer prevention and therapy: therapeutic potentials and mechanisms of action. Front Nutr 2024; 10:1281879. [PMID: 38274206 PMCID: PMC10808607 DOI: 10.3389/fnut.2023.1281879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/09/2023] [Indexed: 01/27/2024] Open
Abstract
Cancer, a leading global cause of mortality, arises from intricate interactions between genetic and environmental factors, fueling uncontrolled cell growth. Amidst existing treatment limitations, vitamins have emerged as promising candidates for cancer prevention and treatment. This review focuses on Vitamins A, C, E, and D because of their protective activity against various types of cancer. They are essential as human metabolic coenzymes. Through a critical exploration of preclinical and clinical studies via PubMed and Google Scholar, the impact of these vitamins on cancer therapy was analyzed, unraveling their complicated mechanisms of action. Interestingly, vitamins impact immune function, antioxidant defense, inflammation, and epigenetic regulation, potentially enhancing outcomes by influencing cell behavior and countering stress and DNA damage. Encouraging clinical trial results have been observed; however, further well-controlled studies are imperative to validate their effectiveness, determine optimal dosages, and formulate comprehensive cancer prevention and treatment strategies. Personalized supplementation strategies, informed by medical expertise, are pivotal for optimal outcomes in both clinical and preclinical contexts. Nevertheless, conclusive evidence regarding the efficacy of vitamins in cancer prevention and treatment is still pending, urging further research and exploration in this compelling area of study.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Faculty of Allied Medical Sciences, Applied Science Private University, Amman, Jordan
| | | | - Zeena Shamil Attallah
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| | - Mohanned Sami Jallad
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| | - Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Rawan Wamidh Hadi
- Faculty of Allied Medical Sciences, Applied Science Private University, Amman, Jordan
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| |
Collapse
|
14
|
Du J, Filipović MR, Wagner BA, Buettner GR. Ascorbate mediates the non-enzymatic reduction of nitrite to nitric oxide. ADVANCES IN REDOX RESEARCH 2023; 9:100079. [PMID: 37692975 PMCID: PMC10486277 DOI: 10.1016/j.arres.2023.100079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Nitric oxide (NO•) generated by nitric oxide synthases is involved in many physiological and pathophysiological processes. However, non-enzymatic formation of NO• also occurs in vivo. Here we investigated the production of NO• from nitrite, as facilitated by ascorbate, over the pH range of 2.4-7.4. Using a nitric oxide electrode, we observed at low pH a rapid generation of NO• from nitrite and ascorbate that slows with increasing pH. The formation of NO• was confirmed by its reaction with oxyhemoglobin. In the ascorbate/nitrite system a steady-state level of NO• was achieved, suggesting that a futile redox cycle of nitrite-reduction by ascorbate and NO•-oxidation by dioxygen was established. However, at pH-values of around 7 and greater, the direct reduction of nitrite by ascorbate is very slow; thus, this route to the non-enzymatic production of NO• is not likely to be significant process in vivo in environments having a pH around 7.4. The production of nitric oxide by nitrite and ascorbate would be important only in areas of lower pH, e.g. stomach/digestive system, sites of inflammation, and areas of hypoxia such as tumor tissue. In patients receiving very large doses of ascorbate delivered by intravenous infusion, plasma levels of ascorbate on the order of 20 - 30 mM can be achieved. After infusion, levels of nitrate and nitrite in plasma were unchanged. Thus, in blood and tissue that maintain a pH of about 7.4, the reduction of nitrite to nitric oxide by ascorbate appears to be insignificant, even at very large, pharmacological levels of ascorbate.
Collapse
Affiliation(s)
- Juan Du
- Free Radical and Radiation Biology & ESR Facility, The University of Iowa, Med Labs B-180, Iowa City, IA, United States
| | | | - Brett A. Wagner
- Free Radical and Radiation Biology & ESR Facility, The University of Iowa, Med Labs B-180, Iowa City, IA, United States
| | - Garry R. Buettner
- Free Radical and Radiation Biology & ESR Facility, The University of Iowa, Med Labs B-180, Iowa City, IA, United States
| |
Collapse
|
15
|
Ou J, Liao Q, Du Y, Xi W, Meng Q, Li K, Cai Q, Pang CLK. SERPINE1 and SERPINB7 as potential biomarkers for intravenous vitamin C treatment in non-small-cell lung cancer. Free Radic Biol Med 2023; 209:96-107. [PMID: 37838303 DOI: 10.1016/j.freeradbiomed.2023.10.391] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
High dose intravenous vitamin C (IVC) has been proposed as a pro-oxidant anticancer agent. However, there is a lack of biomarkers that are specific for this treatment. Here, we explored profiles of gene expression responding to IVC treatment in non-small cell lung cancer (NSCLC) cells as an effort for potential biomarker discovery. Genome-wide RNA-seq was performed in human NSCLC cell lines treated with pharmacological concentrations of vitamin C(VitC) for differential expression of genes. The identified genes were analyzed for correlations with patient prognosis using data from the Kaplan-Meier Plotter and the Human Protein Atlas databases. Further, tumor samples from a retrospective study of 153 NSCLC patients were analyzed with immunohistochemistry for expression of targeted genes, and patient prognosis was correlated to these genes. Two genes, namely SERPINE1 and SERPINB7 were found to be downregulated in NSCLC cells following VitC treatment. Combined patient data from the cohort analysis and online databases revealed that these 2 genes presented an unfavorable prognostic prediction of overall survival (OS) in NSCLC patients receiving standard of care. However, high expression level of these 2 genes were associated with prolonged OS in NSCLC patients receiving IVC in addition to standard of care. These data revealed that SERPINE1 and SERPINB7 have the potential to serve as predictive factors indicating favorable responses to IVC treatment in patients with NSCLC. Further validations are warranted.
Collapse
Affiliation(s)
- Junwen Ou
- Cancer Center, Clifford Hospital, Jinan University, Guangzhou, PR China.
| | - Qiulin Liao
- Pathology Department, Clifford Hospital, Jinan University, Guangzhou, PR China
| | - Yanping Du
- Cancer Center, Clifford Hospital, Jinan University, Guangzhou, PR China
| | - Wentao Xi
- Cancer Center, Clifford Hospital, Jinan University, Guangzhou, PR China
| | - Qiong Meng
- Cancer Center, Clifford Hospital, Jinan University, Guangzhou, PR China
| | - Kexin Li
- Imaging Department, Clifford Hospital, Jinan University, Guangzhou, PR China
| | - Qichun Cai
- Cancer Center, Clifford Hospital, Jinan University, Guangzhou, PR China
| | - Clifford L K Pang
- Cancer Center, Clifford Hospital, Jinan University, Guangzhou, PR China
| |
Collapse
|
16
|
Bailon-Moscoso N, Coronel-Hidalgo J, Duarte-Casar R, Guamán-Ortiz LM, Figueroa JG, Romero-Benavides JC. Exploring the Antioxidant Potential of Tragia volubilis L.: Mitigating Chemotherapeutic Effects of Doxorubicin on Tumor Cells. Antioxidants (Basel) 2023; 12:2003. [PMID: 38001856 PMCID: PMC10669231 DOI: 10.3390/antiox12112003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Several plants of the genus Tragia L. have shown antibacterial, fungicidal, and antiproliferative activity, among other types of activities; however, most species of the genus have not been investigated. Tragia volubilis L. is native to tropical America and Africa, and although it has been reported as medicinal in the literature, it has not been thoroughly investigated. In this study, the phytochemical screening, isolation, and identification of compounds and the determination of the antioxidant activity of the aqueous extract of Tragia volubilis L. and its partitions were carried out. Ethyl acetate and n-butanol partitions of the extract present high antioxidant activity according to the Antioxidant Activity Index. Due to their activity, these partitions were tested on RKO cells as a representative model, both individually and in combination with Doxorubicin. It was found that the partitions significantly reduced the effect of Doxorubicin, as well as the expression of proteins involved in DNA damage and cell death. While the reduction of the chemotherapeutic effect of Doxorubicin on tumor cells may not be a desired outcome in therapeutic settings, the findings of the study are valuable in revealing the antioxidant potential of Tragia volubilis L. and its partitions. This highlights the importance of carefully regulating the application of antioxidants, especially in the context of cancer chemotherapy.
Collapse
Affiliation(s)
- Natalia Bailon-Moscoso
- Departamento de Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (J.C.-H.); (L.M.G.-O.)
| | - José Coronel-Hidalgo
- Departamento de Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (J.C.-H.); (L.M.G.-O.)
- Carrera de Bioquímica y Farmacia, Facultad de Ciencias Exactas y Naturales, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador
| | - Rodrigo Duarte-Casar
- Maestría en Química Aplicada, Facultad de Ciencias Exactas y Naturales, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador;
- Tecnología Superior en Gestión Culinaria, Pontificia Universidad Católica del Ecuador—Sede Manabí, Portoviejo 130103, Ecuador
| | - Luis Miguel Guamán-Ortiz
- Departamento de Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (J.C.-H.); (L.M.G.-O.)
| | - Jorge G. Figueroa
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (J.G.F.); (J.C.R.-B.)
| | - Juan Carlos Romero-Benavides
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (J.G.F.); (J.C.R.-B.)
| |
Collapse
|
17
|
Sanookpan K, Chantaravisoot N, Kalpongnukul N, Chuenjit C, Wattanathamsan O, Shoaib S, Chanvorachote P, Buranasudja V. Pharmacological Ascorbate Elicits Anti-Cancer Activities against Non-Small Cell Lung Cancer through Hydrogen-Peroxide-Induced-DNA-Damage. Antioxidants (Basel) 2023; 12:1775. [PMID: 37760080 PMCID: PMC10525775 DOI: 10.3390/antiox12091775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) poses a significant global health burden with unsatisfactory survival rates, despite advancements in diagnostic and therapeutic modalities. Novel therapeutic approaches are urgently required to improve patient outcomes. Pharmacological ascorbate (P-AscH-; ascorbate at millimolar concentration in plasma) emerged as a potential candidate for cancer therapy for recent decades. In this present study, we explore the anti-cancer effects of P-AscH- on NSCLC and elucidate its underlying mechanisms. P-AscH- treatment induces formation of cellular oxidative distress; disrupts cellular bioenergetics; and leads to induction of apoptotic cell death and ultimately reduction in clonogenic survival. Remarkably, DNA and DNA damage response machineries are identified as vulnerable targets for P-AscH- in NSCLC therapy. Treatments with P-AscH- increase the formation of DNA damage and replication stress markers while inducing mislocalization of DNA repair machineries. The cytotoxic and genotoxic effects of P-AscH- on NSCLC were reversed by co-treatment with catalase, highlighting the roles of extracellular hydrogen peroxide in anti-cancer activities of P-AscH-. The data from this current research advance our understanding of P-AscH- in cancer treatment and support its potential clinical use as a therapeutic option for NSCLC therapy.
Collapse
Affiliation(s)
- Kittipong Sanookpan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (K.S.); (O.W.); (S.S.); (P.C.)
- Nabsolute Co., Ltd., Bangkok 10330, Thailand
| | - Naphat Chantaravisoot
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.C.); (C.C.)
- Center of Excellence in Systems Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Nuttiya Kalpongnukul
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chatchapon Chuenjit
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.C.); (C.C.)
| | - Onsurang Wattanathamsan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (K.S.); (O.W.); (S.S.); (P.C.)
| | - Sara Shoaib
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (K.S.); (O.W.); (S.S.); (P.C.)
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (K.S.); (O.W.); (S.S.); (P.C.)
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Visarut Buranasudja
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (K.S.); (O.W.); (S.S.); (P.C.)
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
18
|
Jiang H, Zuo J, Li B, Chen R, Luo K, Xiang X, Lu S, Huang C, Liu L, Tang J, Gao F. Drug-induced oxidative stress in cancer treatments: Angel or devil? Redox Biol 2023; 63:102754. [PMID: 37224697 DOI: 10.1016/j.redox.2023.102754] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023] Open
Abstract
Oxidative stress (OS), defined as redox imbalance in favor of oxidant burden, is one of the most significant biological events in cancer progression. Cancer cells generally represent a higher oxidant level, which suggests a dual therapeutic strategy by regulating redox status (i.e., pro-oxidant therapy and/or antioxidant therapy). Indeed, pro-oxidant therapy exhibits a great anti-cancer capability, attributing to a higher oxidant accumulation within cancer cells, whereas antioxidant therapy to restore redox homeostasis has been claimed to fail in several clinical practices. Targeting the redox vulnerability of cancer cells by pro-oxidants capable of generating excessive reactive oxygen species (ROS) has surfaced as an important anti-cancer strategy. However, multiple adverse effects caused by the indiscriminate attacks of uncontrolled drug-induced OS on normal tissues and the drug-tolerant capacity of some certain cancer cells greatly limit their further applications. Herein, we review several representative oxidative anti-cancer drugs and summarize their side effects on normal tissues and organs, emphasizing that seeking a balance between pro-oxidant therapy and oxidative damage is of great value in exploiting next-generation OS-based anti-cancer chemotherapeutics.
Collapse
Affiliation(s)
- Hao Jiang
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Jing Zuo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bowen Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Chen
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Kangjia Luo
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Xionghua Xiang
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Shuaijun Lu
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Liu
- Ningbo Women & Children's Hospital, Ningbo, 315012, China.
| | - Jing Tang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
| | - Feng Gao
- The First Hospital of Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
19
|
Krejbich P, Birringer M. The Self-Administered Use of Complementary and Alternative Medicine (CAM) Supplements and Antioxidants in Cancer Therapy and the Critical Role of Nrf-2-A Systematic Review. Antioxidants (Basel) 2022; 11:2149. [PMID: 36358521 PMCID: PMC9686580 DOI: 10.3390/antiox11112149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 07/30/2023] Open
Abstract
Complementary and alternative medicine (CAM) supplements are widely used by cancer patients. Dietary supplements, vitamins and minerals, herbal remedies, and antioxidants are especially popular. In a systematic literature review, 37 studies, each including more than 1000 participants, on CAM, dietary supplement, and vitamin use among cancer patients were identified. Accordingly, cancer patients use antioxidants such as vitamin C (from 2.6% (United Kingdom) to 41.6% (United States)) and vitamin E (from 2.9% (China) to 48% (United States)). Dietary supplements and vitamins are taken for different reasons, but often during conventional cancer treatment involving chemotherapy or radiotherapy and in a self-decided manner without seeking medical advice from healthcare professionals. Drug-drug interactions with dietary supplements or vitamins involving multiple signaling pathways are well described. Since most of the anticancer drugs generate reactive oxygen species (ROS), an adaptive stress response of healthy and malignant cells, mainly driven by the Nrf-2-Keap I network, can be observed. On the one hand, healthy cells should be protected from ROS-overproducing chemotherapy and radiotherapy; on the other hand, ROS production in cancer cells is a "desirable side effect" during anticancer drug treatment. We here describe the paradoxical use of antioxidants and supplements during cancer therapy, possible interactions with anticancer drugs, and the involvement of the Nrf-2 transcription factor.
Collapse
Affiliation(s)
- Paula Krejbich
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Wissenschaftliches Zentrum für Ernährung, Lebensmittel und Nachhaltige Versorgungssysteme (ELVe), Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Public Health Zentrum Fulda, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
| | - Marc Birringer
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Wissenschaftliches Zentrum für Ernährung, Lebensmittel und Nachhaltige Versorgungssysteme (ELVe), Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Public Health Zentrum Fulda, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
| |
Collapse
|
20
|
Zaher A, Stephens LM, Miller AM, Hartwig SM, Stolwijk JM, Petronek MS, Zacharias ZR, Wadas TJ, Monga V, Cullen JJ, Furqan M, Houtman JCD, Varga SM, Spitz DR, Allen BG. Pharmacological ascorbate as a novel therapeutic strategy to enhance cancer immunotherapy. Front Immunol 2022; 13:989000. [PMID: 36072595 PMCID: PMC9444023 DOI: 10.3389/fimmu.2022.989000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Pharmacological ascorbate (i.e., intravenous infusions of vitamin C reaching ~ 20 mM in plasma) is under active investigation as an adjuvant to standard of care anti-cancer treatments due to its dual redox roles as an antioxidant in normal tissues and as a prooxidant in malignant tissues. Immune checkpoint inhibitors (ICIs) are highly promising therapies for many cancer patients but face several challenges including low response rates, primary or acquired resistance, and toxicity. Ascorbate modulates both innate and adaptive immune functions and plays a key role in maintaining the balance between pro and anti-inflammatory states. Furthermore, the success of pharmacological ascorbate as a radiosensitizer and a chemosensitizer in pre-clinical studies and early phase clinical trials suggests that it may also enhance the efficacy and expand the benefits of ICIs.
Collapse
Affiliation(s)
- Amira Zaher
- Cancer Biology Program, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Laura M. Stephens
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Ann M. Miller
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Stacey M. Hartwig
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Jeffrey M. Stolwijk
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Michael S. Petronek
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Zeb R. Zacharias
- Human Immunology Core & Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Thaddeus J. Wadas
- Department of Radiology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Varun Monga
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Joseph J. Cullen
- Department of Surgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Muhammad Furqan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Jon C. D. Houtman
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Steven M. Varga
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Bryan G. Allen,
| |
Collapse
|