1
|
Jiao C, Qiu J, Gong C, Li X, Liang H, He C, Cen S, Xie Y. Ganoderma lucidum extract reverses multidrug resistance in breast cancer cells through inhibiting ATPase activity of the P-glycoprotein via MAPK/ERK signaling pathway. Exp Cell Res 2025; 444:114355. [PMID: 39613022 DOI: 10.1016/j.yexcr.2024.114355] [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: 05/30/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
Breast cancer represents a persistent global health challenge, with multidrug resistance (MDR) posing a significant obstacle to effective treatment. In this study, we investigate the potential of Ganoderma lucidum extract (GLE) in reversing MDR in breast cancer and delve into the underlying mechanisms. We establish a robust in vitro 3D model of breast cancer with acquired MDR induced by paclitaxel. Utilizing the CCK-8 method, we assess the impact of GLE on cytotoxic drug sensitivity to determine its in vitro MDR reversal activity. Our results reveal that GLE enhances the toxicity of paclitaxel in breast cancer cells by inhibiting the ATPase activity of P-glycoprotein (P-gp) and increasing the intracellular and extracellular excretion of P-gp substrates, all without significantly altering P-gp protein expression. Additionally, GLE inhibits the phosphorylation of ERK1/2, suggesting that the enhanced sensitivity of breast cancer cells to paclitaxel by GLE is associated with the MAPK pathway. These findings indicate that GLE may inhibit P-gp-mediated drug efflux via the MAPK pathway, thus effectively overcoming paclitaxel resistance in breast cancer. This study provides valuable insights into the potential clinical applications of GLE in reversing multidrug resistance, offering hope for improved breast cancer treatment strategies.
Collapse
Affiliation(s)
- Chunwei Jiao
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China; Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, 526000, PR China
| | - Jinshou Qiu
- Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, Fujian, 363000, PR China
| | - Congcong Gong
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China; South China University of Technology, PR China
| | - Xiaoyi Li
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China
| | - Huijia Liang
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China
| | - Chunyan He
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, 526000, PR China
| | - Sien Cen
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China
| | - Yizhen Xie
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China; Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, 526000, PR China.
| |
Collapse
|
2
|
Degitz C, Reime S, Baumbach CM, Rauschner M, Thews O. Modulation of mitochondrial function by extracellular acidosis in tumor cells and normal fibroblasts: Role of signaling pathways. Neoplasia 2024; 52:100999. [PMID: 38631214 PMCID: PMC11036092 DOI: 10.1016/j.neo.2024.100999] [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: 01/17/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
In many tumors pronounced extracellular acidosis resulting from glycolytic metabolism is found. Since several environmental stress factors affect the mitochondrial activity the aim of the study was to analyze the impact of acidosis on cellular oxygen consumption and which signaling pathways may be involved in the regulation. In two tumor cell lines and normal fibroblasts cellular oxygen consumption rate (OCR) and mitochondrial function were measured after 3 h at pH 6.6. Besides the activation of ERK1/2, p38 and PI3K signaling in the cytosolic and mitochondrial compartment, the mitochondrial structure and proteins related to mitochondria fission were analyzed. The acidic extracellular environment increased OCR in tumor cells but not in fibroblasts. In parallel, the mitochondrial membrane potential increased at low pH. In both tumor lines (but not in fibroblasts), the phosphorylation of ERK1/2 and PI3K/Akt was significantly increased, and both cascades were involved in OCR modulation. The activation of signaling pathways was located predominantly in the mitochondrial compartment of the cells. At low pH, the mitochondrial structure in tumor cells showed structural changes related to elongation whereas mitochondria fragmentation was reduced indicating mitochondria fusion. However, these morphological changes were not related to ERK1/2 or PI3K signaling. Acidic stress seems to induce an increased oxygen consumption, which might further aggravate tumor hypoxia. Low pH also induces mitochondria fusion that is not mediated by ERK1/2 or PI3K signaling. The mechanism by which these signaling cascades modulate the respiratory activity of tumor cells needs further investigation.
Collapse
Affiliation(s)
- Carmen Degitz
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6 (Saale), Halle, 06112, Germany
| | - Sarah Reime
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6 (Saale), Halle, 06112, Germany
| | - Christina-Marie Baumbach
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6 (Saale), Halle, 06112, Germany
| | - Mandy Rauschner
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6 (Saale), Halle, 06112, Germany
| | - Oliver Thews
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6 (Saale), Halle, 06112, Germany.
| |
Collapse
|
3
|
Li L, Qiu H. Asperulosidic Acid Restrains Hepatocellular Carcinoma Development and Enhances Chemosensitivity Through Inactivating the MEKK1/NF-κB Pathway. Appl Biochem Biotechnol 2024; 196:1-17. [PMID: 37097403 DOI: 10.1007/s12010-023-04500-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
Asperulosidic acid (ASPA) is a plant-extracted iridoid terpenoid with tumor-suppressive and anti-inflammatory properties. At present, the antitumor function of ASPA and its related mechanisms in hepatocellular carcinoma (HCC) cells were explored. Human normal hepatocytes HL-7702 and HCC cells (Huh7 and HCCLM3) were treated with varying concentrations (0 to 200 μg/mL) of ASPA. Cell viability, proliferation, apoptosis, migration, and invasion were checked. The expression of proteins was detected by Western blot. Furthermore, the effect of ASPA (100 μg/mL) on the sensitivity of HCC cells to chemotherapeutic agents, including doxorubicin and cisplatin, was evaluated. A subcutaneous xenografted tumor model was set up in nude mice, and the antitumor effects of ASPA were evaluated. ASPA hindered HCC cells' proliferation, migration, and invasion, and amplified their apoptosis and sensitivity to chemotherapeutic agents. Additionally, ASPA inactivated the MEKK1/NF-κB pathway. Overexpression of MEKK1 increased HCC proliferation, migration, and invasion and facilitated chemoresistance. ASPA treatment alleviated the carcinogenic effect mediated by MEKK1 overexpression. MEKK1 knockdown slowed down HCC progression. However, ASPA could not exert additional antitumor effects in MEKK1 knockdown cells. In vivo results displayed that ASPA substantially curbed tumor growth and inactivated the MEKK1/NF-κB pathway in mice. All over, ASPA exerts antitumor effects in HCC by suppressing the MEKK1/NF-κB pathway.
Collapse
Affiliation(s)
- Liang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, No.1882 South Zhong Huan Road, Jiaxing City, Zhejiang Province, 314001, China
| | - Huiwen Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, No.1882 South Zhong Huan Road, Jiaxing City, Zhejiang Province, 314001, China.
| |
Collapse
|
4
|
Riemann A, Rauschner M, Reime S, Thews O. The Role of microRNAs in Gene Expression and Signaling Response of Tumor Cells to an Acidic Environment. Int J Mol Sci 2023; 24:16919. [PMID: 38069241 PMCID: PMC10707721 DOI: 10.3390/ijms242316919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Many tumors are characterized by marked extracellular acidosis due to increased glycolytic metabolism, which affects gene expression and thereby tumor biological behavior. At the same time, acidosis leads to altered expression of several microRNAs (Mir7, Mir183, Mir203, Mir215). The aim of this study was to analyze whether the acidosis-induced changes in cytokines and tumor-related genes are mediated via pH-sensitive microRNAs. Therefore, the expression of Il6, Nos2, Ccl2, Spp1, Tnf, Acat2, Aox1, Crem, Gls2, Per3, Pink1, Txnip, and Ypel3 was examined in acidosis upon simultaneous transfection with microRNA mimics or antagomirs in two tumor lines in vitro and in vivo. In addition, it was investigated whether microRNA expression in acidosis is affected via known pH-sensitive signaling pathways (MAPK, PKC, PI3K), via ROS, or via altered intracellular Ca2+ concentration. pH-dependent microRNAs were shown to play only a minor role in modulating gene expression. Individual genes (e.g., Ccl2, Txnip, Ypel3) appear to be affected by Mir183, Mir203, or Mir215 in acidosis, but these effects are cell line-specific. When examining whether acid-dependent signaling affects microRNA expression, it was found that Mir203 was modulated by MAPK and ROS, Mir7 was affected by PKC, and Mir215 was dependent on the intracellular Ca2+ concentration. Mir183 could be increased by ROS scavenging. These correlations could possibly result in new therapeutic approaches for acidotic tumors.
Collapse
Affiliation(s)
| | | | | | - Oliver Thews
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, 06108 Halle, Germany
| |
Collapse
|
5
|
Matsui T, Toda Y, Sato H, Itagaki R, Konishi K, Moshnikova A, Andreev OA, Hosogi S, Reshetnyak YK, Ashihara E. Targeting acidic pre-metastatic niche in lungs by pH low insertion peptide and its utility for anti-metastatic therapy. Front Oncol 2023; 13:1258442. [PMID: 38033489 PMCID: PMC10684925 DOI: 10.3389/fonc.2023.1258442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Dysregulated extracellular pH, the universal feature of tumor, works as an evolutional force to drive dissemination of tumor cells. It is well-established that tumor acidity is associated with tumor growth and metastasis. However, the pH of pre-metastatic niche remains unclear. We hypothesized that primary tumor cells remotely prime acidity in secondary organ to achieve metastatic colonization. Herein, we demonstrated that the pH responsive probe pH Low Insertion Peptide (pHLIP) was notably accumulated in pre-metastatic lungs of 4T1.2 breast tumor-bearing mice. The pHLIP-targeted lungs showed high amounts of lactate and overexpressed glycolysis-related proteins. Pharmacological inhibition of glycolysis suppressed the lung acidification induced by 4T1.2 cancer cell culture supernatant and delayed subsequent metastatic burden of disseminated tumor cells. In the acidic lungs, pHLIP was primarily localized in alveolar type 2 cells which strongly expressed glycolysis-related proteins. 4T1.2-derived extracellular vesicles expressed some of the glycolysis-related proteins, and their administration increased pHLIP accumulation and glycolytic enhancement in lungs. pHLIP-conjugated dexamethasone effectively attenuated lung metastatic burden by disrupting pro-inflammatory response in the acidic lungs. From these results, targeting the metastasis-supporting microenvironment by pHLIP technology creates possibility to identify pre-metastatic organ and prevent metastatic recurrence.
Collapse
Affiliation(s)
- Toma Matsui
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yuki Toda
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Haruka Sato
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Rina Itagaki
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kazuya Konishi
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Anna Moshnikova
- Physics Department, University of Rhode Island, Kingston, RI, United States
| | - Oleg A. Andreev
- Physics Department, University of Rhode Island, Kingston, RI, United States
| | - Shigekuni Hosogi
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yana K. Reshetnyak
- Physics Department, University of Rhode Island, Kingston, RI, United States
| | - Eishi Ashihara
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
6
|
Pujals M, Mayans C, Bellio C, Méndez O, Greco E, Fasani R, Alemany-Chavarria M, Zamora E, Padilla L, Mitjans F, Nuciforo P, Canals F, Nonell L, Abad M, Saura C, Tabernero J, Villanueva J. RAGE/SNAIL1 signaling drives epithelial-mesenchymal plasticity in metastatic triple-negative breast cancer. Oncogene 2023; 42:2610-2628. [PMID: 37468678 DOI: 10.1038/s41388-023-02778-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Epithelial/Mesenchymal (E/M) plasticity plays a fundamental role both in embryogenesis and during tumorigenesis. The receptor for advanced glycation end products (RAGE) is a driver of cell plasticity in fibrotic diseases; however, its role and molecular mechanism in triple-negative breast cancer (TNBC) remains unclear. Here, we demonstrate that RAGE signaling maintains the mesenchymal phenotype of aggressive TNBC cells by enforcing the expression of SNAIL1. Besides, we uncover a crosstalk mechanism between the TGF-β and RAGE pathways that is required for the acquisition of mesenchymal traits in TNBC cells. Consistently, RAGE inhibition elicits epithelial features that block migration and invasion capacities. Next, since RAGE is a sensor of the tumor microenvironment, we modeled acute acidosis in TNBC cells and showed it promotes enhanced production of RAGE ligands and the activation of RAGE-dependent invasive properties. Furthermore, acute acidosis increases SNAIL1 levels and tumor cell invasion in a RAGE-dependent manner. Finally, we demonstrate that in vivo inhibition of RAGE reduces metastasis incidence and expands survival, consistent with molecular effects that support the relevance of RAGE signaling in E/M plasticity. These results uncover new molecular insights on the regulation of E/M phenotypes in cancer metastasis and provide rationale for pharmacological intervention of this signaling axis.
Collapse
Affiliation(s)
- Mireia Pujals
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Carla Mayans
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Chiara Bellio
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Olga Méndez
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Emanuela Greco
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Roberta Fasani
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Mercè Alemany-Chavarria
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Esther Zamora
- Medical Oncology Service, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Laura Padilla
- LEITAT Technological Center, 08028, Barcelona, Spain
| | | | - Paolo Nuciforo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Francesc Canals
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Lara Nonell
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - María Abad
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Altos Labs Cambridge Institute of Science, Cambridge, UK
| | - Cristina Saura
- Medical Oncology Service, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Josep Tabernero
- Medical Oncology Service, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- IOB Institute of Oncology, Quiron Group (Quiron-IOB), Barcelona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Villanueva
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
7
|
Bou Antoun N, Chioni AM. Dysregulated Signalling Pathways Driving Anticancer Drug Resistance. Int J Mol Sci 2023; 24:12222. [PMID: 37569598 PMCID: PMC10418675 DOI: 10.3390/ijms241512222] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
One of the leading causes of death worldwide, in both men and women, is cancer. Despite the significant development in therapeutic strategies, the inevitable emergence of drug resistance limits the success and impedes the curative outcome. Intrinsic and acquired resistance are common mechanisms responsible for cancer relapse. Several factors crucially regulate tumourigenesis and resistance, including physical barriers, tumour microenvironment (TME), heterogeneity, genetic and epigenetic alterations, the immune system, tumour burden, growth kinetics and undruggable targets. Moreover, transforming growth factor-beta (TGF-β), Notch, epidermal growth factor receptor (EGFR), integrin-extracellular matrix (ECM), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphoinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR), wingless-related integration site (Wnt/β-catenin), Janus kinase/signal transducers and activators of transcription (JAK/STAT) and RAS/RAF/mitogen-activated protein kinase (MAPK) signalling pathways are some of the key players that have a pivotal role in drug resistance mechanisms. To guide future cancer treatments and improve results, a deeper comprehension of drug resistance pathways is necessary. This review covers both intrinsic and acquired resistance and gives a comprehensive overview of recent research on mechanisms that enable cancer cells to bypass barriers put up by treatments, and, like "satellite navigation", find alternative routes by which to carry on their "journey" to cancer progression.
Collapse
Affiliation(s)
| | - Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Biomolecular Sciences Department, Kingston University London, Kingston-upon-Thames KT1 2EE, UK;
| |
Collapse
|
8
|
Xu Q, Lan X, Lin H, Xi Q, Wang M, Quan X, Yao G, Yu Z, Wang Y, Yu M. Tumor microenvironment-regulating nanomedicine design to fight multi-drug resistant tumors. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1842. [PMID: 35989568 DOI: 10.1002/wnan.1842] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 01/31/2023]
Abstract
The tumor microenvironment (TME) is a very cunning system that enables tumor cells to escape death post-traditional antitumor treatments through the comprehensive effect of different factors, thereby leading to drug resistance. Deep insights into TME characteristics and tumor resistance encourage the construction of nanomedicines that can remodel the TME against drug resistance. Tremendous interest in combining TME-regulation measurement with traditional tumor treatment to fight multidrug-resistant tumors has been inspired by the increasing understanding of the role of TME reconstruction in improving the antitumor efficiency of drug-resistant tumor therapy. This review focuses on the underlying relationships between specific TME characteristics (such as hypoxia, acidity, immunity, microorganisms, and metabolism) and drug resistance in tumor treatments. The exciting antitumor activities strengthened by TME regulation are also discussed in-depth, providing solutions from the perspective of nanomedicine design. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Qinqin Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Xinyue Lan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China.,Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Huimin Lin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Qiye Xi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Manchun Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaolong Quan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Guangyu Yao
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhiqiang Yu
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, People's Republic of China
| | - Yongxia Wang
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, People's Republic of China
| | - Meng Yu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
9
|
Dhawan A, Pifer PM, Sandulache VC, Skinner HD. Metabolic targeting, immunotherapy and radiation in locally advanced non-small cell lung cancer: Where do we go from here? Front Oncol 2022; 12:1016217. [PMID: 36591457 PMCID: PMC9794617 DOI: 10.3389/fonc.2022.1016217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
In the US, there are ~250,000 new lung cancer diagnoses and ~130,000 deaths per year, and worldwide there are an estimated 1.6 million deaths per year from this deadly disease. Lung cancer is the most common cause of cancer death worldwide, and it accounts for roughly a quarter of all cancer deaths in the US. Non-small cell lung cancer (NSCLC) represents 80-85% of these cases. Due to an enormous tobacco cessation effort, NSCLC rates in the US are decreasing, and the implementation of lung cancer screening guidelines and other programs have resulted in a higher percentage of patients presenting with potentially curable locoregional disease, instead of distant disease. Exciting developments in molecular targeted therapy and immunotherapy have resulted in dramatic improvement in patients' survival, in combination with new surgical, pathological, radiographical, and radiation techniques. Concurrent platinum-based doublet chemoradiation therapy followed by immunotherapy has set the benchmark for survival in these patients. However, despite these advances, ~50% of patients diagnosed with locally advanced NSCLC (LA-NSCLC) survive long-term. In patients with local and/or locoregional disease, chemoradiation is a critical component of curative therapy. However, there remains a significant clinical gap in improving the efficacy of this combined therapy, and the development of non-overlapping treatment approaches to improve treatment outcomes is needed. One potential promising avenue of research is targeting cancer metabolism. In this review, we will initially provide a brief general overview of tumor metabolism as it relates to therapeutic targeting. We will then focus on the intersection of metabolism on both oxidative stress and anti-tumor immunity. This will be followed by discussion of both tumor- and patient-specific opportunities for metabolic targeting in NSCLC. We will then conclude with a discussion of additional agents currently in development that may be advantageous to combine with chemo-immuno-radiation in NSCLC.
Collapse
Affiliation(s)
- Annika Dhawan
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States
| | - Phillip M. Pifer
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States
| | - Vlad C. Sandulache
- Bobby R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Heath D. Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States,*Correspondence: Heath D. Skinner,
| |
Collapse
|
10
|
Bogdanov A, Bogdanov A, Chubenko V, Volkov N, Moiseenko F, Moiseyenko V. Tumor acidity: From hallmark of cancer to target of treatment. Front Oncol 2022; 12:979154. [PMID: 36106097 PMCID: PMC9467452 DOI: 10.3389/fonc.2022.979154] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
Abstract
Tumor acidity is one of the cancer hallmarks and is associated with metabolic reprogramming and the use of glycolysis, which results in a high intracellular lactic acid concentration. Cancer cells avoid acid stress major by the activation and expression of proton and lactate transporters and exchangers and have an inverted pH gradient (extracellular and intracellular pHs are acid and alkaline, respectively). The shift in the tumor acid-base balance promotes proliferation, apoptosis avoidance, invasiveness, metastatic potential, aggressiveness, immune evasion, and treatment resistance. For example, weak-base chemotherapeutic agents may have a substantially reduced cellular uptake capacity due to "ion trapping". Lactic acid negatively affects the functions of activated effector T cells, stimulates regulatory T cells, and promotes them to express programmed cell death receptor 1. On the other hand, the inversion of pH gradient could be a cancer weakness that will allow the development of new promising therapies, such as tumor-targeted pH-sensitive antibodies and pH-responsible nanoparticle conjugates with anticancer drugs. The regulation of tumor pH levels by pharmacological inhibition of pH-responsible proteins (monocarboxylate transporters, H+-ATPase, etc.) and lactate dehydrogenase A is also a promising anticancer strategy. Another idea is the oral or parenteral use of buffer systems, such as sodium bicarbonate, to neutralize tumor acidity. Buffering therapy does not counteract standard treatment methods and can be used in combination to increase effectiveness. However, the mechanisms of the anticancer effect of buffering therapy are still unclear, and more research is needed. We have attempted to summarize the basic knowledge about tumor acidity.
Collapse
Affiliation(s)
- Alexey Bogdanov
- Saint Petersburg Clinical Research and Practical Center of Specialized Types of Medical Care (Oncological), Saint Petersburg, Russia
| | | | | | | | | | | |
Collapse
|
11
|
The role of tumor acidification in aggressiveness, cell dissemination and treatment resistance of oral squamous cell carcinoma. Life Sci 2022; 288:120163. [PMID: 34822797 DOI: 10.1016/j.lfs.2021.120163] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 12/30/2022]
Abstract
AIMS To investigate the role of tumor acidification in cell behavior, migration, and treatment resistance of oral squamous cell carcinoma (OSCC). MAIN METHODS The SCC4 and SCC25 cell lines were exposed to acidified (pH 6.8) cell culture medium for 7 days. Alternatively, a long-term acidosis was induced for 21 days. In addition, to mimic dynamic pH fluctuation of the tumor microenvironment, cells were reconditioned to neutral pH after experimental acidosis. This study assessed cell proliferation and viability by sulforhodamine B and flow cytometry. Individual and collective cell migration was analyzed by wound healing, time lapse, and transwell assays. Modifications of cell phenotype, EMT induction and stemness potential were investigated by qRT-PCR, western blot, and immunofluorescence. Finally, resistance to chemo- and radiotherapy of OSCC when exposed to acidified environmental conditions (pH 6.8) was determined. KEY FINDINGS The exposure to an acidic microenvironment caused an initial reduction of OSCC cells viability, followed by an adaptation process. Acidic adapted cells acquired a mesenchymal-like phenotype along with increased migration and motility indexes. Moreover, tumoral extracellular acidity was capable to induce cellular stemness and to increase chemo- and radioresistance of oral cancer cells. SIGNIFICANCE In summary, the results showed that the acidic microenvironment leads to a more aggressive and treatment resistant OSCC cell population.
Collapse
|
12
|
Gonçalves AC, Richiardone E, Jorge J, Polónia B, Xavier CPR, Salaroglio IC, Riganti C, Vasconcelos MH, Corbet C, Sarmento-Ribeiro AB. Impact of cancer metabolism on therapy resistance - Clinical implications. Drug Resist Updat 2021; 59:100797. [PMID: 34955385 DOI: 10.1016/j.drup.2021.100797] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite an increasing arsenal of anticancer therapies, many patients continue to have poor outcomes due to the therapeutic failures and tumor relapses. Indeed, the clinical efficacy of anticancer therapies is markedly limited by intrinsic and/or acquired resistance mechanisms that can occur in any tumor type and with any treatment. Thus, there is an urgent clinical need to implement fundamental changes in the tumor treatment paradigm by the development of new experimental strategies that can help to predict the occurrence of clinical drug resistance and to identify alternative therapeutic options. Apart from mutation-driven resistance mechanisms, tumor microenvironment (TME) conditions generate an intratumoral phenotypic heterogeneity that supports disease progression and dismal outcomes. Tumor cell metabolism is a prototypical example of dynamic, heterogeneous, and adaptive phenotypic trait, resulting from the combination of intrinsic [(epi)genetic changes, tissue of origin and differentiation dependency] and extrinsic (oxygen and nutrient availability, metabolic interactions within the TME) factors, enabling cancer cells to survive, metastasize and develop resistance to anticancer therapies. In this review, we summarize the current knowledge regarding metabolism-based mechanisms conferring adaptive resistance to chemo-, radio-and immunotherapies as well as targeted therapies. Furthermore, we report the role of TME-mediated intratumoral metabolic heterogeneity in therapy resistance and how adaptations in amino acid, glucose, and lipid metabolism support the growth of therapy-resistant cancers and/or cellular subpopulations. We also report the intricate interplay between tumor signaling and metabolic pathways in cancer cells and discuss how manipulating key metabolic enzymes and/or providing dietary changes may help to eradicate relapse-sustaining cancer cells. Finally, in the current era of personalized medicine, we describe the strategies that may be applied to implement metabolic profiling for tumor imaging, biomarker identification, selection of tailored treatments and monitoring therapy response during the clinical management of cancer patients.
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| | - Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Bárbara Polónia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | | | - Chiara Riganti
- Department of Oncology, School of Medicine, University of Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium.
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| |
Collapse
|
13
|
Auzmendi J, Akyuz E, Lazarowski A. The role of P-glycoprotein (P-gp) and inwardly rectifying potassium (Kir) channels in sudden unexpected death in epilepsy (SUDEP). Epilepsy Behav 2021; 121:106590. [PMID: 31706919 DOI: 10.1016/j.yebeh.2019.106590] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the major cause of death that affects patients with epilepsy. The risk of SUDEP increases according to the frequency and severity of uncontrolled seizures; therefore, SUDEP risk is higher in patients with refractory epilepsy (RE), in whom most antiepileptic drugs (AEDs) are ineffective for both seizure control and SUDEP prevention. Consequently, RE and SUDEP share a multidrug resistance (MDR) phenotype, which is mainly associated with brain overexpression of ABC-transporters such as P-glycoprotein (P-gp). The activity of P-gp can also contribute to membrane depolarization and affect the normal function of neurons and cardiomyocytes. Other molecular regulators of membrane potential are the inwardly rectifying potassium channels (Kir), whose genetic variants have been related to both epilepsy and heart dysfunctions. Although it has been suggested that dysfunctions of the cardiac, respiratory, and brainstem arousal systems are the causes of SUDEP, the molecular basis for explaining its dysfunctions remain unknown. In rats, repetitive seizures or status epilepticus induced high expression of P-gp and loss Kir expression in the brain and heart, and promoted membrane depolarization, malignant bradycardia, and the high rate of mortality. Here we reviewed clinical and experimental evidences suggesting that abnormal expression of depolarizing/repolarizing factors as P-gp and Kir could favor persistent depolarization of membranes without any rapid functional recovery capacity. This condition induced by convulsive stress could be the molecular mechanism leading to acquired severe bradycardia, as an ineffective heart response generating the appropriate scenario for SUDEP development. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina; INFIBIOC, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica (FFyB), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Enes Akyuz
- Yozgat Bozok University, Medical Faculty, Department of Biophysics, Erdoğan Akdağ Yerleşkesi, 66100 Yozgat, Turkey
| | - Alberto Lazarowski
- INFIBIOC, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica (FFyB), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.
| |
Collapse
|
14
|
Yang M, Zhong X, Yuan Y. Does Baking Soda Function as a Magic Bullet for Patients With Cancer? A Mini Review. Integr Cancer Ther 2021; 19:1534735420922579. [PMID: 32448009 PMCID: PMC7249593 DOI: 10.1177/1534735420922579] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Sodium bicarbonate, commonly known as baking soda, is widely used in the clinic
as an antacid for treating gastric hyperacidity, among other conditions. Chao et
al have reported a clinical trial about targeting intratumor lactic
acidosis–transarterial chemoembolization. Based on conventional transarterial
chemoembolization, the authors added a 5% sodium bicarbonate solution to
cytotoxic drugs, resulting in a high local control rate. The explanation for the
antitumor effects of sodium bicarbonate is related to acidosis in the tumor
microenvironment. In this review, we summarize the findings from studies
administering sodium bicarbonate alone or in combination with other anticancer
therapies as cancer treatments, and discuss methods for safe and effective use
of sodium bicarbonate in the clinic.
Collapse
Affiliation(s)
- Mengyuan Yang
- The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xian Zhong
- The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying Yuan
- The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Zou T, Lu W, Mezhuev Y, Lan M, Li L, Liu F, Cai T, Wu X, Cai Y. A review of nanoparticle drug delivery systems responsive to endogenous breast cancer microenvironment. Eur J Pharm Biopharm 2021; 166:30-43. [PMID: 34098073 DOI: 10.1016/j.ejpb.2021.05.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/24/2021] [Accepted: 05/31/2021] [Indexed: 12/26/2022]
Abstract
Breast cancer, as a malignant disease that seriously threatens women's health, urgently needs to be researched to develop effective and safe therapeutic drugs. Nanoparticle drug delivery systems (NDDS), provide a powerful means for drug targeting to the breast cancer, enhancing the bioavailability and reducing the adverse effects of anticancer drug. However, the breast cancer microenvironment together with heterogeneity of cancer, impedes the tumor targeting effect of NDDS. Breast cancer microenvironment, exerts endogenous stimuli, such as hypoxia, acidosis, and aberrant protease expression, shape a natural shelter for tumor growth, invasion and migration. On the basis of the ubiquitous of endogenous stimuli in the breast cancer microenvironment, researchers exploited them to design the stimuli-responsive NDDS, which response to endogenous stimulus, targeted release drug in breast cancer microenvironment. In this review, we highlighted the effect of the breast cancer microenvironment, summarized innovative NDDS responsive to the internal stimuli in the tumor microenvironment, including the material, the targeting groups, the loading drugs, targeting position and the function of stimuli-responsive nanoparticle drug delivery system. The limitations and potential applications of the stimuli-responsive nanoparticle drug delivery systems for breast cancer treatment were discussed to further the application.
Collapse
Affiliation(s)
- Tengteng Zou
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Wenping Lu
- Guang an'men Hospital China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yaroslav Mezhuev
- Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russia
| | - Meng Lan
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Lihong Li
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Fengjie Liu
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Tiange Cai
- College of Life Sciences, Liaoning University, Shenyang 110036, PR China.
| | - Xiaoyu Wu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada.
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China; Guangdong Key Lab of Traditional Chinese Medicine Information Technology, Jinan University, Guangzhou 510632, PR China; Cancer Research Institute, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
16
|
Bornes L, Belthier G, van Rheenen J. Epithelial-to-Mesenchymal Transition in the Light of Plasticity and Hybrid E/M States. J Clin Med 2021; 10:jcm10112403. [PMID: 34072345 PMCID: PMC8197992 DOI: 10.3390/jcm10112403] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a cellular program which leads to cells losing epithelial features, including cell polarity, cell-cell adhesion and attachment to the basement membrane, while gaining mesenchymal characteristics, such as invasive properties and stemness. This program is involved in embryogenesis, wound healing and cancer progression. Over the years, the role of EMT in cancer progression has been heavily debated, and the requirement of this process in metastasis even has been disputed. In this review, we discuss previous discrepancies in the light of recent findings on EMT, plasticity and hybrid E/M states. Moreover, we highlight various tumor microenvironmental cues and cell intrinsic signaling pathways that induce and sustain EMT programs, plasticity and hybrid E/M states. Lastly, we discuss how recent findings on plasticity, especially on those that enable cells to switch between hybrid E/M states, have changed our understanding on the role of EMT in cancer metastasis, stemness and therapy resistance.
Collapse
|
17
|
Lam SF, Bishop KW, Mintz R, Fang L, Achilefu S. Calcium carbonate nanoparticles stimulate cancer cell reprogramming to suppress tumor growth and invasion in an organ-on-a-chip system. Sci Rep 2021; 11:9246. [PMID: 33927272 PMCID: PMC8084943 DOI: 10.1038/s41598-021-88687-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/15/2021] [Indexed: 02/08/2023] Open
Abstract
The acidic microenvironment of solid tumors induces the propagation of highly invasive and metastatic phenotypes. However, simulating these conditions in animal models present challenges that confound the effects of pH modulators on tumor progression. To recapitulate the tumor microenvironment and isolate the effect of pH on tumor viability, we developed a bifurcated microfluidic device that supports two different cell environments for direct comparison. RFP-expressing breast cancer cells (MDA-MB-231) were cultured in treatment and control chambers surrounded by fibrin, which received acid-neutralizing CaCO3 nanoparticles (nanoCaCO3) and cell culture media, respectively. Data analysis revealed that nanoCaCO3 buffered the pH within the normal physiological range and inhibited tumor cell proliferation compared to the untreated control (p < 0.05). Co-incubation of cancer cells and fibroblasts, followed by nanoCaCO3 treatment showed that the nanoparticles selectively inhibited the growth of the MDA-MB-231 cells and reduced cellular migration of these cells with no impact on the fibroblasts. Sustainable decrease in the intracellular pH of cancer cells treated with nanoCaCO3 indicates that the extracellular pH induced cellular metabolic reprogramming. These results suggest that the nanoCaCO3 can restrict the aggressiveness of tumor cells without affecting the growth and behavior of the surrounding stromal cells.
Collapse
Affiliation(s)
- Sandra F Lam
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin W Bishop
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel Mintz
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lei Fang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Wu P, Gao W, Su M, Nice EC, Zhang W, Lin J, Xie N. Adaptive Mechanisms of Tumor Therapy Resistance Driven by Tumor Microenvironment. Front Cell Dev Biol 2021; 9:641469. [PMID: 33732706 PMCID: PMC7957022 DOI: 10.3389/fcell.2021.641469] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/05/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer is a disease which frequently has a poor prognosis. Although multiple therapeutic strategies have been developed for various cancers, including chemotherapy, radiotherapy, and immunotherapy, resistance to these treatments frequently impedes the clinical outcomes. Besides the active resistance driven by genetic and epigenetic alterations in tumor cells, the tumor microenvironment (TME) has also been reported to be a crucial regulator in tumorigenesis, progression, and resistance. Here, we propose that the adaptive mechanisms of tumor resistance are closely connected with the TME rather than depending on non-cell-autonomous changes in response to clinical treatment. Although the comprehensive understanding of adaptive mechanisms driven by the TME need further investigation to fully elucidate the mechanisms of tumor therapeutic resistance, many clinical treatments targeting the TME have been successful. In this review, we report on recent advances concerning the molecular events and important factors involved in the TME, particularly focusing on the contributions of the TME to adaptive resistance, and provide insights into potential therapeutic methods or translational medicine targeting the TME to overcome resistance to therapy in clinical treatment.
Collapse
Affiliation(s)
- Peijie Wu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wei Gao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Miao Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Wenhui Zhang
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jie Lin
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
19
|
Rauschner M, Lange L, Hüsing T, Reime S, Nolze A, Maschek M, Thews O, Riemann A. Impact of the acidic environment on gene expression and functional parameters of tumors in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:10. [PMID: 33407762 PMCID: PMC7786478 DOI: 10.1186/s13046-020-01815-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023]
Abstract
Background The low extracellular pH (pHe) of tumors resulting from glycolytic metabolism is a stress factor for the cells independent from concomitant hypoxia. The aim of the study was to analyze the impact of acidic pHe on gene expression on mRNA and protein level in two experimental tumor lines in vitro and in vivo and were compared to hypoxic conditions as well as combined acidosis+hypoxia. Methods Gene expression was analyzed in AT1 prostate and Walker-256 mammary carcinoma of the rat by Next Generation Sequencing (NGS), qPCR and Western blot. In addition, the impact of acidosis on tumor cell migration, adhesion, proliferation, cell death and mitochondrial activity was analyzed. Results NGS analyses revealed that 147 genes were uniformly regulated in both cell lines (in vitro) and 79 genes in both experimental tumors after 24 h at low pH. A subset of 25 genes was re-evaluated by qPCR and Western blot. Low pH consistently upregulated Aox1, Gls2, Gstp1, Ikbke, Per3, Pink1, Tlr5, Txnip, Ypel3 or downregulated Acat2, Brip1, Clspn, Dnajc25, Ercc6l, Mmd, Rif1, Zmpste24 whereas hypoxia alone led to a downregulation of most of the genes. Direct incubation at low pH reduced tumor cell adhesion whereas acidic pre-incubation increased the adhesive potential. In both tumor lines acidosis induced a G1-arrest (in vivo) of the cell cycle and a strong increase in necrotic cell death (but not in apoptosis). The mitochondrial O2 consumption increased gradually with decreasing pH. Conclusions These data show that acidic pHe in tumors plays an important role for gene expression independently from hypoxia. In parallel, acidosis modulates functional properties of tumors relevant for their malignant potential and which might be the result of pH-dependent gene expression.
Collapse
Affiliation(s)
- Mandy Rauschner
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany
| | - Luisa Lange
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany
| | - Thea Hüsing
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany
| | - Sarah Reime
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany
| | - Alexander Nolze
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany
| | - Marcel Maschek
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany
| | - Oliver Thews
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany
| | - Anne Riemann
- Institute of Physiology, University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle (Saale), Germany.
| |
Collapse
|
20
|
Vaidya FU, Sufiyan Chhipa A, Mishra V, Gupta VK, Rawat SG, Kumar A, Pathak C. Molecular and cellular paradigms of multidrug resistance in cancer. Cancer Rep (Hoboken) 2020; 5:e1291. [PMID: 33052041 PMCID: PMC9780431 DOI: 10.1002/cnr2.1291] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The acquisition of resistance to chemotherapy is a major hurdle in the successful application of cancer therapy. Several anticancer approaches, including chemotherapies, radiotherapy, surgery and targeted therapies are being employed for the treatment of cancer. However, cancer cells reprogram themselves in multiple ways to evade the effect of these therapies, and over a period of time, the drug becomes inactive due to the development of multi-drug resistance (MDR). MDR is a complex phenomenon where malignant cells become insensitive to anticancer drugs and attain the ability to survive even after several exposures of anticancer drugs. In this review, we have discussed the molecular and cellular paradigms of multidrug resistance in cancer. RECENT FINDINGS An Extensive research in cancer biology revealed that drug resistance in cancer is the result of perpetuated intracellular and extracellular mechanisms such as drug efflux, drug inactivation, drug target alteration, oncogenic mutations, altered DNA damage repair mechanism, inhibition of programmed cell death signaling, metabolic reprogramming, epithelial mesenchymal transition (EMT), inherent cell heterogeneity, epigenetic changes, redox imbalance, or any combination of these mechanisms. An inevitable cross-link between inflammation and drug resistance has been discussed. This review provided insight molecular mechanism to understand the vulnerabilities of cancer cells to develop drug resistance. CONCLUSION MDR is an outcome of interplays between multiple intricate pathways responsible for the inactivation of drug and development of resistance. MDR is a major obstacle in regimens of successful application of anti-cancer therapy. An improved understanding of the molecular mechanism of multi drug resistance and cellular reprogramming can provide a promising opportunity to combat drug resistance in cancer and intensify anti-cancer therapy for the upcoming future.
Collapse
Affiliation(s)
- Foram U. Vaidya
- Cell Biology Laboratory, School of Biological Sciences & BiotechnologyIndian Institute of Advanced ResearchGandhinagarIndia
| | - Abu Sufiyan Chhipa
- Cell Biology Laboratory, School of Biological Sciences & BiotechnologyIndian Institute of Advanced ResearchGandhinagarIndia
| | - Vinita Mishra
- Cell Biology Laboratory, School of Biological Sciences & BiotechnologyIndian Institute of Advanced ResearchGandhinagarIndia
| | | | | | - Ajay Kumar
- Department of ZoologyBanaras Hindu UniversityVaranasiIndia
| | - Chandramani Pathak
- Cell Biology Laboratory, School of Biological Sciences & BiotechnologyIndian Institute of Advanced ResearchGandhinagarIndia
| |
Collapse
|
21
|
Jin MZ, Jin WL. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther 2020; 5:166. [PMID: 32843638 PMCID: PMC7447642 DOI: 10.1038/s41392-020-00280-x] [Citation(s) in RCA: 745] [Impact Index Per Article: 149.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence shows that cellular and acellular components in tumor microenvironment (TME) can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Cancer research and treatment have switched from a cancer-centric model to a TME-centric one, considering the increasing significance of TME in cancer biology. Nonetheless, the clinical efficacy of therapeutic strategies targeting TME, especially the specific cells or pathways of TME, remains unsatisfactory. Classifying the chemopathological characteristics of TME and crosstalk among one another can greatly benefit further studies exploring effective treating methods. Herein, we present an updated image of TME with emphasis on hypoxic niche, immune microenvironment, metabolism microenvironment, acidic niche, innervated niche, and mechanical microenvironment. We then summarize conventional drugs including aspirin, celecoxib, β-adrenergic antagonist, metformin, and statin in new antitumor application. These drugs are considered as viable candidates for combination therapy due to their antitumor activity and extensive use in clinical practice. We also provide our outlook on directions and potential applications of TME theory. This review depicts a comprehensive and vivid landscape of TME from biology to treatment.
Collapse
Affiliation(s)
- Ming-Zhu Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.,Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.
| |
Collapse
|
22
|
Hijazi K, Iannelli F, Cuppone AM, Desjardins D, Caldwell A, Dereuddre-Bosquet N, Scala C, Smith KA, Mukhopadya I, Frank B, Gwozdz G, Santoro F, Grand RL, Pozzi G, Kelly C. In Vivo Modulation of Cervicovaginal Drug Transporters and Tissue Distribution by Film-Released Tenofovir and Darunavir for Topical Prevention of HIV-1. Mol Pharm 2020; 17:852-864. [PMID: 32017579 DOI: 10.1021/acs.molpharmaceut.9b01121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Clinical trials have demonstrated partial protection against HIV-1 infection by vaginal microbicide formulations based on antiretroviral (ARV) drugs. Improved formulations that will maintain sustained drug concentrations at viral target sites in the cervicovaginal mucosa are needed. We have previously demonstrated that treatment of cervicovaginal cell lines with ARV drugs can alter gene expression of drug transporters, suggesting that the mucosal disposition of ARV drugs delivered vaginally can be modulated by drug transporters. This study aimed to investigate in vivo modulation of drug transporter expression in a nonhuman primate model by tenofovir and darunavir released from film formulations. Cervicovaginal tissues were collected from drug-naïve macaques and from macaques vaginally treated with film formulations of tenofovir or darunavir. Drug release in vaginal fluid as well as drug absorption in cervicovaginal tissues and lymph nodes were verified by mass spectrometry. The effects of exposure to drugs on the expression of transporters relevant to ARV drugs were evaluated by quantitative PCR. We showed expression in cervicovaginal tissue of drug-naïve macaques of transporters important for distribution of ARV drugs, albeit at lower levels compared to human tissue for key transporters including P-glycoprotein. Concentrations of tenofovir and darunavir well above the EC50 values determined in vitro were detected in vaginal fluid and vaginal tissues of macaques treated with drug-dissolving films over 24 h and were also comparable to those shown previously to modulate drug transporter expression. Accordingly, Multidrug Resistance associated Protein 2 (MRP2) in cervicovaginal tissue was upregulated by both tenofovir and darunavir. The two drugs also differentially induced and/or inhibited expression of key uptake transporters for reverse transcriptase inhibitors and protease inhibitors. The lower expression of key transporters in macaques may result in increased retention of ARV drugs at the simian cervicovaginal mucosa compared to the human mucosa and has implications for translation of preclinical data. Modulation of drug transporter expression by tenofovir and darunavir points to the potential benefit of MRP2 inhibition to increase ARV drug penetration through the cervicovaginal epithelium.
Collapse
Affiliation(s)
- Karolin Hijazi
- Institute of Dentistry, School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, U.K
| | - Francesco Iannelli
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Anna Maria Cuppone
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Delphine Desjardins
- Université Paris Sud, INSERM U1184-Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, IBFJ, CEA, Fontenay-aux-Roses, France
| | - Anna Caldwell
- Mass Spectrometry Facility, King's College London, London SE1 9NH, U.K
| | - Nathalie Dereuddre-Bosquet
- Université Paris Sud, INSERM U1184-Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, IBFJ, CEA, Fontenay-aux-Roses, France
| | - Carlo Scala
- Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, U.K
| | - Kieron A Smith
- Institute of Dentistry, School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, U.K
| | - Indrani Mukhopadya
- Institute of Dentistry, School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, U.K
| | - Bruce Frank
- Particle Sciences Inc., Lubrizol LifeSciences, Suite 180 Bethlehem, Pennsylvania 18017, United States
| | - Garry Gwozdz
- Particle Sciences Inc., Lubrizol LifeSciences, Suite 180 Bethlehem, Pennsylvania 18017, United States
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Roger Le Grand
- Université Paris Sud, INSERM U1184-Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, IBFJ, CEA, Fontenay-aux-Roses, France
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Charles Kelly
- Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, U.K
| |
Collapse
|
23
|
Radi AM, Mohammed ET, Abushouk AI, Aleya L, Abdel-Daim MM. The effects of abamectin on oxidative stress and gene expression in rat liver and brain tissues: Modulation by sesame oil and ascorbic acid. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 701:134882. [PMID: 31739238 DOI: 10.1016/j.scitotenv.2019.134882] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 10/06/2019] [Indexed: 06/10/2023]
Abstract
The present work was designed to assess the modulatory effects of sesame oil (SO) and ascorbic acid (AA) on abamectin (ABM)-induced oxidative stress and altered gene expression of hepatic cytochrome P450 2E1 (CYP-2E1), p38 MAPK, and caspase-3 and cerebral P-glycoprotein (Abcb1a receptor). Male rats were distributed into five groups (6 rats/group), receiving distilled water, ABM 2 mg/kg bwt 1/5 LD50 orally for 5 days, ABM + AA 100 mg/kg bwt orally, ABM + SO 5 ml/kg bwt orally, or ABM + SO + AA at the aforementioned doses. Nineteen compounds were identified in the SO sample by GC-MS analysis, including tetradecane,2,6,10-trimethyl, octadecane, 1-hexadecanol,2-methyl, and octadecane,6-methyl. Abamectin significantly upregulated the hepatic CYP-2E1 expression with excess generation of oxidative radicals, as evident by the significant depletion of reduced glutathione and elevation of malondialdehyde concentration (p ≤ 0.05) in rat liver and brain tissues. Further, ABM significantly increased TNF-α concentration, the expression of caspase-3 and p38 MAPK in the liver, as well as p-glycoprotein and GABA-A receptor in the brain. These results were in line with the observed histopathological changes. Sesame oil and/or AA supplementation alleviated ABM-induced cell damage by modulating all tested parameters. In conclusion, ABM induces oxidative stress and increases the expression of CYP-2E1, caspase-3, and p38 MAPK in the liver, as well as P-gp and GABA-A receptor in the brain. These effects could be ameliorated by SO and AA, alone and in combination, probably due to their anti-oxidant, anti-apoptotic, and gene-regulating activities.
Collapse
Affiliation(s)
- Abeer M Radi
- Pharmacology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef 62515, Egypt
| | - Eman T Mohammed
- Biochemistry Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef 62515, Egypt
| | - Abdelrahman Ibrahim Abushouk
- Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt; Department of Medicine, Harvard Medical School, Boston, 02215, MA
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030 Besançon Cedex, France.
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
24
|
Williams ED, Gao D, Redfern A, Thompson EW. Controversies around epithelial-mesenchymal plasticity in cancer metastasis. Nat Rev Cancer 2019; 19:716-732. [PMID: 31666716 PMCID: PMC7055151 DOI: 10.1038/s41568-019-0213-x] [Citation(s) in RCA: 287] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2019] [Indexed: 02/07/2023]
Abstract
Experimental evidence accumulated over decades has implicated epithelial-mesenchymal plasticity (EMP), which collectively encompasses epithelial-mesenchymal transition and the reverse process of mesenchymal-epithelial transition, in tumour metastasis, cancer stem cell generation and maintenance, and therapeutic resistance. However, the dynamic nature of EMP processes, the apparent need to reverse mesenchymal changes for the development of macrometastases and the likelihood that only minor cancer cell subpopulations exhibit EMP at any one time have made such evidence difficult to accrue in the clinical setting. In this Perspectives article, we outline the existing preclinical and clinical evidence for EMP and reflect on recent controversies, including the failure of initial lineage-tracing experiments to confirm a major role for EMP in dissemination, and discuss accumulating data suggesting that epithelial features and/or a hybrid epithelial-mesenchymal phenotype are important in metastasis. We also highlight strategies to address the complexities of therapeutically targeting the EMP process that give consideration to its spatially and temporally divergent roles in metastasis, with the view that this will yield a potent and broad class of therapeutic agents.
Collapse
Affiliation(s)
- Elizabeth D Williams
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
- Translational Research Institute (TRI), Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), Brisbane, Queensland, Australia
| | - Dingcheng Gao
- Department of Cardiothoracic Surgery, Department of Cell and Developmental Biology and Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Andrew Redfern
- Department of Medicine, School of Medicine, University of Western Australia, Fiona Stanley Hospital Campus, Perth, Western Australia, Australia
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia.
- Translational Research Institute (TRI), Brisbane, Queensland, Australia.
| |
Collapse
|
25
|
Koyuncu I, Gonel A, Durgun M, Kocyigit A, Yuksekdag O, Supuran CT. Assessment of the antiproliferative and apoptotic roles of sulfonamide carbonic anhydrase IX inhibitors in HeLa cancer cell line. J Enzyme Inhib Med Chem 2019; 34:75-86. [PMID: 30362386 PMCID: PMC6211230 DOI: 10.1080/14756366.2018.1524380] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 12/22/2022] Open
Abstract
Carbonic anhydrase IX (CA IX) has recently been validated as an antitumor/antimetastatic drug target. In this study, we examined the underlying molecular mechanisms and the anticancer activity of sulfonamide CA IX inhibitors against cervical cancer cell lines. The effects of several sulfonamides on HeLa, MDA-MB-231, HT-29 cancer cell lines, and normal cell lines (HEK-293, PNT-1A) viability were determined. The compounds showed high cytotoxic and apoptotic activities, mainly against HeLa cells overexpressing CA IX. We were also examined for intracellular reactive oxygen species (ROS) production; intra-/extracellular pH changes, for inhibition of cell proliferation, cellular mitochondrial membrane potential change and for the detection of caspase 3, 8, 9, and CA IX protein levels. Of the investigated sulfonamides, one compound was found to possess high cytotoxic and anti-proliferative effects in HeLa cells. The cytotoxic effect occurred via apoptosis, being accompanied by a return of pHe/pHi towards normal values as for other CA IX inhibitors investigated earlier.
Collapse
Affiliation(s)
- Ismail Koyuncu
- Department of Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Ataman Gonel
- Department of Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Mustafa Durgun
- Department of Chemistry, Faculty of Arts and Sciences, Harran University, Sanliurfa, Turkey
| | - Abdurrahim Kocyigit
- Department of Medical Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ozgur Yuksekdag
- Department of Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Claudiu T. Supuran
- Neurofarba Dept., Section of Pharmaceutical and Nutriceutical Sciences, Università degli Studi di Firenze, Sesto Fiorentino (Florence), Italy
| |
Collapse
|
26
|
Ye Z, Zeng Z, Shen Y, Yang Q, Chen D, Chen Z, Shen S. ODC1 promotes proliferation and mobility via the AKT/GSK3β/β-catenin pathway and modulation of acidotic microenvironment in human hepatocellular carcinoma. Onco Targets Ther 2019; 12:4081-4092. [PMID: 31239700 PMCID: PMC6553997 DOI: 10.2147/ott.s198341] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/29/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose: Ornithine decarboxylase 1 (ODC1)–an oncogene involved in the biosynthesis of polyamines–is commonly upregulated and associated with poor prognosis in numerous cancers. However, the role and mechanism of ODC1 in hepatocellular carcinoma (HCC) remains unclear. The aim of the present study was to investigate the role of ODC1 in HCC and clarify the latent molecular mechanisms. Material and methods: We used samples obtained from The Cancer Genome Atlas. The expression of ODC1 was also assessed in our additional HCC samples and HCC cell lines. The roles of ODC1 in HCC cell proliferation, migration and invasion in vitro were investigated using the cell-counting kit-8 assay, 5-ethynyl-2´-deoxyuridine assay, colony formation assay, flow cytometry, wound healing assay and transwell assay, respectively. The effect of ODC1 on HCC cell proliferation in vivo was investigated by constructing a xenotransplanted tumor model in nude mice. Quantitative real-time polymerase chain and western blotting were used to detect the expression levels of ODC1 in mimetic hypoxia, nutrient depleted, and acidotic microenvironment. The relationships between ODC1, the AKT/GSK3β/β-catenin pathway, and acidotic microenvironment were further investigated through western blotting, immunohistochemical staining, and immunofluorescence. Results: ODC1 was upregulated in HCC tissues and cell lines, and co-expressed with KI67 and PCNA (P<0.05). A decrease in the expression of ODC1 inhibits proliferation, migration, invasion, and induces cell cycle arrest in HCC cell lines in vitro, while suppressing HCC cell proliferation in vivo (P<0.05). Furthermore, the expression of ODC1 was increased in the mimetic acidotic microenvironment, while the interference with the expression of ODC1 reversed the effect of the acidotic microenvironment through regulation of AKT/GSK3β/β-catenin and related downstream proteins. Conclusion: ODC1 is an unfavorable gene in HCC patients,promoting HCC cell proliferation, migration and invasion via the AKT/GSK3β/β-catenin pathway and modulation of the acidotic microenvironment.
Collapse
Affiliation(s)
- Zi Ye
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Zhirui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guiyang, Guizhou 550009, People's Republic of China.,Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| | - Yiyi Shen
- Department of Liver-Biliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, People's Republic of China
| | - Qiang Yang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Duidui Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Zubing Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Shiqiang Shen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| |
Collapse
|
27
|
Jia Y, Chen L, Chi D, Cong D, Zhou P, Jin J, Ji H, Liang B, Gao S, Hu S. Photodynamic therapy combined with temozolomide inhibits C6 glioma migration and invasion and promotes mitochondrial-associated apoptosis by inhibiting sodium-hydrogen exchanger isoform 1. Photodiagnosis Photodyn Ther 2019; 26:405-412. [PMID: 31085295 DOI: 10.1016/j.pdpdt.2019.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/23/2019] [Accepted: 05/09/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE As a targeted therapeutic technique for glioma inhibition, photodynamic therapy (PDT) has gradually become a focus of basic research related to glioma treatment. The capacity of PDT to kill glioma cells involves varieties of pathways. In glioma cells, activated sodium-hydrogen exchanger isoform 1 (NHE1) can inhibit the cytotoxic effect of temozolomide (TMZ), promote cell migration and invasion, and inhibit cell apoptosis by changing the acid-base equilibrium. The purpose of our study was to explore if PDT combined with TMZ can effectively inhibit glioma cells by influencing NHE1 in vitro. METHODS We analyzed the expression levels of proteins such as NHE1, ezrin, vimentin, Bcl-2, and Bax by Western blot analysis, we assessed the migration and invasion of rat C6 glioma cells by Transwell assay, and we evaluated C6 cell apoptosis in vitro by flow cytometry. RESULTS Western blot results indicated that NHE1, ezrin and vimentin were downregulated after cotreatment of C6 cells, and intracellular acidification was detected by a fluorometric intracellular pH assay. The migration and invasion capacities of C6 cells were significantly hindered after cotreatment, as shown by the Transwell assay. Experimental data also revealed a significant increase in cell apoptosis after cotreatment, as detected by flow cytometry; corresponding proapoptotic changes in Bcl-2, Bax and caspase-3 were also observed in vitro. CONCLUSION These results demonstrate that PDT combined with TMZ can inhibit C6 cell migration and invasion and promote mitochondrial-associated apoptosis by inhibiting NHE1. Therefore, this study provides supporting evidence for a potential method for the treatment of glioma.
Collapse
Affiliation(s)
- Yulong Jia
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Lei Chen
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Dapeng Chi
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Damin Cong
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Peng Zhou
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Jiaqi Jin
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Hang Ji
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Binbin Liang
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Shuai Gao
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China
| | - Shaoshan Hu
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
28
|
Vander Linden C, Corbet C. Therapeutic Targeting of Cancer Stem Cells: Integrating and Exploiting the Acidic Niche. Front Oncol 2019; 9:159. [PMID: 30941310 PMCID: PMC6433943 DOI: 10.3389/fonc.2019.00159] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 02/25/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSC) or tumor-initiating cells represent a small subpopulation of cells within the tumor bulk that share features with somatic stem cells, such as self-renewal and pluripotency. From a clinical point of view, CSC are thought to be the main drivers of tumor relapse in patients by supporting treatment resistance and dissemination to distant organs. Both genome instability and microenvironment-driven selection support tumor heterogeneity and enable the emergence of resistant cells with stem-like properties, when therapy is applied. Besides hypoxia and nutrient deprivation, acidosis is another selection barrier in the tumor microenvironment (TME) which provides a permissive niche to shape more aggressive and fitter cancer cell phenotypes. This review describes our current knowledge about the influence of the "acidic niche" on the stem-like phenotypic features of cancer cells. In addition, we briefly survey new therapeutic options that may help eradicate CSC by integrating and/or exploiting the acidic niche, and thereby contribute to prevent the occurrence of therapy resistance as well as metastatic dissemination.
Collapse
Affiliation(s)
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| |
Collapse
|
29
|
|
30
|
Riemann A, Reime S, Thews O. Acidic extracellular environment affects miRNA expression in tumorsin vitroandin vivo. Int J Cancer 2018; 144:1609-1618. [DOI: 10.1002/ijc.31790] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/13/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Anne Riemann
- Institute of PhysiologyUniversity Halle Halle (Saale) Germany
| | - Sarah Reime
- Institute of PhysiologyUniversity Halle Halle (Saale) Germany
| | - Oliver Thews
- Institute of PhysiologyUniversity Halle Halle (Saale) Germany
| |
Collapse
|
31
|
Koyuncu I, Gonel A, Kocyigit A, Temiz E, Durgun M, Supuran CT. Selective inhibition of carbonic anhydrase-IX by sulphonamide derivatives induces pH and reactive oxygen species-mediated apoptosis in cervical cancer HeLa cells. J Enzyme Inhib Med Chem 2018; 33:1137-1149. [PMID: 30001631 PMCID: PMC6052416 DOI: 10.1080/14756366.2018.1481403] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/07/2023] Open
Abstract
Selective inhibition with sulphonamides of carbonic anhydrase (CA) IX reduces cell proliferation and induces apoptosis in human cancer cells. The effect on CA IX expression of seven previously synthesised sulphonamide inhibitors, with high affinity for CA IX, as well as their effect on the proliferation/apoptosis of cancer/normal cell lines was investigated. Two normal and three human cancer cell lines were used. Treatment resulted in dose- and time-dependent inhibition of the growth of various cancer cell lines. One compound showed remarkably high toxicity towards CA IX-positive HeLa cells. The mechanisms of apoptosis induction were determined with Annexin-V and AO/EB staining, cleaved caspases (caspase-3, caspase-8, caspase-9) and cleaved PARP activation, reactive oxygen species production (ROS), mitochondrial membrane potential (MMP), intracellular pH (pHi), extracellular pH (pHe), lactate level and cell cycle analysis. The autophagy induction mechanisms were also investigated. The modulation of apoptotic and autophagic genes (Bax, Bcl-2, caspase-3, caspase-8, caspase-9, caspase-12, Beclin and LC3) was measured using real time PCR. The positive staining using γ-H2AX and AO/EB dye, showed increased cleaved caspase-3, caspase-8, caspase-9, increased ROS production, MMP and enhanced mRNA expression of apoptotic genes, suggesting that anticancer effects are also exerted through its apoptosis-inducing properties. Our results show that such sulphonamides might have the potential as new leads for detailed investigations against CA IX-positive cervical cancers.
Collapse
Affiliation(s)
- Ismail Koyuncu
- Department of Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Ataman Gonel
- Department of Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Abdurrahim Kocyigit
- Department of Medical Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ebru Temiz
- Department of Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Mustafa Durgun
- Department of Chemistry, Faculty of Arts and Sciences, Harran University, Sanliurfa, Turkey
| | - Claudiu T. Supuran
- Laboratorio di Chimica Bioinorganica, Università degli Studi di Firenze, Polo Scientifico, Sesto Fiorentino, Florence, Italy
- Neurofarba Department, Section of Pharmaceutical and Nutriceutical Sciences, Università degli Studi di Firenze, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
32
|
Chalmin F, Bruchard M, Vegran F, Ghiringhelli F. Regulation of T cell antitumor immune response by tumor induced metabolic stress. Cell Stress 2018; 3:9-18. [PMID: 31225495 PMCID: PMC6551678 DOI: 10.15698/cst2019.01.171] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adaptive T cell immune response is essential for tumor growth control. The efficacy of immune checkpoint inhibitors is regulated by intratumoral immune response. The tumor microenvironment has a major role in adaptive immune response tuning. Tumor cells generate a particular metabolic environment in comparison to other tissues. Tumors are characterized by glycolysis, hypoxia, acidosis, amino acid depletion and fatty acid metabolism modification. Such metabolic changes promote tumor growth, impair immune response and lead to resistance to therapies. This review will detail how these modifications strongly affect CD8 and CD4 T cell functions and impact immunotherapy efficacy.
Collapse
Affiliation(s)
- Fanny Chalmin
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| | - Mélanie Bruchard
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| | - Frederique Vegran
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| | - Francois Ghiringhelli
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| |
Collapse
|
33
|
El-Sawy HS, Al-Abd AM, Ahmed TA, El-Say KM, Torchilin VP. Stimuli-Responsive Nano-Architecture Drug-Delivery Systems to Solid Tumor Micromilieu: Past, Present, and Future Perspectives. ACS NANO 2018; 12:10636-10664. [PMID: 30335963 DOI: 10.1021/acsnano.8b06104] [Citation(s) in RCA: 296] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The microenvironment characteristics of solid tumors, renowned as barriers that harshly impeded many drug-delivery approaches, were precisely studied, investigated, categorized, divided, and subdivided into a complex diverse of barriers. These categories were further studied with a particular perspective, which makes all barriers found in solid-tumor micromilieu turn into different types of stimuli, and were considered triggers that can increase and hasten drug-release targeting efficacy. This review gathers data concerning the nature of solid-tumor micromilieu. Past research focused on the treatment of such tumors, the recent efforts employed for engineering smart nanoarchitectures with the utilization of the specified stimuli categories, the possibility of combining more than one stimuli for much-greater targeting enhancement, examples of the approved nanoarchitectures that already translated clinically as well as the obstacles faced by the use of these nanostructures, and, finally, an overview of the possible future implementations of smart-chemical engineering for the design of more-efficient drug delivery and theranostic systems and for making nanosystems with a much-higher level of specificity and penetrability features.
Collapse
Affiliation(s)
- Hossam S El-Sawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy , Egyptian Russian University , Badr City , Cairo 63514 , Egypt
| | - Ahmed M Al-Abd
- Department of Pharmaceutical Sciences, College of Pharmacy , Gulf Medical University , Ajman , United Arab Emirates
- Pharmacology Department, Medical Division , National Research Centre , Giza 12622 , Egypt
| | - Tarek A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Khalid M El-Say
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , 140 The Fenway, Room 211/214, 360 Huntington Aveue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
34
|
Effects of extracellular acidity on resistance to chemotherapy treatment: a systematic review. Med Oncol 2018; 35:161. [PMID: 30377828 DOI: 10.1007/s12032-018-1214-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/24/2018] [Indexed: 10/28/2022]
Abstract
Metabolic alterations in the tumor microenvironment have a complex effect on cancer progression. Extracellular acidity is a consequence of metabolic switch in cancer and results in cell phenotypes with higher resistance to chemotherapeutics. However, mechanisms underlying the relationship between the extracellular acidity and chemoresistance are not clearly understood. This systematic review was carried out by searching the databases PubMed and EMBASE using the keywords "cancer" and "acidosis" or "acidic" and "chemoresistance" or "drug resistance." In vitro and in vivo studies that evaluated the effects of acidification of the tumor microenvironment on chemotherapeutic treatments were included. Literature reviews, letters to the editor, and articles that were not published in English were excluded. The search resulted in a total of 352 articles. After discarding 75 duplicate references, 277 articles were analyzed by sequentially reading through their titles, abstracts, and finally full-text. A total of 14 articles was selected. Acidification of the tumor microenvironment can trigger resistance through different mechanisms, such as increase in drug efflux transporters, inhibition of proton pumps, induction of the unfolded protein response (UPR), and cellular autophagy.
Collapse
|
35
|
Lacroix R, Rozeman EA, Kreutz M, Renner K, Blank CU. Targeting tumor-associated acidity in cancer immunotherapy. Cancer Immunol Immunother 2018; 67:1331-1348. [PMID: 29974196 PMCID: PMC11028141 DOI: 10.1007/s00262-018-2195-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/29/2018] [Indexed: 12/21/2022]
Abstract
Checkpoint inhibitors, such as cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and programmed cell death-1 (PD-1) monoclonal antibodies have changed profoundly the treatment of melanoma, renal cell carcinoma, non-small cell lung cancer, Hodgkin lymphoma, and bladder cancer. Currently, they are tested in various tumor entities as monotherapy or in combination with chemotherapies or targeted therapies. However, only a subgroup of patients benefit from checkpoint blockade (combinations). This raises the question, which all mechanisms inhibit T cell function in the tumor environment, restricting the efficacy of these immunotherapeutic approaches. Serum activity of lactate dehydrogenase, likely reflecting the glycolytic activity of the tumor cells and thus acidity within the tumor microenvironment, turned out to be one of the strongest markers predicting response to checkpoint inhibition. In this review, we discuss the impact of tumor-associated acidity on the efficacy of T cell-mediated cancer immunotherapy and possible approaches to break this barrier.
Collapse
Affiliation(s)
- Ruben Lacroix
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Elisa A Rozeman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Christian U Blank
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands.
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
36
|
Dantas E, Erra Díaz F, Pereyra Gerber P, Merlotti A, Varese A, Ostrowski M, Sabatté J, Geffner J. Low pH impairs complement-dependent cytotoxicity against IgG-coated target cells. Oncotarget 2018; 7:74203-74216. [PMID: 27716623 PMCID: PMC5342046 DOI: 10.18632/oncotarget.12412] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 09/20/2016] [Indexed: 12/25/2022] Open
Abstract
Local acidosis is a common feature of allergic, vascular, autoimmune, and cancer diseases. However, few studies have addressed the effect of extracellular pH on the immune response. Here, we analyzed whether low pH could modulate complement-dependent cytotoxicity (CDC) against IgG-coated cells. Using human serum as a complement source, we found that extracellular pH values of 5.5 and 6.0 strongly inhibit CDC against either B lymphoblast cell lines coated with the chimeric anti-CD20 mAb rituximab or PBMCs coated with the humanized anti-CD52 mAb alemtuzumab. Suppression of CDC by low pH was observed either in cells suspended in culture medium or in whole blood assays. Interestingly, not only CDC against IgG-coated cells, but also the activation of the complement system induced by the alternative and lectin pathways was prevented by low pH. Tumor-targeting mAbs represent one of the most successful tools for cancer therapy, however, the use of mAb monotherapy has only modest effects on solid tumors. Our present results suggest that severe acidosis, a hallmark of solid tumors, might impair complement-mediated tumor destruction directed by mAb.
Collapse
Affiliation(s)
- Ezequiel Dantas
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Fernando Erra Díaz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Pehuén Pereyra Gerber
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Antonela Merlotti
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Augusto Varese
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Matías Ostrowski
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Juan Sabatté
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| |
Collapse
|
37
|
Riemann A, Reime S, Thews O. Tumor Acidosis and Hypoxia Differently Modulate the Inflammatory Program: Measurements In Vitro and In Vivo. Neoplasia 2017; 19:1033-1042. [PMID: 29149667 PMCID: PMC5695649 DOI: 10.1016/j.neo.2017.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 12/29/2022] Open
Abstract
Inflammatory mediators produced by the tumor cells are of importance for immune response but also for malignant progression. The aim of the study was to analyze the expression of monocyte chemoattractant protein-1, interleukin-6 (IL-6), tumor necrosis factor-α, inducible isoform of nitric oxide synthase (iNOS), cyclooxygenase-2, and osteopontin in vitro in two different tumor cell lines under hypoxia (pO2 ≈ 1.5 mmHg) and/or acidosis (pH = 6.6) for up to 24 hours since hypoxia and acidosis are common characteristics of solid tumors. Additionally, the same tumor cell lines implanted in vivo were made hypoxic and acidotic artificially for 24 hours, after which the cytokine expression was measured. Finally, the activation of ERK1/2 and p38 by acidosis/hypoxia and their impact on cytokine expression were studied. The results indicate that acidosis and hypoxia have fundamentally different (often opposing) effects on cytokine expression. In addition, these effects were tumor cell line specific. When combining hypoxia and acidosis, the overall changes reflect an additive effect of both conditions alone, indicating that hypoxia and acidosis act by independent mechanisms. The in vivo changes corresponded well with the results obtained in the isolated tumor cells. Only iNOS expression was downregulated in vivo but increased in cell culture. For IL-6 expression, the acidosis-induced changes were dependent on ERK1/2 activation. In conclusion, it was demonstrated that the environmental pO2 and pH strongly affect the expression of inflammatory mediators in tumor cells. In vivo, most of the inflammatory mediators were downregulated, which could limit the activation of immune cells and by this foster the immune escape of tumors.
Collapse
Affiliation(s)
- Anne Riemann
- Julius Bernstein Institute of Physiology, University Halle-Wittenberg, Germany.
| | - Sarah Reime
- Julius Bernstein Institute of Physiology, University Halle-Wittenberg, Germany
| | - Oliver Thews
- Julius Bernstein Institute of Physiology, University Halle-Wittenberg, Germany
| |
Collapse
|
38
|
Impact of the Tumor Microenvironment on the Expression of Inflammatory Mediators in Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 923:105-111. [PMID: 27526131 DOI: 10.1007/978-3-319-38810-6_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Hypoxia and extracellular acidosis are common features of solid malignant tumors. The aim of the study was to analyze whether these pathophysiological parameters affect the expression of inflammatory mediators in tumor cells. Therefore the mRNA expression of MCP-1 (monocyte chemotactic protein 1), iNOS and osteopontin was measured under hypoxic (pO2 1 mmHg) and acidotic (pH 6.6) conditions by qPCR in AT1 R-3327 prostate cancer cells. In addition, the underlying signaling cascades were analyzed by using inhibitors of the p38 and ERK1/2 MAP kinase pathways.Hypoxia led to a significant decrease of the expression of MCP-1 and osteopontin over the complete observation period of 24 h, whereas the iNOS expression after an initial reduction slightly increased. Acidotic conditions for up to 6 h increased the iNOS expression significantly which was functional as indicated by an elevated level of nitrate/nitrite formation by 30 %. Acidosis had almost no impact on the MCP-1 expression of tumor cells, whereas the osteopontin level tended to increase leading to a significantly elevated level after 24 h at pH 6.6. Inhibiting the p38 and ERK1/2 under control conditions revealed that the MAPKs play a significant role for the regulation of the expression of inflammatory mediators. MCP-1 expression could be lowered by inhibiting ERK1/2 whereas iNOS expression was dependent on both p38 and ERK1/2 MAPK. These results indicate that the adverse tumor microenvironment affects the expression of inflammatory mediators by tumors cells and may therefore modulate the immune response within the tumor tissue.
Collapse
|
39
|
Peppicelli S, Andreucci E, Ruzzolini J, Laurenzana A, Margheri F, Fibbi G, Del Rosso M, Bianchini F, Calorini L. The acidic microenvironment as a possible niche of dormant tumor cells. Cell Mol Life Sci 2017; 74:2761-2771. [PMID: 28331999 PMCID: PMC11107711 DOI: 10.1007/s00018-017-2496-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/01/2017] [Accepted: 02/27/2017] [Indexed: 12/31/2022]
Abstract
Although surgical excision, chemo-, and radio-therapy are clearly advanced, tumors may relapse due to cells of the so-called "minimal residual disease". Indeed, small clusters of tumor cells persist in host tissues after treatment of the primary tumor elaborating strategies to survive and escape from immunological attacks before their relapse: this variable period of remission is known as "cancer dormancy". Therefore, it is crucial to understand and consider the major concepts addressing dormancy, to identify new targets and disclose potential clinical strategies. Here, we have particularly focused the relationships between tumor microenvironment and cancer dormancy, looking at a re-appreciated aspect of this compartment that is the low extracellular pH. Accumulating evidences indicate that acidity of tumor microenvironment is associated with a poor prognosis of tumor-bearing patients, stimulates a chemo- and radio-therapy resistant phenotype, and suppresses the tumoricidal activity of cytotoxic lymphocytes and natural killer cells, and all these aspects are useful for dormancy. Therefore, this review discusses the possibility that acidity of tumor microenvironment may provide a new, not previously suggested, adequate milieu for "dormancy" of tumor cells.
Collapse
MESH Headings
- Acidosis/complications
- Acidosis/immunology
- Acidosis/pathology
- Animals
- Apoptosis
- Cell Proliferation
- Humans
- Hydrogen-Ion Concentration
- Immunologic Surveillance
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Neoplasm Recurrence, Local/etiology
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm, Residual/complications
- Neoplasm, Residual/immunology
- Neoplasm, Residual/pathology
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/pathology
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Prognosis
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- Tumor Microenvironment
Collapse
Affiliation(s)
- Silvia Peppicelli
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Elena Andreucci
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Jessica Ruzzolini
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Anna Laurenzana
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Francesca Margheri
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Gabriella Fibbi
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Mario Del Rosso
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Francesca Bianchini
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy.
- Istituto Toscano Tumori, Firenze, Italy.
| | - Lido Calorini
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy.
- Istituto Toscano Tumori, Firenze, Italy.
| |
Collapse
|
40
|
Riemann A, Güttler A, Haupt V, Wichmann H, Reime S, Bache M, Vordermark D, Thews O. Inhibition of Carbonic Anhydrase IX by Ureidosulfonamide Inhibitor U104 Reduces Prostate Cancer Cell Growth, But Does Not Modulate Daunorubicin or Cisplatin Cytotoxicity. Oncol Res 2017. [PMID: 28631600 PMCID: PMC7844713 DOI: 10.3727/096504017x14965111926391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Carbonic anhydrase (CA) IX has emerged as a promising target for cancer therapy. It is highly upregulated in hypoxic regions and mediates pH regulation critical for tumor cell survival as well as extracellular acidification of the tumor microenvironment, which promotes tumor aggressiveness via various mechanisms, such as augmenting metastatic potential. Therefore, the aim of this study was to analyze the complex interdependency between CA IX and the tumor microenvironment in prostate tumor cells with regard to potential therapeutic implications. CA IX was upregulated by hypoxia as well as acidosis in prostate cancer cells. This induction did not modulate intracellular pH but led to extracellular acidification. Pharmacological inhibition of CA IX activity by U104 (SLC-0111) resulted in a reduction in tumor cell growth and an increase in apoptotic cell death. Intracellular pH was reduced under normoxic and even more so under hypoxic conditions when CA IX level was high. However, although intracellular pH regulation was disturbed, targeting CA IX in combination with daunorubicin or cisplatin did not intensify apoptotic tumor cell death. Hence, targeting CA IX in prostate cancer cells can lead to intracellular pH dysregulation and, consequently, can reduce cellular growth and elevate apoptotic cell death. Attenuation of extracellular acidification by blocking CA IX might additionally impede tumor progression and metastasis. However, no beneficial effect was seen when targeting CA IX in combination with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Anne Riemann
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Halle, Germany
| | - Antje Güttler
- Klinik und Poliklinik für Strahlentherapie, Universität Halle-Wittenberg, Halle, Germany
| | - Verena Haupt
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Halle, Germany
| | - Henri Wichmann
- Klinik und Poliklinik für Strahlentherapie, Universität Halle-Wittenberg, Halle, Germany
| | - Sarah Reime
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Halle, Germany
| | - Matthias Bache
- Klinik und Poliklinik für Strahlentherapie, Universität Halle-Wittenberg, Halle, Germany
| | - Dirk Vordermark
- Klinik und Poliklinik für Strahlentherapie, Universität Halle-Wittenberg, Halle, Germany
| | - Oliver Thews
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Halle, Germany
| |
Collapse
|
41
|
Kobayashi H. Cancer Chemotherapy Specific to Acidic Nests. Cancers (Basel) 2017; 9:cancers9040036. [PMID: 28425953 PMCID: PMC5406711 DOI: 10.3390/cancers9040036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 04/17/2017] [Accepted: 04/18/2017] [Indexed: 12/15/2022] Open
Abstract
The realization of cancer therapeutics specific to cancer cells with less of an effect on normal tissues is our goal. Many trials have been carried out for this purpose, but this goal is still far from being realized. It was found more than 80 years ago that solid cancer nests are acidified, but in vitro studies under acidic conditions have not been extensively studied. Recently, in vitro experiments under acidic conditions were started and anti-cancer drugs specific to acidic areas have been identified. Many genes have been reported to be expressed at a high level under acidic conditions, and such genes may be potent targets for anti-cancer drugs specific to acidic nests. In this review article, recent in vitro, in vivo, and clinical achievements in anti-cancer drugs with marked efficacy under acidic conditions are summarized, and the clinical use of anti-cancer drugs specific to acidic nests is discussed.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan.
| |
Collapse
|
42
|
Harguindey S, Stanciu D, Devesa J, Alfarouk K, Cardone RA, Polo Orozco JD, Devesa P, Rauch C, Orive G, Anitua E, Roger S, Reshkin SJ. Cellular acidification as a new approach to cancer treatment and to the understanding and therapeutics of neurodegenerative diseases. Semin Cancer Biol 2017; 43:157-179. [PMID: 28193528 DOI: 10.1016/j.semcancer.2017.02.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/06/2017] [Indexed: 12/27/2022]
Abstract
During the last few years, the understanding of the dysregulated hydrogen ion dynamics and reversed proton gradient of cancer cells has resulted in a new and integral pH-centric paradigm in oncology, a translational model embracing from cancer etiopathogenesis to treatment. The abnormalities of intracellular alkalinization along with extracellular acidification of all types of solid tumors and leukemic cells have never been described in any other disease and now appear to be a specific hallmark of malignancy. As a consequence of this intracellular acid-base homeostatic failure, the attempt to induce cellular acidification using proton transport inhibitors and other intracellular acidifiers of different origins is becoming a new therapeutic concept and selective target of cancer treatment, both as a metabolic mediator of apoptosis and in the overcoming of multiple drug resistance (MDR). Importantly, there is increasing data showing that different ion channels contribute to mediate significant aspects of cancer pH regulation and etiopathogenesis. Finally, we discuss the extension of this new pH-centric oncological paradigm into the opposite metabolic and homeostatic acid-base situation found in human neurodegenerative diseases (HNDDs), which opens novel concepts in the prevention and treatment of HNDDs through the utilization of a cohort of neural and non-neural derived hormones and human growth factors.
Collapse
Affiliation(s)
- Salvador Harguindey
- Institute of Clinical Biology and Metabolism, c) Postas 13, 01004 Vitoria, Spain.
| | - Daniel Stanciu
- Institute of Clinical Biology and Metabolism, c) Postas 13, 01004 Vitoria, Spain
| | - Jesús Devesa
- Department of Physiology, School of Medicine, University of Santiago de Compostela, Spain and Scientific Director of Foltra Medical Centre, Teo, Spain
| | - Khalid Alfarouk
- Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah, Saudi Arabia
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | | | - Pablo Devesa
- Research and Development, Medical Centre Foltra, Teo, Spain
| | - Cyril Rauch
- School of Veterinary Medicine and Science, University of Nottingham,College Road, Sutton Bonington, LE12 5RD, UK
| | - Gorka Orive
- Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country, Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, SLFPB-EHU, 01006 Vitoria, Spain
| | - Eduardo Anitua
- BTI Biotechnology Institute ImasD, S.L. C/Jacinto Quincoces, 39, 01007 Vitoria, Spain
| | - Sébastien Roger
- Inserm UMR1069, University François-Rabelais of Tours,10 Boulevard Tonnellé, 37032 Tours, France; Institut Universitaire de France, 1 Rue Descartes, Paris 75231, France
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
43
|
Sowa T, Menju T, Chen-Yoshikawa TF, Takahashi K, Nishikawa S, Nakanishi T, Shikuma K, Motoyama H, Hijiya K, Aoyama A, Sato T, Sonobe M, Harada H, Date H. Hypoxia-inducible factor 1 promotes chemoresistance of lung cancer by inducing carbonic anhydrase IX expression. Cancer Med 2016; 6:288-297. [PMID: 28028936 PMCID: PMC5269694 DOI: 10.1002/cam4.991] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/01/2016] [Accepted: 11/11/2016] [Indexed: 01/04/2023] Open
Abstract
Lung cancer treatment is difficult owing to chemoresistance. Hypoxia‐inducible factor 1 (HIF‐1) and HIF‐1‐induced glycolysis are correlated with chemoresistance; however, this is not evident in lung cancer. We investigated the effect of HIF‐1α and carbonic anhydrase IX (CAIX), a transmembrane protein neutralizing intracellular acidosis, on chemoresistance and prognosis of lung cancer patients after induction chemoradiotherapy. Associations of HIF‐1α, glucose transporter 1 (GLUT1), and CAIX with chemoresistance of lung cancer were investigated using A549 lung cancer cells under normoxia or hypoxia in vitro. HIF‐1α‐induced reprogramming of glucose metabolic pathway in A549 cells and the effects of HIF‐1 and CAIX on the cytotoxicity of vinorelbine were investigated. Immunohistochemical analyses were performed to determine HIF‐1α, GLUT1, and CAIX expression levels in cancer specimens from lung cancer patients after induction chemoradiotherapy. Hypoxia induced HIF‐1α expression in A549 cells. Moreover, hypoxia induced GLUT1 and CAIX expression in A549 cells in a HIF‐1‐dependent manner. Glucose metabolic pathway was shifted from oxidative phosphorylation to glycolysis by inducing HIF‐1α in A549 cells. HIF‐1 and CAIX induced chemoresistance under hypoxia, and their inhibition restored the chemosensitivity of A549 cells. The expression levels of HIF‐1α, GLUT1, and CAIX were associated with poor overall survival of lung cancer patients after induction chemoradiotherapy. HIF‐1 and CAIX affected the chemosensitivity of A549 cells and prognosis of lung cancer patients. Therefore, inhibition of HIF‐1 and CAIX might improve prognosis of lung cancer patients after induction chemoradiotherapy. Further analysis might be helpful in developing therapies for lung cancer.
Collapse
Affiliation(s)
- Terumasa Sowa
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toyofumi F Chen-Yoshikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Koji Takahashi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shigeto Nishikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takao Nakanishi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kei Shikuma
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hideki Motoyama
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kyoko Hijiya
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akihiro Aoyama
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toshihiko Sato
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Makoto Sonobe
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Radiation Biology Center, Kyoto University, Yoshida Konoecho, Sakyo-ku, Kyoto, 606-8501, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Saitama, 332-0012, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
44
|
Park SH, Seong MA, Lee HY. p38 MAPK-induced MDM2 degradation confers paclitaxel resistance through p53-mediated regulation of EGFR in human lung cancer cells. Oncotarget 2016; 7:8184-99. [PMID: 26799187 PMCID: PMC4884985 DOI: 10.18632/oncotarget.6945] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/06/2016] [Indexed: 12/31/2022] Open
Abstract
Paclitaxel (PTX) is a chemotherapeutic agent that is used to treat a variety of cancers, including non-small cell lung cancer (NSCLC). However, the emergence of drug resistance limits the utility of PTX. This study determined the signaling pathway that contributes to PTX resistance. We first established PTX resistant cell lines (H460/R and 226B/R) using a dose-escalating maintenance of PTX. We found that p38 MAPK and epidermal growth factor receptor (EGFR) were constitutively activated in these cell lines. The inhibition of p38 MAPK activity by SB203580 treatment or the transfection of dominant-negative p38 MAPK sensitized both cell lines to PTX treatment. Erlotinib, an EGFR inhibitor, also increased PTX-induced apoptosis in PTX resistant cells, which suggests a role for p38 MAPK and EGFR in the development of PTX resistance. We demonstrated that p38 MAPK enhanced EGFR expression via the induction of the rapid degradation of mouse double-minute 2 homolog (MDM2) and the consequent stabilization of p53, a transcription factor of EGFR. These results suggest for the first time that the p38 MAPK/p53/EGFR axis is crucial for the facilitation of PTX resistance in NSCLCs. We also propose a mechanism for the role of the tumor-suppressor p53 in drug resistance. These results provide a foundation for the future development of potential therapeutic strategies to regulate the p38 MAPK/p53/EGFR pathway for the treatment of lung cancer patients with PTX resistance.
Collapse
Affiliation(s)
- Shin-Hyung Park
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Myeong-A Seong
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ho-Young Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
45
|
Martin JD, Fukumura D, Duda DG, Boucher Y, Jain RK. Reengineering the Tumor Microenvironment to Alleviate Hypoxia and Overcome Cancer Heterogeneity. Cold Spring Harb Perspect Med 2016; 6:a027094. [PMID: 27663981 PMCID: PMC5131751 DOI: 10.1101/cshperspect.a027094] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Solid tumors consist of cancer cells and stromal cells, including resident and transiting immune cells-all ensconced in an extracellular matrix (ECM)-nourished by blood vessels and drained by lymphatic vessels. The microenvironment constituents are abnormal and heterogeneous in morphology, phenotype, and physiology. Such irregularities include an inefficient tumor vascular network comprised of leaky and compressed vessels, which impair blood flow and oxygen delivery. Low oxygenation in certain tumor regions-or focal hypoxia-is a mediator of cancer progression, metastasis, immunosuppression, and treatment resistance. Thus, repairing an abnormal and heterogeneous microenvironment-and hypoxia in particular-can significantly improve treatments of solid tumors. Here, we summarize two strategies to reengineer the tumor microenvironment (TME)-vessel normalization and decompression-that can alleviate hypoxia. In addition, we discuss how these two strategies alone and in combination with each other-or other therapeutic strategies-may overcome the challenges posed by cancer heterogeneity.
Collapse
Affiliation(s)
- John D Martin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Yves Boucher
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
46
|
Brown JS, Cunningham JJ, Gatenby RA. Aggregation Effects and Population-Based Dynamics as a Source of Therapy Resistance in Cancer. IEEE Trans Biomed Eng 2016; 64:512-518. [PMID: 28113286 DOI: 10.1109/tbme.2016.2623564] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Evolution of resistance allows cancer cells to adapt and continue proliferating even when therapy is initially very effective. Most investigations of treatment resistance focus on the adaptive phenotypic properties of individual cells. We propose that the resistance of a single cell to therapy may extend beyond its own phenotypic and molecular properties and be influenced by the phenotypic properties of surrounding cells and variations in cell density. Similar variation exists in population densities of animals living in groups and can significantly affect the outcome of an external threat. METHODS We investigate aggregation effects in cancer therapy using Darwinian models that integrate phenotypic properties of individual cells and common population effects found in nature to simulate the dynamics of resistance and sensitivity in the diverse cellular environments within cancers. RESULTS We demonstrate that the density of cancer cell populations can profoundly influence response to chemotherapy independent of the properties of individual cells. Most commonly, these aggregation effects benefit the tumor allowing cells to survive even with phenotypic properties that would render them highly vulnerable to therapy in the absence of population effects. CONCLUSION We demonstrate aggregation effects likely play a significant role in conferring resistance to therapy on tumor cells that would otherwise be sensitive to treatment. SIGNIFICANCE The potential role of aggregation in outcomes from cancer therapy has not been previously investigated. Our results demonstrate these dynamics may play a key role in resistance to therapy and could be used to design evolutionarily-enlightened therapies that exploit aggregation effects to improve treatment outcomes.
Collapse
|
47
|
Longo DL, Bartoli A, Consolino L, Bardini P, Arena F, Schwaiger M, Aime S. In Vivo Imaging of Tumor Metabolism and Acidosis by Combining PET and MRI-CEST pH Imaging. Cancer Res 2016; 76:6463-6470. [PMID: 27651313 DOI: 10.1158/0008-5472.can-16-0825] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/13/2016] [Accepted: 08/15/2016] [Indexed: 11/16/2022]
Abstract
The vast majority of cancers exhibit increased glucose uptake and glycolysis regardless of oxygen availability. This metabolic shift leads to an enhanced production of lactic acid that decreases extracellular pH (pHe), a hallmark of the tumor microenvironment. In this way, dysregulated tumor pHe and upregulated glucose metabolism are linked tightly and their relative assessment may be useful to gain understanding of the underlying biology. Here we investigated noninvasively the in vivo correlation between tumor 18F-FDG uptake and extracellular pH values in a murine model of HER2+ breast cancer. Tumor extracellular pH and perfusion were assessed by acquiring MRI-CEST (chemical exchange saturation transfer) images on a 3T scanner after intravenous administration of a pH-responsive contrast agent (iopamidol). Static PET images were recorded immediately after MRI acquisitions to quantify the extent of 18F-FDG uptake. We demonstrated the occurrence of tumor pHe changes that report on acidification of the interstitial fluid caused by an accelerated glycolysis. Combined PET and MRI-CEST images reported complementary spatial information of the altered glucose metabolism. Notably, a significant inverse correlation was found between extracellular tumor pH and 18F-FDG uptake, as a high 18F-FDG uptake corresponds to lower extracellular pH values. These results show how merging the information from 18F-FDG-uptake and extracellular pH measurements can improve characterization of the tumor microenvironment. Cancer Res; 76(22); 6463-70. ©2016 AACR.
Collapse
Affiliation(s)
- Dario L Longo
- Institute of Biostructure and Bioimaging (CNR) c/o Molecular Biotechnologies Center, Torino, Italy.,Molecular Imaging Center, University of Torino, Torino, Italy
| | - Antonietta Bartoli
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lorena Consolino
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Paola Bardini
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Francesca Arena
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universitat Munchen, Munich, Germany
| | - Silvio Aime
- Molecular Imaging Center, University of Torino, Torino, Italy. .,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
48
|
Zhu H, Wu J, Zhang W, Luo H, Shen Z, Cheng H, Zhu X. PKM2 enhances chemosensitivity to cisplatin through interaction with the mTOR pathway in cervical cancer. Sci Rep 2016; 6:30788. [PMID: 27492148 PMCID: PMC4974606 DOI: 10.1038/srep30788] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 07/11/2016] [Indexed: 12/22/2022] Open
Abstract
Pyruvate kinase M2 (PKM2) is a key driver of aerobic glycolysis in cancer cells and has been shown to be up-regulated by mTOR in vitro. Our previous proteomic profiling studies showed that PKM2 was significantly upregulated in cervical cancer tissues after treatment with neoadjuvant chemotherapy (NACT). Whether PKM2 expression predicts cisplatin-based NACT sensitivity and is mTOR dependent in cervical cancer patients remains unclear. Using paired tumor samples (pre- and post-chemotherapy) from 36 cervical cancer patients, we examined mTOR, HIF-1α, c-Myc, and PKM2 expression in cervical cancer samples and investigated the response to cisplatin-based NACT. In addition, we established PKM2 suppressed cervical cancer cell lines and evaluated their sensitivity to cisplatin in vitro. We found that the mTOR/HIF-1α/c-Myc/PKM2 signaling pathway was significantly downregulated in post-chemotherapy cervical cancer tissues. High levels of mTOR, HIF-1α, c-Myc, and PKM2 were associated with a positive chemotherapy response in cervical cancer patients treated with cisplatin-based NACT. In vitro, PKM2 knockdown desensitized cervical cancer cells to cisplatin. Moreover, PKM2 had complex interactions with mTOR pathways. mTOR, HIF1α, c-Myc, and PKM2 expression in cervical cancer may serve as predictive biomarkers to cisplatin-based chemotherapy. PKM2 enhances chemosensitivity to cisplatin through interaction with the mTOR pathway in cervical cancer.
Collapse
Affiliation(s)
- Haiyan Zhu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jun Wu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Wenwen Zhang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Hui Luo
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhaojun Shen
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huihui Cheng
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
49
|
Filatova A, Seidel S, Böğürcü N, Gräf S, Garvalov BK, Acker T. Acidosis Acts through HSP90 in a PHD/VHL-Independent Manner to Promote HIF Function and Stem Cell Maintenance in Glioma. Cancer Res 2016; 76:5845-5856. [PMID: 27488520 DOI: 10.1158/0008-5472.can-15-2630] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 07/13/2016] [Indexed: 11/16/2022]
Abstract
Hypoxia is a common feature of solid tumors, which controls multiple aspects of cancer progression. One important function of hypoxia and the hypoxia-inducible factors (HIF) is the maintenance of cancer stem-like cells (CSC), a population of tumor cells that possess stem cell-like properties and drives tumor growth. Among the changes promoted by hypoxia is a metabolic shift resulting in acidification of the tumor microenvironment. Here, we show that glioma hypoxia and acidosis functionally cooperate in inducing HIF transcription factors and CSC maintenance. We found that these effects did not involve the classical PHD/VHL pathway for HIF upregulation, but instead involved the stress-induced chaperone protein HSP90. Genetic or pharmacologic inactivation of HSP90 inhibited the increase in HIF levels and abolished the self-renewal and tumorigenic properties of CSCs induced by acidosis. In clinical specimens of glioma, HSP90 was upregulated in the hypoxic niche and was correlated with a CSC phenotype. Our findings highlight the role of tumor acidification within the hypoxic niche in the regulation of HIF and CSC function through HSP90, with implications for therapeutic strategies to target CSC in gliomas and other hypoxic tumors. Cancer Res; 76(19); 5845-56. ©2016 AACR.
Collapse
Affiliation(s)
- Alina Filatova
- Institute of Neuropathology, University of Giessen, Giessen, Germany
| | - Sascha Seidel
- Institute of Neuropathology, University of Giessen, Giessen, Germany. Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Nuray Böğürcü
- Institute of Neuropathology, University of Giessen, Giessen, Germany
| | - Sabine Gräf
- Institute of Neuropathology, University of Giessen, Giessen, Germany
| | - Boyan K Garvalov
- Institute of Neuropathology, University of Giessen, Giessen, Germany
| | - Till Acker
- Institute of Neuropathology, University of Giessen, Giessen, Germany.
| |
Collapse
|
50
|
Mondal SK, Jinka S, Pal K, Nelli S, Dutta SK, Wang E, Ahmad A, AlKharfy KM, Mukhopadhyay D, Banerjee R. Glucocorticoid Receptor-Targeted Liposomal Codelivery of Lipophilic Drug and Anti-Hsp90 Gene: Strategy to Induce Drug-Sensitivity, EMT-Reversal, and Reduced Malignancy in Aggressive Tumors. Mol Pharm 2016; 13:2507-23. [PMID: 27184196 DOI: 10.1021/acs.molpharmaceut.6b00230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many cancers including the late stage ones become drug-resistant and undergo epithelial-to-mesenchymal transition (EMT). These lead to enhanced invasion, migration, and metastasis toward manifesting its aggressiveness and malignancy. One of the key hallmarks of cancer is its overdependence on glycolysis as its preferred energy metabolism pathway. The strict avoidance of alternate energy pathway gluconeogenesis by cancer cells points to a yet-to-be hoisted role of glucocorticoid receptor (GR) especially in tumor microenvironment, where cells are known to become drug-sensitive through induction of gluconeogenesis. However, since GR is involved in metabolism, anti-inflammatory reactions, immunity besides inducing gluconeogenesis, a greater role of GR in tumor microenvironment is envisaged. We have shown previously that GR, although ubiquitously expressed in all cells; afford to be an effective cytoplasmic target for killing cancer cells selectively. Herein, we report the therapeutic use of a newly developed GR-targeted liposomal concoction (DXE) coformulating a lipophilic drug (ESC8) and an anti-Hsp90 anticancer gene against aggressive tumor models. This induced drug-sensitivity and apoptosis while reversing EMT in tumor cells toward effective retardation of aggressive growth in pancreas and skin tumor models. Additionally, the ESC8-free lipid formulation upon cotreatment with hydrophilic drugs, gemcitabine and doxorubicin, could effectively sensitize and kill pancreatic cancer and melanoma cells, respectively. The formulation-triggered EMT-reversal was GR-dependent. Overall, we found a new strategy for drug sensitization that led to the advent of new GR-targeted anticancer therapeutics.
Collapse
Affiliation(s)
- Sujan Kumar Mondal
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India.,Academy of Scientific & Innovative Research (AcSIR) , 2 Rafi Marg, New Delhi 110001, India
| | - Sudhakar Jinka
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India.,Academy of Scientific & Innovative Research (AcSIR) , 2 Rafi Marg, New Delhi 110001, India
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Swetha Nelli
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, King Saud University , Riyadh 11451, Saudi Arabia
| | - Khalid M AlKharfy
- Department of Clinical Pharmacy, King Saud University , Riyadh 11451, Saudi Arabia
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Rajkumar Banerjee
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India.,Academy of Scientific & Innovative Research (AcSIR) , 2 Rafi Marg, New Delhi 110001, India
| |
Collapse
|