1
|
Lavudi K, Nuguri SM, Pandey P, Kokkanti RR, Wang QE. ALDH and cancer stem cells: Pathways, challenges, and future directions in targeted therapy. Life Sci 2024; 356:123033. [PMID: 39222837 DOI: 10.1016/j.lfs.2024.123033] [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: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Human ALDH comprise 19 subfamilies in which ALDH1A1, ALDH1A3, ALDH3A1, ALDH5A1, ALDH7A1, and ALDH18A1 are implicated in CSC. Studies have shown that ALDH can also be involved in drug resistance and standard chemotherapy regimens are ineffective in treating patients at the stage of disease recurrence. Existing chemotherapeutic drugs eliminate the bulk of tumors but are usually not effective against CSC which express ALDH+ population. Henceforth, targeting ALDH is convincing to treat the patient's post-relapse. Combination therapies that interlink signaling mechanisms seem promising to increase the overall disease-free survival rate. Therefore, targeting ALDH through ALDH inhibitors along with immunotherapies may create a novel platform for translational research. This review aims to fill in the gap between ALDH1 family members in relation to its cell signaling mechanisms, highlighting their potential as molecular targets to sensitize recurrent tumors and bring forward the future development concerning the current progress and draw backs. This review summarizes the role of cancer stem cells and their upregulation by maintaining the tumor microenvironment in which ALDH is specifically highlighted. It discusses the regulation of ALDH family proteins and the crosstalk between ALDH and CSC in relation to cancer metabolism. Furthermore, it establishes the correlation between ALDH involved signaling mechanisms and their specific targeted inhibitors, as well as their functional modularity, bioavailability, and mechanistic role in various cancers.
Collapse
Affiliation(s)
- Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States
| | - Shreya Madhav Nuguri
- Department of Food science and Technology, The Ohio State University, Columbus, OH, United States
| | - Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, U.P., India; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | | | - Qi-En Wang
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
2
|
Acharya SK, Shai S, Choon YF, Gunardi I, Hartanto FK, Kadir K, Roychoudhury A, Amtha R, Vincent-Chong VK. Cancer Stem Cells in Oral Squamous Cell Carcinoma: A Narrative Review on Experimental Characteristics and Methodological Challenges. Biomedicines 2024; 12:2111. [PMID: 39335624 PMCID: PMC11429394 DOI: 10.3390/biomedicines12092111] [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: 07/14/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation of cancer cells that are believed to initiate and drive cancer progression. In animal models, xenotransplanted CSCs have demonstrated the ability to produce tumors. Since their initial isolation in blood cancers, CSCs have been identified in various solid human cancers, including oral squamous cell carcinoma (OSCC). In addition to their tumorigenic properties, dysregulated stem-cell-related signaling pathways-Wnt family member (Wnt), neurogenic locus notch homolog protein (Notch), and hedgehog-have been shown to endow CSCs with characteristics like self-renewal, phenotypic plasticity, and chemoresistance, contributing to recurrence and treatment failure. Consequently, CSCs have become targets for new therapeutic agents, with some currently in different phases of clinical trials. Notably, small molecule inhibitors of the hedgehog signaling pathway, such as vismodegib and glasdegib, have been approved for the treatment of basal cell carcinoma and acute myeloid leukemia, respectively. Other strategies for eradicating CSCs include natural compounds, nano-drug delivery systems, targeting mitochondria and the CSC microenvironment, autophagy, hyperthermia, and immunotherapy. Despite the extensive documentation of CSCs in OSCC since its first demonstration in head and neck (HN) SCC in 2007, none of these novel pharmacological approaches have yet entered clinical trials for OSCC patients. This narrative review summarizes the in vivo and in vitro evidence of CSCs and CSC-related signaling pathways in OSCC, highlighting their role in promoting chemoresistance and immunotherapy resistance. Additionally, it addresses methodological challenges and discusses future research directions to improve experimental systems and advance CSC studies.
Collapse
Affiliation(s)
- Surendra Kumar Acharya
- Department of Oral Medicine, Radiology and Surgery, Faculty of Dentistry, Lincoln University College, Petaling Jaya 47301, Selangor, Malaysia
| | - Saptarsi Shai
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Yee Fan Choon
- Department of Oral and Maxillofacial Surgical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom 42610, Selangor, Malaysia;
| | - Indrayadi Gunardi
- Oral Medicine Department, Faculty of Dentistry, Universitas Trisakti, Jakarta 11440, Indonesia; (I.G.); (F.K.H.)
| | - Firstine Kelsi Hartanto
- Oral Medicine Department, Faculty of Dentistry, Universitas Trisakti, Jakarta 11440, Indonesia; (I.G.); (F.K.H.)
| | - Kathreena Kadir
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Ajoy Roychoudhury
- Department of Oral and Maxillofacial Surgery, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Rahmi Amtha
- Oral Medicine Department, Faculty of Dentistry, Universitas Trisakti, Jakarta 11440, Indonesia; (I.G.); (F.K.H.)
| | - Vui King Vincent-Chong
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
3
|
Guo S, Zheng S, Liu M, Wang G. Novel Anti-Cancer Stem Cell Compounds: A Comprehensive Review. Pharmaceutics 2024; 16:1024. [PMID: 39204369 PMCID: PMC11360402 DOI: 10.3390/pharmaceutics16081024] [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: 05/30/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer stem cells (CSCs) possess a significant ability to renew themselves, which gives them a strong capacity to form tumors and expand to encompass additional body areas. In addition, they possess inherent resistance to chemotherapy and radiation therapies used to treat many forms of cancer. Scientists have focused on investigating the signaling pathways that are highly linked to the ability of CSCs to renew themselves and maintain their stem cell properties. The pathways encompassed are Notch, Wnt/β-catenin, hedgehog, STAT3, NF-κB, PI-3K/Akt/mTOR, sirtuin, ALDH, MDM2, and ROS. Recent studies indicate that directing efforts towards CSC cells is essential in eradicating the overall cancer cell population and reducing the likelihood of tumor metastasis. As our comprehension of the mechanisms that stimulate CSC activity, growth, and resistance to chemotherapy advances, the discovery of therapeutic drugs specifically targeting CSCs, such as small-molecule compounds, holds the potential to revolutionize cancer therapy. This review article examines and analyzes the novel anti-CSC compounds that have demonstrated effective and selective targeting of pathways associated with the renewal and stemness of CSCs. We also discussed their special drug metabolism and absorption mechanisms. CSCs have been the subject of much study in cancer biology. As a possible treatment for malignancies, small-molecule drugs that target CSCs are gaining more and more attention. This article provides a comprehensive review of the current state of key small-molecule compounds, summarizes their recent developments, and anticipates the future discovery of even more potent and targeted compounds, opening up new avenues for cancer treatment.
Collapse
Affiliation(s)
- Shanchun Guo
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Shilong Zheng
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Mingli Liu
- Department of Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Guangdi Wang
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| |
Collapse
|
4
|
Li Z, Yang J, Ren B, Fan Q, Huang L, Guo S, Zhou R, Chen S, Feng J, Yan C, Chen X, Shen Z. Double-Layered Hollow Mesoporous Cuprous Oxide Nanoparticles for Double Drug Sequential Therapy of Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313212. [PMID: 38670140 DOI: 10.1002/adma.202313212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/08/2024] [Indexed: 04/28/2024]
Abstract
Cancer stem cells (CSCs) are one of the determinants of tumor heterogeneity and are characterized by self-renewal, high tumorigenicity, invasiveness, and resistance to various therapies. To overcome the resistance of traditional tumor therapies resulting from CSCs, a strategy of double drug sequential therapy (DDST) for CSC-enriched tumors is proposed in this study and is realized utilizing the developed double-layered hollow mesoporous cuprous oxide nanoparticles (DL-HMCONs). The high drug-loading contents of camptothecin (CPT) and all-trans retinoic acid (ATRA) demonstrate that the DL-HMCON can be used as a generic drug delivery system. ATRA and CPT can be sequentially loaded in and released from CPT3@ATRA3@DL-HMCON@HA. The DDST mechanisms of CPT3@ATRA3@DL-HMCON@HA for CSC-containing tumors are demonstrated as follows: 1) the first release of ATRA from the outer layer induces differentiation from CSCs with high drug resistance to non-CSCs with low drug resistance; 2) the second release of CPT from the inner layer causes apoptosis of non-CSCs; and 3) the third release of Cu+ from DL-HMCON itself triggers the Fenton-like reaction and glutathione depletion, resulting in ferroptosis of non-CSCs. This CPT3@ATRA3@DL-HMCON@HA is verified to possess high DDST efficacy for CSC-enriched tumors with high biosafety.
Collapse
Affiliation(s)
- Zongheng Li
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jing Yang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Bin Ren
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Qingdeng Fan
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Lin Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Shuai Guo
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - RuiLong Zhou
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Sijin Chen
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jie Feng
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Chenggong Yan
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
5
|
Ajmeera D, Ajumeera R. Drug repurposing: A novel strategy to target cancer stem cells and therapeutic resistance. Genes Dis 2024; 11:148-175. [PMID: 37588226 PMCID: PMC10425757 DOI: 10.1016/j.gendis.2022.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/21/2022] [Accepted: 12/08/2022] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy is an effortless and frequently used approach in cancer therapy. However, in most cases, it can only prolong life expectancy and does not guarantee a complete cure. Furthermore, chemotherapy is associated with severe adverse effects, one of the major complications of effective cancer therapy. In addition, newly published research outputs show that cancer stem cells are involved in cancer disease progression, drug resistance, metastasis, and recurrence and that they are functional in the trans-differentiation capacity of cancer stem cells to cancer cells in response to treatments. Novel strategies are therefore required for better management of cancer therapy. The prime approach would be to synthesize and develop novel drugs that need extensive resources, time, and endurance to be brought into therapeutic use. The subsequent approach would be to screen the anti-cancer activity of available non-cancerous drugs. This concept of repurposing non-cancer drugs as an alternative to current cancer therapy has become popular in recent years because using existing anticancer drugs has several adverse effects. Micronutrients have also been investigated for cancer therapy due to their significant anti-cancer effects with negligible or no side effects and availability in food sources. In this paper, we discuss an ideal hypothesis for screening available non-cancerous drugs with anticancer activity, with a focus on cancer stem cells and their clinical application for cancer treatment. Further, drug repurposing and the combination of micronutrients that can target both cancers and cancer stem cells may result in a better therapeutic approach leading to maximum tumor growth control.
Collapse
Affiliation(s)
- Divya Ajmeera
- Cell Biology Department, ICMR-National Institute of Nutrition (NIN), Hyderabad, Telangana 500007, India
| | - Rajanna Ajumeera
- Cell Biology Department, ICMR-National Institute of Nutrition (NIN), Hyderabad, Telangana 500007, India
| |
Collapse
|
6
|
Jan N, Sofi S, Qayoom H, Haq BU, Shabir A, Mir MA. Targeting breast cancer stem cells through retinoids: A new hope for treatment. Crit Rev Oncol Hematol 2023; 192:104156. [PMID: 37827439 DOI: 10.1016/j.critrevonc.2023.104156] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/09/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023] Open
Abstract
Breast cancer is a complex and diverse disease accounting for nearly 30% of all cancers diagnosed in females. But unfortunately, patients develop resistance to the existing chemotherapeutic regimen, resulting in approximately 90% treatment failure. With over half a million deaths annually, it is imperative to explore new therapeutic approaches to combat the disease. Within a breast tumor, a small sub-population of heterogeneous cells, with a unique ability of self-renew and differentiation and responsible for tumor formation, initiation, and recurrence are referred to as breast cancer stem cells (BCSCs). These BCSCs have been identified as one of the main contributors to chemoresistance in breast cancer, making them an attractive target for developing novel therapeutic strategies. These cells exhibit surface biomarkers such as CD44+, CD24-/LOW, ALDH, CD133, and CD49f phenotypes. Higher expression of CD44+ and ALDH activity has been associated with the formation of tumors in various cancers. Moreover, the abnormal regulation of signaling pathways, including Hedgehog, Notch, β-catenin, JAK/STAT, and P13K/AKT/mTOR, leads to the formation of cancer stem cells, resulting in the development of tumors. The growing drug resistance in BC is a significant challenge, highlighting the need for new therapeutic strategies to combat this dreadful disease. Retinoids, a large group of synthetic derivatives of vitamin A, have been studied as chemopreventive agents in clinical trials and have been shown to regulate various crucial biological functions including vision, development, inflammation, and metabolism. On a cellular level, the retinoid activity has been well characterized and translated and is known to induce differentiation and apoptosis, which play important roles in the outcome of the transformation of tissues into malignant. Retinoids have been investigated extensively for their use in the treatment and prevention of cancer due to their high receptor-binding affinity to directly modulate gene expression programs. Therefore, in this study, we aim to summarize the current understanding of BCSCs, their biomarkers, and the associated signaling pathways. Retinoids, such as Adapalene, a third-generation retinoid, have shown promising anti-cancer potential and may serve as therapeutic agents to target BCSCs.
Collapse
Affiliation(s)
- Nusrat Jan
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Burhan Ul Haq
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Aisha Shabir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India.
| |
Collapse
|
7
|
Yehya A, Youssef J, Hachem S, Ismael J, Abou-Kheir W. Tissue-specific cancer stem/progenitor cells: Therapeutic implications. World J Stem Cells 2023; 15:323-341. [PMID: 37342220 PMCID: PMC10277968 DOI: 10.4252/wjsc.v15.i5.323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 05/26/2023] Open
Abstract
Surgical resection, chemotherapy, and radiation are the standard therapeutic modalities for treating cancer. These approaches are intended to target the more mature and rapidly dividing cancer cells. However, they spare the relatively quiescent and intrinsically resistant cancer stem cells (CSCs) subpopulation residing within the tumor tissue. Thus, a temporary eradication is achieved and the tumor bulk tends to revert supported by CSCs' resistant features. Based on their unique expression profile, the identification, isolation, and selective targeting of CSCs hold great promise for challenging treatment failure and reducing the risk of cancer recurrence. Yet, targeting CSCs is limited mainly by the irrelevance of the utilized cancer models. A new era of targeted and personalized anti-cancer therapies has been developed with cancer patient-derived organoids (PDOs) as a tool for establishing pre-clinical tumor models. Herein, we discuss the updated and presently available tissue-specific CSC markers in five highly occurring solid tumors. Additionally, we highlight the advantage and relevance of the three-dimensional PDOs culture model as a platform for modeling cancer, evaluating the efficacy of CSC-based therapeutics, and predicting drug response in cancer patients.
Collapse
Affiliation(s)
- Amani Yehya
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Joe Youssef
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Sana Hachem
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Jana Ismael
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
8
|
An anti-alcoholism drug, disulfiram and copper complex improves radio-resistance of tumor-initiating cells in esophageal squamous cell carcinoma. Esophagus 2023; 20:134-142. [PMID: 36121574 DOI: 10.1007/s10388-022-00948-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/13/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a malignant cancer with a poor prognosis. Chemoradiotherapy is one of the most important strategies for patients with locally advanced unresectable ESCC; however, its therapeutic effect is unsatisfactory. Tumor-initiating cells (TICs) have been reported to be resistant to conventional chemotherapy and radiotherapy so far. Therefore, we aimed to develop a treatment strategy targeting TICs in ESCC to improve radiosensitivity. METHODS First, we validated aldehyde dehydrogenase 1 (ALDH1) as a TIC marker and investigated its ability to mediate resistance in human ESCC cell lines using flow cytometry, Western blotting, and functional analyses. Then, we focused on disulfiram (DSF), an aldehyde dehydrogenase inhibitor, used to treat alcohol use disorder. We investigated the effect of DSF and copper (II) D-gluconate (Cu) on the radiosensitivity of ESCC in xenograft mouse models. RESULTS ALDH1-positive cells showed an upregulation of SOX2 and Nanog, exhibiting much stronger tumor-initiating properties than ALDH1-negative cells. Furthermore, inhibition of ALDH1 attenuated the tumor-initiating properties of ESCC cell lines. Our results also showed that ALDH1-positive cells were resistant to chemotherapy and radiotherapy, and the inhibition of ALDH1 led to the mitigation of therapeutic resistance. Our in vitro and in vivo studies revealed that the DSF/Cu complex could radiosensitize ALDH1-positive ESCC cells and downregulate the phosphoinositide 3-kinase/Akt pathway. CONCLUSION ALDH1 inhibition by the DSF/Cu complex enhances the radiosensitivity of TICs in ESCC. The drug repositioning approach using disulfiram is a potential treatment option to overcome radioresistance in patients with locally advanced ESCC.
Collapse
|
9
|
Chu Y, Luo Y, Su B, Li C, Guo Q, Zhang Y, Liu P, Chen H, Zhao Z, Zhou Z, Wang Y, Jiang C, Sun T. A neutrophil-biomimic platform for eradicating metastatic breast cancer stem-like cells by redox microenvironment modulation and hypoxia-triggered differentiation therapy. Acta Pharm Sin B 2023; 13:298-314. [PMID: 36815033 PMCID: PMC9939302 DOI: 10.1016/j.apsb.2022.05.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/10/2022] [Accepted: 05/15/2022] [Indexed: 11/18/2022] Open
Abstract
Metastasis accounts for 90% of breast cancer deaths, where the lethality could be attributed to the poor drug accumulation at the metastatic loci. The tolerance to chemotherapy induced by breast cancer stem cells (BCSCs) and their particular redox microenvironment further aggravate the therapeutic dilemma. To be specific, therapy-resistant BCSCs can differentiate into heterogeneous tumor cells constantly, and simultaneously dynamic maintenance of redox homeostasis promote tumor cells to retro-differentiate into stem-like state in response to cytotoxic chemotherapy. Herein, we develop a specifically-designed biomimic platform employing neutrophil membrane as shell to inherit a neutrophil-like tumor-targeting capability, and anchored chemotherapeutic and BCSCs-differentiating reagents with nitroimidazole (NI) to yield two hypoxia-responsive prodrugs, which could be encapsulated into a polymeric nitroimidazole core. The platform can actively target the lung metastasis sites of triple negative breast cancer (TNBC), and release the escorted drugs upon being triggered by the hypoxia microenvironment. During the responsiveness, the differentiating agent could promote transferring BCSCs into non-BCSCs, and simultaneously the nitroimidazole moieties conjugated on the polymer and prodrugs could modulate the tumor microenvironment by depleting nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) and amplifying intracellular oxidative stress to prevent tumor cells retro-differentiation into BCSCs. In combination, the BCSCs differentiation and tumor microenvironment modulation synergistically could enhance the chemotherapeutic cytotoxicity, and remarkably suppress tumor growth and lung metastasis. Hopefully, this work can provide a new insight in to comprehensively treat TNBC and lung metastasis using a versatile platform.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Chen Jiang
- Corresponding author. Tel./fax: +86 21 5198 0079.
| | - Tao Sun
- Corresponding author. Tel./fax: +86 21 5198 0079.
| |
Collapse
|
10
|
Nallasamy P, Nimmakayala RK, Parte S, Are AC, Batra SK, Ponnusamy MP. Tumor microenvironment enriches the stemness features: the architectural event of therapy resistance and metastasis. Mol Cancer 2022; 21:225. [PMID: 36550571 PMCID: PMC9773588 DOI: 10.1186/s12943-022-01682-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer divergence has many facets other than being considered a genetic term. It is a tremendous challenge to understand the metastasis and therapy response in cancer biology; however, it postulates the opportunity to explore the possible mechanism in the surrounding tumor environment. Most deadly solid malignancies are distinctly characterized by their tumor microenvironment (TME). TME consists of stromal components such as immune, inflammatory, endothelial, adipocytes, and fibroblast cells. Cancer stem cells (CSCs) or cancer stem-like cells are a small sub-set of the population within cancer cells believed to be a responsible player in the self-renewal, metastasis, and therapy response of cancer cells. The correlation between TME and CSCs remains an enigma in understanding the events of metastasis and therapy resistance in cancer biology. Recent evidence suggests that TME dictates the CSCs maintenance to arbitrate cancer progression and metastasis. The immune, inflammatory, endothelial, adipocyte, and fibroblast cells in the TME release growth factors, cytokines, chemokines, microRNAs, and exosomes that provide cues for the gain and maintenance of CSC features. These intricate cross-talks are fueled to evolve into aggressive, invasive, migratory phenotypes for cancer development. In this review, we have abridged the recent developments in the role of the TME factors in CSC maintenance and how these events influence the transition of tumor progression to further translate into metastasis and therapy resistance in cancer.
Collapse
Affiliation(s)
- Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Seema Parte
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Abhirup C Are
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
11
|
Lo Iacono M, Gaggianesi M, Bianca P, Brancato OR, Muratore G, Modica C, Roozafzay N, Shams K, Colarossi L, Colarossi C, Memeo L, Turdo A, Veschi V, Di Franco S, Todaro M, Stassi G. Destroying the Shield of Cancer Stem Cells: Natural Compounds as Promising Players in Cancer Therapy. J Clin Med 2022; 11:6996. [PMID: 36498571 PMCID: PMC9737492 DOI: 10.3390/jcm11236996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
In a scenario where eco-sustainability and a reduction in chemotherapeutic drug waste are certainly a prerogative to safeguard the biosphere, the use of natural products (NPs) represents an alternative therapeutic approach to counteract cancer diseases. The presence of a heterogeneous cancer stem cell (CSC) population within a tumor bulk is related to disease recurrence and therapy resistance. For this reason, CSC targeting presents a promising strategy for hampering cancer recurrence. Increasing evidence shows that NPs can inhibit crucial signaling pathways involved in the maintenance of CSC stemness and sensitize CSCs to standard chemotherapeutic treatments. Moreover, their limited toxicity and low costs for large-scale production could accelerate the use of NPs in clinical settings. In this review, we will summarize the most relevant studies regarding the effects of NPs derived from major natural sources, e.g., food, botanical, and marine species, on CSCs, elucidating their use in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Melania Lo Iacono
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Paola Bianca
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Ornella Roberta Brancato
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Giampaolo Muratore
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Chiara Modica
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Narges Roozafzay
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Kimiya Shams
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Lorenzo Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Cristina Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Alice Turdo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Veronica Veschi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Simone Di Franco
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
12
|
Relationship between Aldehyde Dehydrogenase, PD-L1 and Tumor-Infiltrating Lymphocytes with Pathologic Response and Survival in Breast Cancer. Cancers (Basel) 2022; 14:cancers14184418. [PMID: 36139578 PMCID: PMC9496850 DOI: 10.3390/cancers14184418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Aldehyde dehydrogenase 1A1 (ALDH1A1) is a cancer stem cell (CSC) marker related to clinical outcomes in breast cancer (BC). The aim of this study was to analyze the relationship between ALDH1A1, programmed death ligand 1 (PD-L1) and tumor-infiltrating lymphocytes (TILs) in triple negative (TN) and human epidermal growth factor receptor 2-positive (HER2+) BC tumors, and its association with clinicopathological characteristics and outcomes. A retrospective, historical cohort study of patients diagnosed with early or locally advanced BC treated with neoadjuvant chemotherapy was conducted. ALDH1A1, PD-L1 expression and TILs were assessed using immunohistochemistry. A total of 75 patients were analyzed (42.7% TN, 57.3% HER2+ tumors). ALDH1A1+ was related to HTILs (p = 0.005) and PD-L1+ tumors (p = 0.004). ALDH1A1+ tumors presented higher CD3+ (p = 0.008), CD4+ (p = 0.005), CD8+ (p = 0.003) and CD20+ (p = 0.006) TILs. ALDH1A1+ (p = 0.018), PD-L1+ (p = 0.004) and HTILs (p < 0.001) were related to smaller tumors. ALDH1A1+ was related to pathologic complete response (pCR) (p = 0.048). At the end of the follow-up (54.4 [38.3−87.6] months), 47 patients (62.7%) remained disease-free, and 20 (26.7%) had died. HTILs were related to improved disease-free survival (p = 0.027). ALDH1A1+ was related to PD-L1+ and HITLs, that might be related to higher pCR rates with neoadjuvant therapy.
Collapse
|
13
|
Osmond B, Facey COB, Zhang C, Boman BM. HOXA9 Overexpression Contributes to Stem Cell Overpopulation That Drives Development and Growth of Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23126799. [PMID: 35743243 PMCID: PMC9224160 DOI: 10.3390/ijms23126799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 01/22/2023] Open
Abstract
HOX proteins are transcription factors that regulate stem cell (SC) function, but their role in the SC origin of cancer is under-studied. Aberrant expression of HOX genes occurs in many cancer types. Our goal is to ascertain how retinoic acid (RA) signaling and the regulation of HOXA9 expression might play a role in the SC origin of human colorectal cancer (CRC). Previously, we reported that aldehyde dehydrogenase (ALDH) and other RA pathway components are co-expressed in colonic cancer SCs (CSCs) and that overpopulation of ALDH-positive CSCs occurs during colon tumorigenesis. Our hypothesis is RA signaling regulates HOXA9 expression, and dysregulated RA signaling results in HOXA9 overexpression, which contributes to CSC overpopulation in CRC. Immunostaining showed that HOXA9 was selectively expressed in ALDH-positive SCs, and HOXA9 expression was increased in CRCs compared to normal epithelium. Modulating RA signaling in CRC cells (HT29 and SW480) with ATRA and DEAB decreased cell proliferation and reduced HOXA9 expression. Bioinformatics analyses identified a network of proteins that functionally interact with HOXA9, and the genes that encode these proteins, as well as HOXA9, contain RA receptor binding sites. These findings indicate that the expression of HOXA9 and its functional network is regulated by RA signaling in normal colonic SCs, and, when dysregulated, HOXA9 may contribute to CSC overpopulation that drives CRC development and growth. Our study provides a regulatory mechanism that might be useful in developing treatments against CSC overpopulation in CRC.
Collapse
Affiliation(s)
- Brian Osmond
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (B.O.); (C.O.B.F.); (C.Z.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19713, USA
| | - Caroline O. B. Facey
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (B.O.); (C.O.B.F.); (C.Z.)
| | - Chi Zhang
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (B.O.); (C.O.B.F.); (C.Z.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19713, USA
| | - Bruce M. Boman
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (B.O.); (C.O.B.F.); (C.Z.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19713, USA
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Correspondence: ; Tel.: +1-267-303-9241
| |
Collapse
|
14
|
Ibrahim AIM, Batlle E, Sneha S, Jiménez R, Pequerul R, Parés X, Rüngeler T, Jha V, Tuccinardi T, Sadiq M, Frame F, Maitland NJ, Farrés J, Pors K. Expansion of the 4-(Diethylamino)benzaldehyde Scaffold to Explore the Impact on Aldehyde Dehydrogenase Activity and Antiproliferative Activity in Prostate Cancer. J Med Chem 2022; 65:3833-3848. [PMID: 35212533 PMCID: PMC9007462 DOI: 10.1021/acs.jmedchem.1c01367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
Aldehyde dehydrogenases (ALDHs) are
overexpressed in various tumor
types including prostate cancer and considered a potential target
for therapeutic intervention. 4-(Diethylamino)benzaldehyde (DEAB)
has been extensively reported as a pan-inhibitor of ALDH isoforms,
and here, we report on the synthesis, ALDH isoform selectivity, and
cellular potencies in prostate cancer cells of 40 DEAB analogues;
three analogues (14, 15, and 16) showed potent inhibitory activity against ALDH1A3, and two analogues
(18 and 19) showed potent inhibitory activity
against ALDH3A1. Significantly, 16 analogues displayed increased cytotoxicity
(IC50 = 10–200 μM) compared with DEAB (>200
μM) against three different prostate cancer cell lines. Analogues 14 and 18 were more potent than DEAB against
patient-derived primary prostate tumor epithelial cells, as single
agents or in combination treatment with docetaxel. In conclusion,
our study supports the use of DEAB as an ALDH inhibitor but also reveals
closely related analogues with increased selectivity and potency.
Collapse
Affiliation(s)
- Ali I M Ibrahim
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Yorkshire BD7 1DP, U.K.,Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Elisabet Batlle
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Yorkshire BD7 1DP, U.K.,Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona E-08193, Spain
| | - Smarakan Sneha
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Yorkshire BD7 1DP, U.K
| | - Rafael Jiménez
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona E-08193, Spain
| | - Raquel Pequerul
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona E-08193, Spain
| | - Xavier Parés
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona E-08193, Spain
| | - Till Rüngeler
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona E-08193, Spain
| | - Vibhu Jha
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Maria Sadiq
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Yorkshire BD7 1DP, U.K.,Cancer Research Unit, Department of Biology, University of York, Heslington, Yorkshire YO10 5DD, U.K
| | - Fiona Frame
- Cancer Research Unit, Department of Biology, University of York, Heslington, Yorkshire YO10 5DD, U.K
| | - Norman J Maitland
- Cancer Research Unit, Department of Biology, University of York, Heslington, Yorkshire YO10 5DD, U.K
| | - Jaume Farrés
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona E-08193, Spain
| | - Klaus Pors
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Yorkshire BD7 1DP, U.K
| |
Collapse
|
15
|
Dogan E, Kisim A, Bati-Ayaz G, Kubicek GJ, Pesen-Okvur D, Miri AK. Cancer Stem Cells in Tumor Modeling: Challenges and Future Directions. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100017. [PMID: 34927168 PMCID: PMC8680587 DOI: 10.1002/anbr.202100017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Microfluidic tumors-on-chips models have revolutionized anticancer therapeutic research by creating an ideal microenvironment for cancer cells. The tumor microenvironment (TME) includes various cell types and cancer stem cells (CSCs), which are postulated to regulate the growth, invasion, and migratory behavior of tumor cells. In this review, the biological niches of the TME and cancer cell behavior focusing on the behavior of CSCs are summarized. Conventional cancer models such as three-dimensional cultures and organoid models are reviewed. Opportunities for the incorporation of CSCs with tumors-on-chips are then discussed for creating tumor invasion models. Such models will represent a paradigm shift in the cancer community by allowing oncologists and clinicians to predict better which cancer patients will benefit from chemotherapy treatments.
Collapse
Affiliation(s)
- Elvan Dogan
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028
| | - Asli Kisim
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey
| | - Gizem Bati-Ayaz
- Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Gregory J. Kubicek
- Department of Radiation Oncology, MD Anderson Cancer Center at Cooper, 2 Cooper Plaza, Camden, NJ 08103
| | - Devrim Pesen-Okvur
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey; Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Amir K. Miri
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028; School of Medical Engineering, Science, and Health, Rowan University, Camden, NJ 08103
| |
Collapse
|
16
|
O.B. Facey C, M. Boman B. Retinoids in Treatment of Colorectal Cancer. COLORECTAL CANCER 2021. [DOI: 10.5772/intechopen.93699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Retinoids are vitamin A metabolites best known for their role in embryonic development. Indeed, retinoid acid (RA) signaling plays a key role in regulating the development of the embryo body-plan by controlling embryonic stem cells (SCs). Retinoids function through their ability to induce cellular differentiation. Mutations in RA signaling pathway genes occur in most human cancers. The classic example is the chromosomal translocation involving RA receptor alpha in acute promyelocytic leukemia (APL). Because all-trans retinoic acid (ATRA) is a highly effective and often curative treatment for APL patients, determining if retinoids are efficacious for other cancer types is imperative. We review the current research on retinoids in colorectal cancer (CRC) and provide bioinformatics analyses of RA signaling. Our results show that most RA pathway genes are overexpressed and often mutated in CRC. Moreover, aberrant expression of many RA signaling proteins predicts decreased CRC patient survival. We also review aldehyde dehydrogenase (ALDH) expression in CRC because ALDH is a key enzyme in RA signaling, which regulates colonic SCs. Further investigation of RA signaling mechanisms that regulate colon SCs and how dysregulation contributes to the SC overpopulation that drives CRC growth should provide insight into strategies for designing new SC-targeted therapies for CRC.
Collapse
|
17
|
Abu-Serie MM, Andrade F, Cámara-Sánchez P, Seras-Franzoso J, Rafael D, Díaz-Riascos ZV, Gener P, Abasolo I, Schwartz S. Pluronic F127 micelles improve the stability and enhance the anticancer stem cell efficacy of citral in breast cancer. Nanomedicine (Lond) 2021; 16:1471-1485. [PMID: 34160295 DOI: 10.2217/nnm-2021-0013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aim: Improving the stability and anti-cancer stem cell (CSC) activity of citral, a natural ALDH1A inhibitor. Materials & methods: Citral-loaded micelles (CLM) were obtained using Pluronic® F127 and its efficacy tested on the growth of four breast cancer cell lines. The impact of the CLM on the growth and functional hallmarks of breast CSCs were also evaluated using mammosphere and CSC reporter cell lines. Results: CLM improved the stability and growth inhibitory effects of citral. Importantly, CLM fully blocking the stemness features of CSCs (self-renewal, differentiation and migration) and in combination with paclitaxel CLM sensitized breast cancer cells to the chemotherapy. Conclusion: Targeting CSCs with CLM could improve the treatment of advanced breast cancer in combination with the standard chemotherapy.
Collapse
Affiliation(s)
- Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering, & Biotechnology Research Institute, City of Scientific Research & Technological Applications (SRTA-City), New Borg EL-Arab, 21934, Alexandria, Egypt
| | - Fernanda Andrade
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Patricia Cámara-Sánchez
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain.,Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Joaquin Seras-Franzoso
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Diana Rafael
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Zamira V Díaz-Riascos
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain.,Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Petra Gener
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Ibane Abasolo
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain.,Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Simó Schwartz
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| |
Collapse
|
18
|
Slepicka PF, Somasundara AVH, Dos Santos CO. The molecular basis of mammary gland development and epithelial differentiation. Semin Cell Dev Biol 2021; 114:93-112. [PMID: 33082117 PMCID: PMC8052380 DOI: 10.1016/j.semcdb.2020.09.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Our understanding of the molecular events underpinning the development of mammalian organ systems has been increasing rapidly in recent years. With the advent of new and improved next-generation sequencing methods, we are now able to dig deeper than ever before into the genomic and epigenomic events that play critical roles in determining the fates of stem and progenitor cells during the development of an embryo into an adult. In this review, we detail and discuss the genes and pathways that are involved in mammary gland development, from embryogenesis, through maturation into an adult gland, to the role of pregnancy signals in directing the terminal maturation of the mammary gland into a milk producing organ that can nurture the offspring. We also provide an overview of the latest research in the single-cell genomics of mammary gland development, which may help us to understand the lineage commitment of mammary stem cells (MaSCs) into luminal or basal epithelial cells that constitute the mammary gland. Finally, we summarize the use of 3D organoid cultures as a model system to study the molecular events during mammary gland development. Our increased investigation of the molecular requirements for normal mammary gland development will advance the discovery of targets to predict breast cancer risk and the development of new breast cancer therapies.
Collapse
Affiliation(s)
- Priscila Ferreira Slepicka
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | | | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
19
|
Ahmadi Z, Jena H, Singh M, Dhawan G, Kumar P. Self-Assembled Biodegradable Core-Shell Nanocomposites of Amphiphilic Retinoic Acid-LMW bPEI Conjugates Exhibit Enhanced Transgene Expression in Hepatocellular Carcinoma Cells With Inherent Anticancer Properties. J Pharm Sci 2021; 110:3047-3060. [PMID: 33933435 DOI: 10.1016/j.xphs.2021.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/02/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022]
Abstract
Low molecular weight branched polyethylenimines (LMW bPEIs) are almost nontoxic but display poor transfection efficiency due to lack of adequate complexation ability with nucleic acids followed by transportation across the cell membrane. Here, a series of amphiphilic retinoyl-bPEI conjugates (RP-1, RP-2 and RP-3) has been synthesized by allowing the reaction between bPEI (1.8 kDa) and a bioactive and hydrophobic vitamin A metabolite, all-trans-retinoic acid (ATRA), in varying amounts. In aqueous medium, these conjugates self-assembled into core-shell RP nanocomposites with size ranging from ~113-178 nm and zeta potential from ~ +15-35 mV. Evaluation of pDNA complexes of RP nanocomposites revealed that all the complexes exhibited significantly enhanced transfection efficiency without compromising on the cytocompatibility. RP-3/pDNA complex, with the highest content of retinoic acid, exhibited the best transfection efficiency. Further, due to anticancer properties of ATRA, these nanocomposites significantly reduced the viability of cancer cells (HepG2 and MCF-7 cells) without affecting the viability of non-cancerous cells (HEK 293 cells) demonstrating the cell-selective nature of the formulated nanocomposites. The intracellular trafficking and co-localization studies involving RP-3 nanocomposites also showed their higher uptake with intracellular and nuclear accumulation properties. Altogether, the results demonstrate the promising potential of the RP conjugates that can be used in future hepatocellular carcinoma targeted gene delivery applications.
Collapse
Affiliation(s)
- Zeba Ahmadi
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi-110007, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad- 201002, India
| | - Harekrushna Jena
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi-110007, India; Department of Biomedical Sciences, Acharya Narendra Dev College (University of Delhi), Govindpuri, Kalkaji, New Delhi- 110019, India
| | - Mahak Singh
- Department of Chemistry, Ramjas College (University of Delhi), University Enclave, Delhi-110007, India
| | - Gagan Dhawan
- Department of Biomedical Sciences, Acharya Narendra Dev College (University of Delhi), Govindpuri, Kalkaji, New Delhi- 110019, India.
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi-110007, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad- 201002, India.
| |
Collapse
|
20
|
All-trans retinoic acid and protein kinase C α/β1 inhibitor combined treatment targets cancer stem cells and impairs breast tumor progression. Sci Rep 2021; 11:6044. [PMID: 33723318 PMCID: PMC7961031 DOI: 10.1038/s41598-021-85344-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 02/28/2021] [Indexed: 01/08/2023] Open
Abstract
Breast cancer is the leading cause of cancer death among women worldwide. Blocking a single signaling pathway is often an ineffective therapy, especially in the case of aggressive or drug-resistant tumors. Since we have previously described the mechanism involved in the crosstalk between Retinoic Acid system and protein kinase C (PKC) pathway, the rationale of our study was to evaluate the effect of combining all-trans-retinoic acid (ATRA) with a classical PCK inhibitor (Gö6976) in preclinical settings. Employing hormone-independent mammary cancer models, Gö6976 and ATRA combined treatment induced a synergistic reduction in proliferative potential that correlated with an increased apoptosis and RARs modulation towards an anti-oncogenic profile. Combined treatment also impairs growth, self-renewal and clonogenicity potential of cancer stem cells and reduced tumor growth, metastatic spread and cancer stem cells frequency in vivo. An in-silico analysis of “Kaplan–Meier plotter” database indicated that low PKCα together with high RARα mRNA expression is a favorable prognosis factor for hormone-independent breast cancer patients. Here we demonstrate that a classical PKC inhibitor potentiates ATRA antitumor effects also targeting cancer stem cells growth, self-renewal and frequency.
Collapse
|
21
|
Koual M, Tomkiewicz C, Guerrera IC, Sherr D, Barouki R, Coumoul X. Aggressiveness and Metastatic Potential of Breast Cancer Cells Co-Cultured with Preadipocytes and Exposed to an Environmental Pollutant Dioxin: An in Vitro and in Vivo Zebrafish Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:37002. [PMID: 33683140 PMCID: PMC7939125 DOI: 10.1289/ehp7102] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 01/27/2021] [Accepted: 02/09/2021] [Indexed: 05/27/2023]
Abstract
BACKGROUND Breast cancer (BC) is a major public health concern, and its prognosis is very poor once metastasis occurs. The tumor microenvironment and chemical pollution have been suggested recently to contribute, independently, to the development of metastatic cells. The BC microenvironment consists, in part, of adipocytes and preadipocytes in which persistent organic pollutants (POPs) can be stored. OBJECTIVES We aimed to test the hypothesis that these two factors (2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an extensively studied, toxic POP and the microenvironment) may interact to increase tumor aggressiveness. METHODS We used a co-culture model using BC MCF-7 cells or MDA-MB-231 cells together with hMADS preadipocytes to investigate the contribution of the microenvironment and 2,3,7,8-tetrachlorodibenzo-p-dioxin TCDD on BC cells. Global differences were characterized using a high-throughput proteomic assay. Subsequently we measured the BC stem cell-like activity, analyzed the cell morphology, and used a zebrafish larvae model to study the metastatic potential of the BC cells. RESULTS We found that coexposure to TCDD and preadipocytes modified BC cell properties; moreover, it induced the expression of ALDH1A3, a cancer stem cell marker, and the appearance of giant cancer cells with cell-in-cell structures (CICs), which are associated with malignant metastatic progression, that we demonstrated in vivo. DISCUSSION The results of our study using BC cell lines co-cultured with preadipocytes and a POP and an in vivo zebrafish model of metastasis suggest that the interactions between BC cells and their microenvironment could affect their invasive or metastatic potential. https://doi.org/10.1289/EHP7102.
Collapse
Affiliation(s)
- Meriem Koual
- UMR-S1124, Institut national de la santé et de la recherché médicale (Inserm), T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, Paris, France
- Service de Chirurgie Cancérologique Gynécologique et du Sein, Hôpital Européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, France
- Université de Paris, Paris, France
| | - Céline Tomkiewicz
- UMR-S1124, Institut national de la santé et de la recherché médicale (Inserm), T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, Paris, France
| | - Ida Chiara Guerrera
- Plateforme protéomique 3P5-Necker, Structure Fédérative de Recherche Necker, Université de Paris, US24/CNRS UMS3633, Inserm, Paris, France
| | - David Sherr
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Robert Barouki
- UMR-S1124, Institut national de la santé et de la recherché médicale (Inserm), T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, Paris, France
- Université de Paris, Paris, France
| | - Xavier Coumoul
- UMR-S1124, Institut national de la santé et de la recherché médicale (Inserm), T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, Paris, France
- Université de Paris, Paris, France
| |
Collapse
|
22
|
Zheng Q, Zhang M, Zhou F, Zhang L, Meng X. The Breast Cancer Stem Cells Traits and Drug Resistance. Front Pharmacol 2021; 11:599965. [PMID: 33584277 PMCID: PMC7876385 DOI: 10.3389/fphar.2020.599965] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
Drug resistance is a major challenge in breast cancer (BC) treatment at present. Accumulating studies indicate that breast cancer stem cells (BCSCs) are responsible for the BC drugs resistance, causing relapse and metastasis in BC patients. Thus, BCSCs elimination could reverse drug resistance and improve drug efficacy to benefit BC patients. Consequently, mastering the knowledge on the proliferation, resistance mechanisms, and separation of BCSCs in BC therapy is extremely helpful for BCSCs-targeted therapeutic strategies. Herein, we summarize the principal BCSCs surface markers and signaling pathways, and list the BCSCs-related drug resistance mechanisms in chemotherapy (CT), endocrine therapy (ET), and targeted therapy (TT), and display therapeutic strategies for targeting BCSCs to reverse drug resistance in BC. Even more importantly, more attention should be paid to studies on BCSC-targeted strategies to overcome the drug resistant dilemma of clinical therapies in the future.
Collapse
Affiliation(s)
- Qinghui Zheng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Mengdi Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xuli Meng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
23
|
Identification of significantly mutated subnetworks in the breast cancer genome. Sci Rep 2021; 11:642. [PMID: 33436820 PMCID: PMC7804148 DOI: 10.1038/s41598-020-80204-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/17/2020] [Indexed: 11/24/2022] Open
Abstract
Recent studies showed that somatic cancer mutations target genes that are in specific signaling and cellular pathways. However, in each patient only a few of the pathway genes are mutated. Current approaches consider only existing pathways and ignore the topology of the pathways. For this reason, new efforts have been focused on identifying significantly mutated subnetworks and associating them with cancer characteristics. We applied two well-established network analysis approaches to identify significantly mutated subnetworks in the breast cancer genome. We took network topology into account for measuring the mutation similarity of a gene-pair to allow us to infer the significantly mutated subnetworks. Our goals are to evaluate whether the identified subnetworks can be used as biomarkers for predicting breast cancer patient survival and provide the potential mechanisms of the pathways enriched in the subnetworks, with the aim of improving breast cancer treatment. Using the copy number alteration (CNA) datasets from the METABRIC (Molecular Taxonomy of Breast Cancer International Consortium) study, we identified a significantly mutated yet clinically and functionally relevant subnetwork using two graph-based clustering algorithms. The mutational pattern of the subnetwork is significantly associated with breast cancer survival. The genes in the subnetwork are significantly enriched in retinol metabolism KEGG pathway. Our results show that breast cancer treatment with retinoids may be a potential personalized therapy for breast cancer patients since the CNA patterns of the breast cancer patients can imply whether the retinoids pathway is altered. We also showed that applying multiple bioinformatics algorithms at the same time has the potential to identify new network-based biomarkers, which may be useful for stratifying cancer patients for choosing optimal treatments.
Collapse
|
24
|
Aldehyde Dehydrogenase 1B1 Is Associated with Altered Cell Morphology, Proliferation, Migration and Chemosensitivity in Human Colorectal Adenocarcinoma Cells. Biomedicines 2021; 9:biomedicines9010044. [PMID: 33419031 PMCID: PMC7825346 DOI: 10.3390/biomedicines9010044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 12/28/2022] Open
Abstract
Aldehyde dehydrogenases (ALDHs) are NAD(P)+-dependent enzymes that catalyze the oxidation of endogenous and exogenous aldehydes to their corresponding carboxylic acids. ALDHs participate in a variety of cellular mechanisms, such as metabolism, cell proliferation and apoptosis, as well as differentiation and stemness. Over the last few years, ALDHs have emerged as cancer stem cell markers in a wide spectrum of solid tumors and hematological malignancies. In this study, the pathophysiological role of ALDH1B1 in human colorectal adenocarcinoma was investigated. Human colon cancer HT29 cells were stably transfected either with human green fluorescent protein (GFP)-tagged ALDH1B1 or with an empty lentiviral expression vector. The overexpression of ALDH1B1 was correlated with altered cell morphology, decreased proliferation rate and reduced clonogenic efficiency. Additionally, ALDH1B1 triggered a G2/M arrest at 24 h post-cell synchronization, probably through p53 and p21 upregulation. Furthermore, ALDH1B1-overexpressing HT29 cells exhibited enhanced resistance against doxorubicin, fluorouracil (5-FU) and etoposide. Finally, ALDH1B1 induced increased migratory potential and displayed epithelial–mesenchymal transition (EMT) through the upregulation of ZEB1 and vimentin and the consequent downregulation of E-cadherin. Taken together, ALDH1B1 confers alterations in the cell morphology, cell cycle progression and gene expression, accompanied by significant changes in the chemosensitivity and migratory potential of HT29 cells, underlying its potential significance in cancer progression.
Collapse
|
25
|
Soltanian S, Sheikhbahaei M. Effect of Menadione and Combination of Gemcitabine and Cisplatin on Cancer Stem Cells in Human Non-small Cell Lung Cancer (NSCLC) Cell Line A549. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:105-117. [PMID: 34400945 PMCID: PMC8170754 DOI: 10.22037/ijpr.2020.112373.13715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. Chemotherapy-induced adverse effects and resistance of NSCLC to conventional drugs reduce the efficacy of current therapies. Tumors contain a small population of cancer stem cells (CSCs) that play a critical role in tumor initiation, maintenance, and drug resistance that finally lead to cancer recurrence. Therefore, CSC-targeting therapies can offer the best hope for developing curative cancer therapies. Vitamins have a high potential for cancer prevention and treatment. Vitamins also ameliorate the side effects which occur in chemo-radio therapy. Menadione (2-methyl-1,4-naphthoquinone/vitamin-K3) is a synthetic form of vitamin K that indicated antitumor activities. The purpose of this study was to evaluate the anti-CSCs effect of menadione and combination of cisplatin and gemcitabine as a first-line treatment in patients with NSCLC on the NSCLC cell line A549. MTT results displayed decreased cell survival after treatment with cisplatin/gemcitabine for 48 h treatment (IC50 values 0.25 µM for cisplatin and 5 µM for gemcitabine). Menadione also inhibited the cell growth in A549 cells (IC50: 16 µM). Quantitative RT-PCR showed significant downregulation of CSC markers (Oct4, Nanog, Sox2, Aldh1, Abcb1, CD44, and CD133) and Snail, epithelial-mesenchymal transition marker, after treatment with menadione and cisplatin/gemcitabine. Flow cytometry showed CD44-positive cells that constitute a high percentage (70%) of A549 cells reduced significantly after treatment with cisplatin/gemcitabine or menadione. However, A549 cells did not show a significant population positive for CD133 and ABCB1 (less than 0.05%), and these fractions did not change after treatment with two agents.
Collapse
|
26
|
Shen S, Xu X, Lin S, Zhang Y, Liu H, Zhang C, Mo R. A nanotherapeutic strategy to overcome chemotherapeutic resistance of cancer stem-like cells. NATURE NANOTECHNOLOGY 2021; 16:104-113. [PMID: 33437035 DOI: 10.1038/s41565-020-00793-0] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 10/13/2020] [Indexed: 05/06/2023]
Abstract
Tumour heterogeneity remains a major challenge in cancer therapy owing to the different susceptibility of cells to chemotherapy within a solid tumour. Cancer stem-like cells (CSCs), which reside in hypoxic tumour regions, are characterized by high tumourigenicity and chemoresistance and are often responsible for tumour progression and recurrence. Here we report a nanotherapeutic strategy to kill CSCs in tumours using nanoparticles that are co-loaded with the differentiation-inducing agent, all-trans retinoic acid, and the chemotherapeutic drug, camptothecin. All-trans retinoic acid is released under hypoxic conditions, leading to CSC differentiation in the hypoxic niche. In differentiating CSC, the reactive oxygen species levels increase, which then causes the release of camptothecin and subsequent cell death. This dual strategy enables controlled drug release in CSCs and reduces stemness-related drug resistance, enhancing the chemotherapeutic response. In breast tumour mouse models, treatment with the nanoparticles suppresses tumour growth and prevents post-surgical tumour relapse and metastasis.
Collapse
Affiliation(s)
- Shiyang Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China
| | - Xiao Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China
| | - Shiqi Lin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China
| | - Huayu Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
27
|
He L, Yu A, Deng L, Zhang H. Eradicating the Roots: Advanced Therapeutic Approaches Targeting Breast Cancer Stem Cells. Curr Pharm Des 2020; 26:2009-2021. [PMID: 32183663 DOI: 10.2174/1381612826666200317132949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/08/2020] [Indexed: 12/30/2022]
Abstract
Accumulating evidences have demonstrated that the existence of breast cancer-initiating cells, which drives the original tumorigenicity, local invasion and migration propensity of breast cancer. These cells, termed as breast cancer stem cells (BCSCs), possess properties including self-renewal, multidirectional differentiation and proliferative potential, and are believed to play important roles in the intrinsic drug resistance of breast cancer. One of the reasons why BCBCs cause difficulties in breast cancer treating is that BCBCs can control both genetic and non-genetic elements to keep their niches safe and sound, which allows BCSCs for constant self-renewal and differentiation. Therapeutic strategies designed to target BCSCs may ultimately result in effective interventions for the treatment of breast cancer. Novel strategies including nanomedicine, oncolytic virus therapy, immunotherapy and induced differentiation therapy are emerging and proved to be efficient in anti-BCSCs therapy. In this review, we summarized breast tumor biology and the current challenges of breast cancer therapies, focused on breast cancer stem cells, and introduced promising therapeutic strategies targeting BCSCs.
Collapse
Affiliation(s)
- Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Anran Yu
- The State University of New York, Buffalo, NY 12246, United States
| | - Li Deng
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hongwei Zhang
- School of Pharmacy, MCPHS University, Boston, MA 02115, United States
| |
Collapse
|
28
|
Ávalos-Moreno M, López-Tejada A, Blaya-Cánovas JL, Cara-Lupiañez FE, González-González A, Lorente JA, Sánchez-Rovira P, Granados-Principal S. Drug Repurposing for Triple-Negative Breast Cancer. J Pers Med 2020; 10:E200. [PMID: 33138097 PMCID: PMC7711505 DOI: 10.3390/jpm10040200] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer which presents a high rate of relapse, metastasis, and mortality. Nowadays, the absence of approved specific targeted therapies to eradicate TNBC remains one of the main challenges in clinical practice. Drug discovery is a long and costly process that can be dramatically improved by drug repurposing, which identifies new uses for existing drugs, both approved and investigational. Drug repositioning benefits from improvements in computational methods related to chemoinformatics, genomics, and systems biology. To the best of our knowledge, we propose a novel and inclusive classification of those approaches whereby drug repurposing can be achieved in silico: structure-based, transcriptional signatures-based, biological networks-based, and data-mining-based drug repositioning. This review specially emphasizes the most relevant research, both at preclinical and clinical settings, aimed at repurposing pre-existing drugs to treat TNBC on the basis of molecular mechanisms and signaling pathways such as androgen receptor, adrenergic receptor, STAT3, nitric oxide synthase, or AXL. Finally, because of the ability and relevance of cancer stem cells (CSCs) to drive tumor aggressiveness and poor clinical outcome, we also focus on those molecules repurposed to specifically target this cell population to tackle recurrence and metastases associated with the progression of TNBC.
Collapse
Affiliation(s)
- Marta Ávalos-Moreno
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Francisca E. Cara-Lupiañez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Adrián González-González
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Jose A. Lorente
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- Department of Legal Medicine, School of Medicine—PTS—University of Granada, 18016 Granada, Spain
| | | | - Sergio Granados-Principal
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| |
Collapse
|
29
|
Zhang T, Ahn K, Emerick B, Modarai SR, Opdenaker LM, Palazzo J, Schleiniger G, Fields JZ, Boman BM. APC mutations in human colon lead to decreased neuroendocrine maturation of ALDH+ stem cells that alters GLP-2 and SST feedback signaling: Clue to a link between WNT and retinoic acid signalling in colon cancer development. PLoS One 2020; 15:e0239601. [PMID: 33112876 PMCID: PMC7592776 DOI: 10.1371/journal.pone.0239601] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
APC mutations drive human colorectal cancer (CRC) development. A major contributing factor is colonic stem cell (SC) overpopulation. But, the mechanism has not been fully identified. A possible mechanism is the dysregulation of neuroendocrine cell (NEC) maturation by APC mutations because SCs and NECs both reside together in the colonic crypt SC niche where SCs mature into NECs. So, we hypothesized that sequential inactivation of APC alleles in human colonic crypts leads to progressively delayed maturation of SCs into NECs and overpopulation of SCs. Accordingly, we used quantitative immunohistochemical mapping to measure indices and proportions of SCs and NECs in human colon tissues (normal, adenomatous, malignant), which have different APC-zygosity states. In normal crypts, many cells staining for the colonic SC marker ALDH1 co-stained for chromogranin-A (CGA) and other NEC markers. In contrast, in APC-mutant tissues from familial adenomatous polyposis (FAP) patients, the proportion of ALDH+ SCs progressively increased while NECs markedly decreased. To explain how these cell populations change in FAP tissues, we used mathematical modelling to identify kinetic mechanisms. Computational analyses indicated that APC mutations lead to: 1) decreased maturation of ALDH+ SCs into progenitor NECs (not progenitor NECs into mature NECs); 2) diminished feedback signaling by mature NECs. Biological experiments using human CRC cell lines to test model predictions showed that mature GLP-2R+ and SSTR1+ NECs produce, via their signaling peptides, opposing effects on rates of NEC maturation via feedback regulation of progenitor NECs. However, decrease in this feedback signaling wouldn't explain the delayed maturation because both progenitor and mature NECs are depleted in CRCs. So the mechanism for delayed maturation must explain how APC mutation causes the ALDH+ SCs to remain immature. Given that ALDH is a key component of the retinoic acid (RA) signaling pathway, that other components of the RA pathway are selectively expressed in ALDH+ SCs, and that exogenous RA ligands can induce ALDH+ cancer SCs to mature into NECs, RA signaling must be attenuated in ALDH+ SCs in CRC. Thus, attenuation of RA signaling explains why ALDH+ SCs remain immature in APC mutant tissues. Since APC mutation causes increased WNT signaling in FAP and we found that sequential inactivation of APC in FAP patient tissues leads to progressively delayed maturation of colonic ALDH+ SCs, the hypothesis is developed that human CRC evolves due to an imbalance between WNT and RA signaling.
Collapse
Affiliation(s)
- Tao Zhang
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
- Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Koree Ahn
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
- Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Brooks Emerick
- Center for Applications of Mathematics in Medicine, Department of Mathematical Sciences, University of Delaware, Newark, DE, United States of America
| | - Shirin R. Modarai
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
| | - Lynn M. Opdenaker
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
| | - Juan Palazzo
- Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Gilberto Schleiniger
- Center for Applications of Mathematics in Medicine, Department of Mathematical Sciences, University of Delaware, Newark, DE, United States of America
| | | | - Bruce M. Boman
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
- Thomas Jefferson University, Philadelphia, PA, United States of America
- Center for Applications of Mathematics in Medicine, Department of Mathematical Sciences, University of Delaware, Newark, DE, United States of America
| |
Collapse
|
30
|
Soltanian S, Sheikhbahaei M, Ziasistani M. Phytol Down-Regulates Expression of Some Cancer Stem Cell Markers and Decreases Side Population Proportion in Human Embryonic Carcinoma NCCIT Cells. Nutr Cancer 2020; 73:1520-1533. [PMID: 32700607 DOI: 10.1080/01635581.2020.1795695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cancer stem cells (CSCs), a subgroup of cancer cells, have self-renewal capacity and differentiation potential and drive tumor growth. CSCs are highly-resistant to conventional chemo-radio therapy. Phytochemicals were shown to be able to eliminate CSCs. Phytol is a diterpene alcohol with demonstrated anticancer effects. The current study compared the effect of phytol with retinoic acid (RA) as a well-known inducers of CSC differentiation and cisplatin, a common chemotherapy drug, on CSC markers in human embryonic carcinoma NCCIT cells. NCCIT cells were exposed to 10 mM RA for 14 day to induce differentiation. Moreover, NCCIT cells were treated with IC50 dose of cisplatin (12 µM) and phytol (40 µM) for 7 day. Real-time PCR showed that phytol was more effective that RA and cisplatin in down-regulating the CSC markers OCT4, NANOG, SOX2, ALDH1, ABCB1, CD44 and CD133. Percentage of SP (13%) and ABCB1+ (0.34%) in NCCIT cells decreased to 7% and 0.1% respectively after treatment with phytol. A very small proportion of NCCIT cells were positive for CD44 (0.2%) and CD133 (0.48%) and this fraction did not change significantly after treatment with three agents. In conclusion, phytol has the greatest inhibitory effect on CSC population and markers than RA and cisplatin.
Collapse
Affiliation(s)
- Sara Soltanian
- Department of Biology, Faculty of Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mahboubeh Sheikhbahaei
- Department of Biology, Faculty of Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mahsa Ziasistani
- Pathology and Stem Cell Research Center, Afzalipour Medical School, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
31
|
Wu J, Mamidi TKK, Zhang L, Hicks C. Unraveling the Genomic-Epigenomic Interaction Landscape in Triple Negative and Non-Triple Negative Breast Cancer. Cancers (Basel) 2020; 12:cancers12061559. [PMID: 32545594 PMCID: PMC7352267 DOI: 10.3390/cancers12061559] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 01/01/2023] Open
Abstract
Background: The recent surge of next generation sequencing of breast cancer genomes has enabled development of comprehensive catalogues of somatic mutations and expanded the molecular classification of subtypes of breast cancer. However, somatic mutations and gene expression data have not been leveraged and integrated with epigenomic data to unravel the genomic-epigenomic interaction landscape of triple negative breast cancer (TNBC) and non-triple negative breast cancer (non-TNBC). Methods: We performed integrative data analysis combining somatic mutation, epigenomic and gene expression data from The Cancer Genome Atlas (TCGA) to unravel the possible oncogenic interactions between genomic and epigenomic variation in TNBC and non-TNBC. We hypothesized that within breast cancers, there are differences in somatic mutation, DNA methylation and gene expression signatures between TNBC and non-TNBC. We further hypothesized that genomic and epigenomic alterations affect gene regulatory networks and signaling pathways driving the two types of breast cancer. Results: The investigation revealed somatic mutated, epigenomic and gene expression signatures unique to TNBC and non-TNBC and signatures distinguishing the two types of breast cancer. In addition, the investigation revealed molecular networks and signaling pathways enriched for somatic mutations and epigenomic changes unique to each type of breast cancer. The most significant pathways for TNBC were: retinal biosynthesis, BAG2, LXR/RXR, EIF2 and P2Y purigenic receptor signaling pathways. The most significant pathways for non-TNBC were: UVB-induced MAPK, PCP, Apelin endothelial, Endoplasmatic reticulum stress and mechanisms of viral exit from host signaling Pathways. Conclusion: The investigation revealed integrated genomic, epigenomic and gene expression signatures and signing pathways unique to TNBC and non-TNBC, and a gene signature distinguishing the two types of breast cancer. The study demonstrates that integrative analysis of multi-omics data is a powerful approach for unravelling the genomic-epigenomic interaction landscape in TNBC and non-TNBC.
Collapse
Affiliation(s)
- Jiande Wu
- Health Sciences Center, Department of Genetic, Louisiana State University School of Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA;
| | - Tarun Karthik Kumar Mamidi
- Center for Computational Genomics and Data Science, Departments of Pediatrics and Pathology, University of Alabama–Birmingham School of Medicine, Birmingham, AL 35233, USA;
| | - Lu Zhang
- Department of Public Health Sciences, Clemson University, 513 Edwards Hall, Clemson, SC 29634, USA;
| | - Chindo Hicks
- Health Sciences Center, Department of Genetic, Louisiana State University School of Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA;
- Correspondence: ; Tel.: +1-504-568-2657
| |
Collapse
|
32
|
Li C, Han X. Co-delivery of Dacarbazine and All-Trans Retinoic Acid (ATRA) Using Lipid Nanoformulations for Synergistic Antitumor Efficacy Against Malignant Melanoma. NANOSCALE RESEARCH LETTERS 2020; 15:113. [PMID: 32430641 PMCID: PMC7237551 DOI: 10.1186/s11671-020-3293-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/04/2020] [Indexed: 06/10/2023]
Abstract
Malignant melanoma is a highly aggressive skin cancer responsible for 80% of mortality, and the overall median survival in patients with metastatic melanoma is only 6-9 months. Combination treatment through the simultaneous administration of dual drugs in a single nanocarrier has been demonstrated to be elegant and effective in combatting cancer. Herein, we employ a combination therapy based on dacarbazine (DBZ), FDA approved drug for melanoma and all-trans retinoic acid (ATRA), promising anticancer agents loaded on lipid nanoformulations (RD-LNF) as a new treatment strategy for malignant melanoma. We have successfully encapsulated both the drugs in lipid nanoformulations and showed a controlled release of payload over time. We demonstrated that the simultaneous delivery of DBZ and ATRA could effectively reduce cell proliferation in a concentration-dependent manner. The combinational nanoparticles significantly reduced the colony formation ability of B16F10 melanoma cells. Flow cytometer analysis showed that RD-LNF induced a greater proportion of apoptosis cells with significant inhibition of cell cycle progression and cell migration. These results suggest the promising potential of RD-LNF in the treatment of malignant melanoma with high efficacy.
Collapse
Affiliation(s)
- Chenyang Li
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Xiuping Han
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
33
|
Lin H, Liu S, Gao W, Liu H. DDIT3 modulates cancer stemness in gastric cancer by directly regulating CEBPβ. J Pharm Pharmacol 2020; 72:807-815. [PMID: 32189359 DOI: 10.1111/jphp.13243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 02/09/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Cancer stem cells (CSCs) have been identified to correlate with the initiation and metastasis of tumours, and DNA damage-inducible transcript 3 (DDIT3) is associated with the poor prognosis in gastric cancer (GC). However, whether DDIT3 mediates CSCs stemness in GC is still unclear. METHODS Microarray analysis and Gene Ontology (GO) were conducted to identify the differentially expressed genes in GC tissues from GC patients. The interaction between DDIT3 and CEBPβ was determined using immunoprecipitation (IP) analysis. KEY FINDINGS Herein, microarray analysis showed that DDIT3 expression is increased in GC tissues. qRT-PCR confirmed that DDIT3 is significantly increased in GC tissues and cancer cell lines compared with healthy tissues and normal cell lines, individually. Genetic overexpression of DDIT3 enhanced GC cell proliferation, colony-forming ability, sphere formation and CSCs stemness. Mechanistically, DDIT3 directly up-regulated the expression of transcription factor CEBPβ, leading to the increased expression of CSCs markers SOX2, NANOG, OCT4 and CD133 in gastric CSCs. Genetic downregulation of CEBPβ significantly abolishes DDIT3-mediated increased cell proliferation, colony-forming ability, sphere formation and CSCs stemness. CONCLUSION Our results demonstrated that DDIT3 promotes CSCs stemness by up-regulating CEBPβ in GC that provides novel targets for the further GC therapy.
Collapse
Affiliation(s)
- Hai Lin
- Department of digestive medicine, Linyi Central Hospital, Linyi, Shandong, China
| | - Shufang Liu
- Department of Laboratory, Linyi Central Hospital, Linyi, Shandong, China
| | - Weidong Gao
- Department of Gastroenterology, Lianshui County People's Hospital, The Affiliated Lianshui County People's Hospital of Kangda College of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Hongyu Liu
- Department of Pathology, The First Hospital of Qiqihar, The Affiliated Qiqihar Hospital of Southern Medical University, Qiqihar, Heilongjiang, China
| |
Collapse
|
34
|
Devalaraja S, To TKJ, Folkert IW, Natesan R, Alam MZ, Li M, Tada Y, Budagyan K, Dang MT, Zhai L, Lobel GP, Ciotti GE, Eisinger-Mathason TSK, Asangani IA, Weber K, Simon MC, Haldar M. Tumor-Derived Retinoic Acid Regulates Intratumoral Monocyte Differentiation to Promote Immune Suppression. Cell 2020; 180:1098-1114.e16. [PMID: 32169218 DOI: 10.1016/j.cell.2020.02.042] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/13/2019] [Accepted: 02/19/2020] [Indexed: 12/13/2022]
Abstract
The immunosuppressive tumor microenvironment (TME) is a major barrier to immunotherapy. Within solid tumors, why monocytes preferentially differentiate into immunosuppressive tumor-associated macrophages (TAMs) rather than immunostimulatory dendritic cells (DCs) remains unclear. Using multiple murine sarcoma models, we find that the TME induces tumor cells to produce retinoic acid (RA), which polarizes intratumoral monocyte differentiation toward TAMs and away from DCs via suppression of DC-promoting transcription factor Irf4. Genetic inhibition of RA production in tumor cells or pharmacologic inhibition of RA signaling within TME increases stimulatory monocyte-derived cells, enhances T cell-dependent anti-tumor immunity, and synergizes with immune checkpoint blockade. Furthermore, an RA-responsive gene signature in human monocytes correlates with an immunosuppressive TME in multiple human tumors. RA has been considered as an anti-cancer agent, whereas our work demonstrates its tumorigenic capability via myeloid-mediated immune suppression and provides proof of concept for targeting this pathway for tumor immunotherapy.
Collapse
Affiliation(s)
- Samir Devalaraja
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tsun Ki Jerrick To
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian W Folkert
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ramakrishnan Natesan
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Md Zahidul Alam
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Minghong Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Yuma Tada
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Konstantin Budagyan
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Mai T Dang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Li Zhai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Graham P Lobel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gabrielle E Ciotti
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - T S Karin Eisinger-Mathason
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Irfan A Asangani
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristy Weber
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malay Haldar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Xu A, Zhang N, Cao J, Zhu H, Yang B, He Q, Shao X, Ying M. Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation. Biochem Pharmacol 2020; 171:113696. [DOI: 10.1016/j.bcp.2019.113696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/05/2019] [Indexed: 12/22/2022]
|
36
|
Tomaselli D, Lucidi A, Rotili D, Mai A. Epigenetic polypharmacology: A new frontier for epi-drug discovery. Med Res Rev 2020; 40:190-244. [PMID: 31218726 PMCID: PMC6917854 DOI: 10.1002/med.21600] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022]
Abstract
Recently, despite the great success achieved by the so-called "magic bullets" in the treatment of different diseases through a marked and specific interaction with the target of interest, the pharmacological research is moving toward the development of "molecular network active compounds," embracing the related polypharmacology approach. This strategy was born to overcome the main limitations of the single target therapy leading to a superior therapeutic effect, a decrease of adverse reactions, and a reduction of potential mechanism(s) of drug resistance caused by robustness and redundancy of biological pathways. It has become clear that multifactorial diseases such as cancer, neurological, and inflammatory disorders, may require more complex therapeutic approaches hitting a certain biological system as a whole. Concerning epigenetics, the goal of the multi-epi-target approach consists in the development of small molecules able to simultaneously and (often) reversibly bind different specific epi-targets. To date, two dual histone deacetylase/kinase inhibitors (CUDC-101 and CUDC-907) are in an advanced stage of clinical trials. In the last years, the growing interest in polypharmacology encouraged the publication of high-quality reviews on combination therapy and hybrid molecules. Hence, to update the state-of-the-art of these therapeutic approaches avoiding redundancy, herein we focused only on multiple medication therapies and multitargeting compounds exploiting epigenetic plus nonepigenetic drugs reported in the literature in 2018. In addition, all the multi-epi-target inhibitors known in literature so far, hitting two or more epigenetic targets, have been included.
Collapse
Affiliation(s)
- Daniela Tomaselli
- Department of Chemistry and Technologies of Drugs,
“Sapienza” University of Rome, P.le A. Moro 5, 00185 Roma, Italy
| | - Alessia Lucidi
- Department of Chemistry and Technologies of Drugs,
“Sapienza” University of Rome, P.le A. Moro 5, 00185 Roma, Italy
| | - Dante Rotili
- Department of Chemistry and Technologies of Drugs,
“Sapienza” University of Rome, P.le A. Moro 5, 00185 Roma, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs,
“Sapienza” University of Rome, P.le A. Moro 5, 00185 Roma, Italy
- Pasteur Institute - Cenci Bolognetti Foundation, Viale
Regina Elena 291, 00161 Roma, Italy
| |
Collapse
|
37
|
The Effect of Arsenic Trioxide on All-trans Retinoic Acid Binding to Human Serum Albumin. J Fluoresc 2019; 29:1277-1283. [DOI: 10.1007/s10895-019-02458-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/31/2019] [Indexed: 01/30/2023]
|
38
|
Sun HR, Wang S, Yan SC, Zhang Y, Nelson PJ, Jia HL, Qin LX, Dong QZ. Therapeutic Strategies Targeting Cancer Stem Cells and Their Microenvironment. Front Oncol 2019; 9:1104. [PMID: 31709180 PMCID: PMC6821685 DOI: 10.3389/fonc.2019.01104] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) have been demonstrated in a variety of tumors and are thought to act as a clonogenic core for the genesis of new tumor growth. This small subpopulation of cancer cells has been proposed to help drive tumorigenesis, metastasis, recurrence and conventional therapy resistance. CSCs show self-renewal and flexible clonogenic properties and help define specific tumor microenvironments (TME). The interaction between CSCs and TME is thought to function as a dynamic support system that fosters the generation and maintenance of CSCs. Investigation of the interaction between CSCs and the TME is shedding light on the biologic mechanisms underlying the process of tumor malignancy, metastasis, and therapy resistance. We summarize recent advances in CSC biology and their environment, and discuss the challenges and future strategies for targeting this biology as a new therapeutic approach.
Collapse
Affiliation(s)
- Hao-Ran Sun
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shun Wang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shi-Can Yan
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Zhang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter J Nelson
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Hu-Liang Jia
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiong-Zhu Dong
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
39
|
Two Opposing Faces of Retinoic Acid: Induction of Stemness or Induction of Differentiation Depending on Cell-Type. Biomolecules 2019; 9:biom9100567. [PMID: 31590252 PMCID: PMC6843238 DOI: 10.3390/biom9100567] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Stem cells have the capacity of self-renewal and, through proliferation and differentiation, are responsible for the embryonic development, postnatal development, and the regeneration of tissues in the adult organism. Cancer stem cells, analogous to the physiological stem cells, have the capacity of self-renewal and may account for growth and recurrence of tumors. Development and regeneration of healthy tissues and tumors depend on the balance of different genomic and nongenomic signaling pathways that regulate stem cell quiescence, proliferation, and differentiation. During evolution, this balance became dependent on all-trans retinoic acid (RA), a molecule derived from the environmental factor vitamin A. Here we summarize some recent findings on the prominent role of RA on the proliferation of stem and progenitor cells, in addition to its well-known function as an inductor of cell differentiation. A better understanding of the regulatory mechanisms of stemness and cell differentiation by RA may improve the therapeutic options of this molecule in regenerative medicine and cancer.
Collapse
|
40
|
Novohradsky V, Markova L, Kostrhunova H, Trávníček Z, Brabec V, Kasparkova J. An anticancer Os(II) bathophenanthroline complex as a human breast cancer stem cell-selective, mammosphere potent agent that kills cells by necroptosis. Sci Rep 2019; 9:13327. [PMID: 31527683 PMCID: PMC6746710 DOI: 10.1038/s41598-019-49774-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Conventional chemotherapy is mostly effective in the treatment of rapidly-dividing differentiated tumor cells but has limited application toward eliminating cancer stem cell (CSC) population. The presence of a very small number of CSCs may contribute to the development of therapeutic resistance, metastases, and relapse. Thus, treatment failure by developing novel anticancer drugs capable of effective targeting of CSCs is at present a major challenge for research focused on chemotherapy of cancer. Here, we show that Os(II) complex 2 [Os(η6-pcym)(bphen)(dca)]PF6 (pcym = p-cymene, bphen = bathophenanthroline, and dca = dichloroacetate), is capable of efficient and selective killing CSCs in heterogeneous populations of human breast cancer cells MCF-7 and SKBR-3. Notably, its remarkable submicromolar potency to kill CSCs is considerably higher than that of its Ru analog, [Ru(η6-pcym)(bphen)(dca)]PF6 (complex 1) and salinomycin, one of the most selective CSC-targeting compounds hitherto identified. Furthermore, Os(II) complex 2 reduces the formation, size, and viability of three-dimensional mammospheres which more closely reflect the tumor microenvironment than cells in traditional two-dimensional cultures. The antiproliferation studies and propidium iodide staining using flow cytometry suggest that Os(II) complex 2 induces human breast cancer stem cell death predominantly by necroptosis, a programmed form of necrosis. The results of this study demonstrate the promise of Os(II) complex 2 in treating human breast tumors. They also represent the foundation for further preclinical and clinical studies and applications of Os(II) complex 2 to comply with the emergent need for human breast CSCs-specific chemotherapeutics capable to treat chemotherapy-resistant and relapsed human breast tumors.
Collapse
Affiliation(s)
- Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Lenka Markova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Zdeněk Trávníček
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Slechtitelu 27, 783 71, Olomouc, Czech Republic
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 612 65, Brno, Czech Republic.,Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71, Olomouc, Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 612 65, Brno, Czech Republic.
| |
Collapse
|
41
|
Shah S, Pocard M, Mirshahi M. Targeting the differentiation of gastric cancer cells (KATO‑III) downregulates epithelial‑mesenchymal and cancer stem cell markers. Oncol Rep 2019; 42:670-678. [PMID: 31233198 PMCID: PMC6609315 DOI: 10.3892/or.2019.7198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 05/17/2019] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to analyze the acquisition of the differentiated phenotype in the human gastric signet ring cell adenoma cancer KATO-III cell line in vitro. The morphology of KATO-III cells was explored by microcinematography. Different cytokines secreted by both adherent and non-adherent KATO-III cells into medium were observed. The cancer stem cell phenotypes were identified by reverse transcription-quantitative polymerase chain reaction using primers (E-Cad, Slug, Snail, vimentin, NANOG, NESTIN, OCT3/4 and C-X-C motif chemokine receptor 4) or antibodies [cluster of differentiation (CD)90 and CD117] by flow cytometry (FACS). The influence of the induction media for the differentiation of mesenchymal cells was studied through viability and proliferation assays, by evaluating gene expression and the expression of markers via FACS. Cell viability and cell cycle distribution were evaluated following the treatment of KATO-III with acetyl salicylic acid and using the induction media as an inhibitor of epithelial-mesenchymal transition (EMT) and heparanase. A total of 3 phenotypes of KATO-III were observed (adherent, non-adherent and cell cluster), which have internal potential for cell transition into one of the other phenotypes. KATO-III was differentiated into adipocyte-, chondrocyte-, osteocyte- and neurocyte-like cells by the induction media. Identification of the induced cells was conducted using cell dyes. Reduced mRNA expression of EMT-associated molecules, stem cell markers and heparanase was observed with acetyl salicylic acid and induction media. An inhibitory effect of acetyl salicylic acid and the induction media was also noted in regard to cell proliferation. In addition, acetyl salicylic acid induced G0/G1 phase cell cycle arrest in KATO-III cells. In conclusion, the induction of the differentiation of cancer stem cells into non-proliferating cells offers the possibility for novel drug design to overcome the issues associated with metastasis, drug resistance and systemic toxicity with improved therapeutic efficacy.
Collapse
Affiliation(s)
- Shahid Shah
- University of Sorbonne Paris Cité‑Paris 7, Lariboisière Hospital, INSERM U965, 75010 Paris, France
| | - Marc Pocard
- University of Sorbonne Paris Cité‑Paris 7, Lariboisière Hospital, INSERM U965, 75010 Paris, France
| | - Massoud Mirshahi
- University of Sorbonne Paris Cité‑Paris 7, Lariboisière Hospital, INSERM U965, 75010 Paris, France
| |
Collapse
|
42
|
Wang Y, Zhao B, Wang L, Bu W, Liu S, Sun B. Nanoparticles based on retinoic acid caped with ferrocenium: a novel synthesized targetable nanoparticle both with anti-cancer effect and drug loading capacity. RSC Adv 2019; 9:16208-16214. [PMID: 35521379 PMCID: PMC9064344 DOI: 10.1039/c9ra02472g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/30/2019] [Indexed: 11/21/2022] Open
Abstract
To date, there is an urgent need for cancer treatment to improve in many ways in order to successfully cure all cancers. Retinoic acid (RA) is a promising anti-cancer drug through influencing cancer stem cells (CSCs). Taxol is a chemotherapy drug for many cancers. To increase the anti-cancer effects of RA and taxol, we created a novel RA nanoparticle, FCRAN, which has the ability of carrying a second anti-cancer drug, taxol, using nanotechnological methods. The results of this study demonstrated that this RA nanoparticle was water-soluble and retained the same effects as RA on cancer cells, such as inhibiting the proliferation of CSCs, inducing the differentiation of CSCs, and enhancing the sensitivity of CSCs to chemotherapeutic drugs. In addition, this RA nanoparticle can be used to carry a second anticancer drug, taxol, to become FCRAN/T and synergistically enhance the anti-cancer effects of both drugs in vivo. Interestingly, the FCRAN/T is a targetable anti-cancer nanoparticle in the presence of higher levels of glutathione (GSH) in cancer cells. Our results demonstrate that our novel synthesized nanoparticles not only retain the RA functions, but can also carry a second anticancer drug to play a synergistic anticancer role with good water solubility, in particular FCRAN/T can target cancer cells. Therefore, our novel synthesized targetable anti-cancer nanoparticles have a better application prospect than that of RA or taxol alone. A retinoic acid nanoparticle with the ability of carrying a second anti-cancer drug, taxol, was developed. The anti-cancer nanoparticles were shown to have a better application prospect than that of RA or taxol alone.![]()
Collapse
Affiliation(s)
- Yibo Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| | - Bin Zhao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China.,Department of Periodontosis, School and Hospital of Stomatology, Jilin University Changchun 130041 People's Republic of China
| | - Lu Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| | - Wenhuan Bu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| | - Shuwei Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University Changchun 130012 People's Republic of China
| | - Bin Sun
- Department of Oral and Maxilloficial Surgery, School and Hospital of Stomatology, Jilin University Changchun 130041 People's Republic of China .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| |
Collapse
|
43
|
Vassalli G. Aldehyde Dehydrogenases: Not Just Markers, but Functional Regulators of Stem Cells. Stem Cells Int 2019; 2019:3904645. [PMID: 30733805 PMCID: PMC6348814 DOI: 10.1155/2019/3904645] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/25/2018] [Indexed: 12/26/2022] Open
Abstract
Aldehyde dehydrogenase (ALDH) is a superfamily of enzymes that detoxify a variety of endogenous and exogenous aldehydes and are required for the biosynthesis of retinoic acid (RA) and other molecular regulators of cellular function. Over the past decade, high ALDH activity has been increasingly used as a selectable marker for normal cell populations enriched in stem and progenitor cells, as well as for cell populations from cancer tissues enriched in tumor-initiating stem-like cells. Mounting evidence suggests that ALDH not only may be used as a marker for stem cells but also may well regulate cellular functions related to self-renewal, expansion, differentiation, and resistance to drugs and radiation. ALDH exerts its functional actions partly through RA biosynthesis, as all-trans RA reverses the functional effects of pharmacological inhibition or genetic suppression of ALDH activity in many cell types in vitro. There is substantial evidence to suggest that the role of ALDH as a stem cell marker comes down to the specific isoform(s) expressed in a particular tissue. Much emphasis has been placed on the ALDH1A1 and ALDH1A3 members of the ALDH1 family of cytosolic enzymes required for RA biosynthesis. ALDH1A1 and ALDH1A3 regulate cellular function in both normal stem cells and tumor-initiating stem-like cells, promoting tumor growth and resistance to drugs and radiation. An improved understanding of the molecular mechanisms by which ALDH regulates cellular function will likely open new avenues in many fields, especially in tissue regeneration and oncology.
Collapse
Affiliation(s)
- Giuseppe Vassalli
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
- Center for Molecular Cardiology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
44
|
Parejo S, Tschan MP, Muraro MG, Garattini E, Spagnoli GC, Schläfli AM. Assessing Autophagy During Retinoid Treatment of Breast Cancer Cells. Methods Mol Biol 2019; 2019:237-256. [PMID: 31359401 DOI: 10.1007/978-1-4939-9585-1_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Retinoids are derived from vitamin A through a multi-step process. Within a target cell, retinoids regulate gene expression by activating the retinoid acid receptors (RAR) and retinoid x receptors (RXR), which are ligand-dependent transcription factors. Besides its therapeutic use in dermatological disorders, all-trans retinoic acid (ATRA) is successfully utilized to treat acute promyelocytic leukemia (APL) patients. The use of ATRA in APL patients is the first example of clinically useful differentiation therapy. Therapeutic strategies aiming at cancer cell differentiation have great potential for solid tumors, including breast cancer. The few clinical studies conducted with ATRA in breast cancer are rather disappointing. However, these studies did not take into account the heterogeneity of the disease and were conducted on unselected cohorts of patients.We recently showed that ATRA treatment of breast cancer cells induces autophagy, a highly conserved process aiming at degrading and recycling superfluous or harmful cellular components. In addition, autophagy inhibition significantly increases the therapeutic activity of ATRA. This finding is of fundamental importance, since autophagy has a dual role in cancer. Whereas autophagy may be a protective mechanism during the initial phases of cancer development, it may support cancer cell survival in already established tumors. Furthermore, autophagy can lower or enhance therapeutic efficiency, depending on the tumor type and the anticancer agent considered. Therefore, it is important to investigate the role of autophagy in the context of specific tumors and therapeutic approaches. Accurate autophagy studies are challenging given the dynamic nature of the process and the difficulty of measuring the rate of autophagosome degradation (autophagic flux). In this chapter, we provide protocols for a careful assessment of the autophagic flux in ATRA treated 2D and 3D breast cancer cultures.
Collapse
Affiliation(s)
- Sarah Parejo
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Manuele G Muraro
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Enrico Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giulio C Spagnoli
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Anna M Schläfli
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
45
|
Toledo-Guzmán ME, Hernández MI, Gómez-Gallegos ÁA, Ortiz-Sánchez E. ALDH as a Stem Cell Marker in Solid Tumors. Curr Stem Cell Res Ther 2019; 14:375-388. [PMID: 30095061 DOI: 10.2174/1574888x13666180810120012] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023]
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme that participates in important cellular mechanisms as aldehyde detoxification and retinoic acid synthesis; moreover, ALDH activity is involved in drug resistance, a characteristic of cancer stem cells (CSCs). Even though ALDH is found in stem cells, CSCs and progenitor cells, this enzyme has been successfully used to identify and isolate cell populations with CSC properties from several tumor origins. ALDH is allegedly involved in cell differentiation through its product, retinoic acid. However, direct or indirect ALDH inhibition, using specific inhibitors or retinoic acid, has shown a reduction in ALDH activity, along with the loss of stem cell traits, reduction of cell proliferation, invasion, and drug sensitization. For these reasons, ALDH and retinoic acid are promising therapeutic targets. This review summarizes the current evidence for ALDH as a CSCs marker in solid tumors, as well as current knowledge about the functional roles of ALDH in CSCs. We discuss the controversy of ALDH activity to maintain CSC stemness, or conversely, to promote cell differentiation. Finally, we review the advances in using ALDH inhibitors as anti-cancer drugs.
Collapse
Affiliation(s)
- Mariel E Toledo-Guzmán
- Departamento de Bioquimica, Laboratorio de Terapia Genica, Escuela Nacional de Ciencias Biologicas, Posgrado de Biomedicina y Biotecnologia Molecular, Instituto Politecnico Nacional, Mexico City, Mexico
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
| | - Miguel Ibañez Hernández
- Departamento de Bioquimica, Laboratorio de Terapia Genica, Escuela Nacional de Ciencias Biologicas, Posgrado de Biomedicina y Biotecnologia Molecular, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Ángel A Gómez-Gallegos
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
- Posgrado de Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
| |
Collapse
|
46
|
ALDH1-positive intratumoral stromal cells indicate differentiated epithelial-like phenotype and good prognosis in prostate cancer. Transl Res 2019; 203:49-56. [PMID: 30287243 DOI: 10.1016/j.trsl.2018.08.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/28/2018] [Accepted: 08/16/2018] [Indexed: 01/16/2023]
Abstract
Aldehyde dehydrogenase 1 (ALDH1) characterizes tumor-initiating cells in solid tumors; however, little is known about its expression in intratumoral stromal cells. Herein, we aimed to dissect its potential dual relevance in prostate cancer (PCa). ALDH1 expression was evaluated immunohistochemically in tumor and stromal cells in primary PCa and metastases. It was correlated to clinico-pathologic parameters, patients' outcome, and selected proteins (CK5/6, CK14, CK8/18, CK19, EpCAM, Ki-67, E-cadherin, N-cadherin, and vimentin). ALDH1 protein was detected in tumor and stromal cells in 16% and 67% of 348 primary PCa, respectively. Tumor cell ALDH1 expression was associated with advanced T stage (P = 0.009), higher Gleason score (P = 0.016), shorter time to biochemical recurrence (TBR P = 0.010) and CK14 expression (P = 0.023). Stromal cell ALDH1 expression correlated to lower T stage (P = 0.008) and Gleason score (P = 0.016), N0 stage (P = 0.017), and longer TBR (P = 0.017). It occurred to be an independent predictor of good prognosis in the subgroup of d'Amico high-risk patients (multivariate analysis, P = 0.050). ALDH1-positive stromal cells were found in tumors characterized frequently by CK8/18 (P = 0.033) or EpCAM expression (P < 0.001) and rarely by epithelial-mesenchymal transition defined as CK8/18(-)vimentin(+) phenotype (P = 0.003). ALDH1-positive tumor and stromal cells were detected in 33% and 41% of hormone naive lymph node metastases (n = 63), 52% and 24% of castration resistant bone metastases, as well as 89% and 28% of castration resistant visceral metastases (n = 21), respectively. We have determined that contrary to tumor cell ALDH1, the presence of stromal ALDH1 is associated with epithelial phenotype of primary PCa, improved clinical outcome, and is less frequent in PCa metastases.
Collapse
|
47
|
Toledo-Guzmán ME, Bigoni-Ordóñez GD, Ibáñez Hernández M, Ortiz-Sánchez E. Cancer stem cell impact on clinical oncology. World J Stem Cells 2018; 10:183-195. [PMID: 30613312 PMCID: PMC6306557 DOI: 10.4252/wjsc.v10.i12.183] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/15/2018] [Accepted: 11/15/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer is a widespread worldwide chronic disease. In most cases, the high mortality rate from cancer correlates with a lack of clear symptoms, which results in late diagnosis for patients, and consequently, advanced tumor disease with poor probabilities for cure, since many patients will show chemo- and radio-resistance. Several mechanisms have been studied to explain chemo- and radio-resistance to anti-tumor therapies, including cell signaling pathways, anti-apoptotic mechanisms, stemness, metabolism, and cellular phenotypes. Interestingly, the presence of cancer stem cells (CSCs), which are a subset of cells within the tumors, has been related to therapy resistance. In this review, we focus on evaluating the presence of CSCs in different tumors such as breast cancer, gastric cancer, lung cancer, and hematological neoplasias, highlighting studies where CSCs were identified in patient samples. It is evident that there has been a great drive to identify the cell surface phenotypes of CSCs so that they can be used as a tool for anti-tumor therapy treatment design. We also review the potential effect of nanoparticles, drugs, natural compounds, aldehyde dehydrogenase inhibitors, cell signaling inhibitors, and antibodies to treat CSCs from specific tumors. Taken together, we present an overview of the role of CSCs in tumorigenesis and how research is advancing to target these highly tumorigenic cells to improve oncology patient outcomes.
Collapse
Affiliation(s)
- Mariel E Toledo-Guzmán
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
| | | | - Miguel Ibáñez Hernández
- Departamento de Bioquímica, Laboratorio de Terapia Génica, Escuela Nacional de Ciencias Biológicas, Posgrado de Biomedicina y Biotecnología Molecular, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico.
| |
Collapse
|
48
|
Dasatinib sensitises triple negative breast cancer cells to chemotherapy by targeting breast cancer stem cells. Br J Cancer 2018; 119:1495-1507. [PMID: 30482914 PMCID: PMC6288167 DOI: 10.1038/s41416-018-0287-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/21/2018] [Accepted: 09/12/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Patients with triple negative breast cancer (TNBC) exhibit poor prognosis and are at high risk of tumour relapse, due to the resistance to chemotherapy. These aggressive phenotypes are in part attributed to the presence of breast cancer stem cells (BCSCs). Therefore, targeting BCSCs is a priority to overcoming chemotherapy failure in TNBCs. METHODS We generated paclitaxel (pac)-resistant TNBC cells which displayed higher sphere forming potential and percentage of BCSC subpopulations compared to the parental cells. A screen with various kinase inhibitors revealed dasatinib, a Src kinase family inhibitor, as a potent suppressor of BCSC expansion/sphere formation in pac-resistant TNBC cells. RESULTS We found dasatinib to block pac-induced BCSC enrichment and Src activation in both parental and pac-resistant TNBC cells. Interestingly, dasatinib induced an epithelial differentiation of the pac-resistant mesenchymal cells, resulting in their enhanced sensitivity to paclitaxel. The combination treatment of dasatinib and paclitaxel not only decreased the BCSCs numbers and their sphere forming capacity but also synergistically reduced cell viability of pac-resistant cells. Preclinical models of breast cancer further demonstrated the efficiency of the dasatinib/paclitaxel combination treatment in inhibiting tumour growth. CONCLUSIONS Dasatinib is a promising anti-BCSC drug that could be used in combination with paclitaxel to overcome chemoresistance in TNBC.
Collapse
|
49
|
Merino VF, Cho S, Nguyen N, Sadik H, Narayan A, Talbot C, Cope L, Zhou XC, Zhang Z, Győrffy B, Sukumar S. Induction of cell cycle arrest and inflammatory genes by combined treatment with epigenetic, differentiating, and chemotherapeutic agents in triple-negative breast cancer. Breast Cancer Res 2018; 20:145. [PMID: 30486871 PMCID: PMC6263070 DOI: 10.1186/s13058-018-1068-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/24/2018] [Indexed: 12/21/2022] Open
Abstract
Background A combination of entinostat, all-trans retinoic acid, and doxorubicin (EAD) induces cell death and differentiation and causes significant regression of xenografts of triple-negative breast cancer (TNBC). Methods We investigated the mechanisms underlying the antitumor effects of each component of the EAD combination therapy by high-throughput gene expression profiling of drug-treated cells. Results Microarray analysis showed that entinostat and doxorubicin (ED) altered expression of genes related to growth arrest, inflammation, and differentiation. ED downregulated MYC, E2F, and G2M cell cycle genes. Accordingly, entinostat sensitized the cells to doxorubicin-induced growth arrest at G2. ED induced interferon genes, which correlated with breast tumors containing a higher proportion of tumor-infiltrating lymphocytes. ED also increased the expression of immune checkpoint agonists and cancer testis antigens. Analysis of TNBC xenografts showed that EAD enhanced the inflammation score in nude mice. Among the genes differentially regulated between the EAD and ED groups, an all-trans retinoic acid (ATRA)-regulated gene, DHRS3, was induced in EAD-treated xenografts. DHRS3 was expressed at lower levels in human TNBC metastases compared to normal breast or primary tumors. High expression of ED-induced growth arrest and inflammatory genes was associated with better prognosis in TNBC patients. Conclusions Entinostat potentiated doxorubicin-mediated cell death and the combination induced inflammatory signatures. The ED-induced immunomodulation may improve immunotherapy. Addition of ATRA to ED may potentiate inflammation and contribute to TNBC regression. Electronic supplementary material The online version of this article (10.1186/s13058-018-1068-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vanessa F Merino
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Soonweng Cho
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nguyen Nguyen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Helen Sadik
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Athira Narayan
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Conover Talbot
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie Cope
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xian C Zhou
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhe Zhang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary.,2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
50
|
Dorokhov YL, Sheshukova EV, Bialik TE, Komarova TV. Human Endogenous Formaldehyde as an Anticancer Metabolite: Its Oxidation Downregulation May Be a Means of Improving Therapy. Bioessays 2018; 40:e1800136. [PMID: 30370669 DOI: 10.1002/bies.201800136] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/27/2018] [Indexed: 02/06/2023]
Abstract
Malignant cells are characterized by an increased content of endogenous formaldehyde formed as a by-product of biosynthetic processes. Accumulation of formaldehyde in cancer cells is combined with activation of the processes of cellular formaldehyde clearance. These mechanisms include increased ALDH and suppressed ADH5/FDH activity, which oncologists consider poor and favorable prognostic markers, respectively. Here, the sources and regulation of formaldehyde metabolism in cancer cells are reviewed. The authors also analyze the participation of oncoproteins such as fibulins, FGFR1, HER2/neu, FBI-1, and MUC1-C in the control of genes related to formaldehyde metabolism, suggesting the existence of two mutually exclusive processes in cancer cells: 1) production and 2) oxidation and elimination of formaldehyde from the cell. The authors hypothesize that the study of the anticancer properties of disulfiram and alpha lipoic acid - which affect the balance of formaldehyde in the body - may serve as the basis of future anticancer therapy.
Collapse
Affiliation(s)
- Yuri L Dorokhov
- N.I. Vavilov Institute of General Genetics of RAS, 119991, Moscow, Russia.,A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | | | - Tatiana E Bialik
- N.N. Blokhin National Medical Research Center of Oncology, 115478, Moscow, Russia
| | - Tatiana V Komarova
- N.I. Vavilov Institute of General Genetics of RAS, 119991, Moscow, Russia.,A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| |
Collapse
|