1
|
Zarrella S, Miranda MR, Covelli V, Restivo I, Novi S, Pepe G, Tesoriere L, Rodriquez M, Bertamino A, Campiglia P, Tecce MF, Vestuto V. Endoplasmic Reticulum Stress and Its Role in Metabolic Reprogramming of Cancer. Metabolites 2025; 15:221. [PMID: 40278350 PMCID: PMC12029571 DOI: 10.3390/metabo15040221] [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: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Endoplasmic reticulum (ER) stress occurs when ER homeostasis is disrupted, leading to the accumulation of misfolded or unfolded proteins. This condition activates the unfolded protein response (UPR), which aims to restore balance or trigger cell death if homeostasis cannot be achieved. In cancer, ER stress plays a key role due to the heightened metabolic demands of tumor cells. This review explores how metabolomics can provide insights into ER stress-related metabolic alterations and their implications for cancer therapy. Methods: A comprehensive literature review was conducted to analyze recent findings on ER stress, metabolomics, and cancer metabolism. Studies examining metabolic profiling of cancer cells under ER stress conditions were selected, with a focus on identifying potential biomarkers and therapeutic targets. Results: Metabolomic studies highlight significant shifts in lipid metabolism, protein synthesis, and oxidative stress management in response to ER stress. These metabolic alterations are crucial for tumor adaptation and survival. Additionally, targeting ER stress-related metabolic pathways has shown potential in preclinical models, suggesting new therapeutic strategies. Conclusions: Understanding the metabolic impact of ER stress in cancer provides valuable opportunities for drug development. Metabolomics-based approaches may help identify novel biomarkers and therapeutic targets, enhancing the effectiveness of antitumor therapies.
Collapse
Affiliation(s)
- Salvatore Zarrella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Verdiana Covelli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Manuela Rodriquez
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| |
Collapse
|
2
|
Dhar S, Sarkar T, Bose S, Pati S, Chakraborty D, Roy D, Panda AK, Guin A, Mukherjee S, Jana K, Sarkar DK, Sa G. FOXP3 Transcriptionally Activates Fatty Acid Scavenger Receptor CD36 in Tumour-Induced Treg Cells. Immunology 2025; 174:296-309. [PMID: 39736083 DOI: 10.1111/imm.13887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
The host immune system is adapted in a variety of ways by tumour microenvironment and growing tumour interacts to promote immune escape. One of these adaptations is manipulating the metabolic processes of cells in the tumour microenvironment. The growing tumour aggressively utilise glucose, its primary energy source available in tumour site, and produce lactate by Warburg effect. In such a hostile environment, tumour-infiltrating immune cells are unable to survive metabolically. Tumour-infiltrating CD4+ Treg cells, on the other hand, adapted to an alternative energy-generating system, switching from the highly-competitive glucose to the fatty-acid metabolic pathway, by down-regulating glucose-metabolising genes and up-regulating fatty-acid metabolising genes. Tregs with high-levels of the fatty acid scavenger receptor CD36, a key component of the fatty-acid metabolic pathway, aided this metabolic shift. Treg cell formation was hampered when the fatty-acid metabolic pathway was disrupted, showing that it is necessary for Treg cell development. FOXP3, the Treg lineage-specific transcription factor, regulates fatty-acid metabolism by inducing CD36 transcription. A high-fat diet enhanced Treg development while suppressing anti-tumour immunity, whereas a low-fat diet suppressed Treg development. The altered metabolism of tumour-infiltrating Treg cells enables their rapid generation and survival in the hostile tumour microenvironment, aiding cancer progression. Fascinatingly, mice fed with a low-fat diet showed a positive prognosis with chemotherapy than mice fed with a high-fat diet. Thus, a maximum efficacy of chemotherapy might be achieved by altering diet composition during chemotherapy, providing a promising indication for future cancer treatment.
Collapse
Affiliation(s)
- Subhanki Dhar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Tania Sarkar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sayantan Bose
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Subhadip Pati
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Dia Roy
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Abir K Panda
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Aharna Guin
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sumon Mukherjee
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
3
|
Mosadegh M, Noori Goodarzi N, Erfani Y. A Comprehensive Insight into Apoptosis: Molecular Mechanisms, Signaling Pathways, and Modulating Therapeutics. Cancer Invest 2025; 43:33-58. [PMID: 39760426 DOI: 10.1080/07357907.2024.2445528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 11/15/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Apoptosis, or programmed cell death, is a fundamental biological process essential for maintaining tissue homeostasis. Dysregulation of apoptosis is implicated in a variety of diseases, including cancer, neurodegenerative disorders, and autoimmune conditions. This review provides an in-depth insight into the molecular mechanisms and signaling pathways that regulate apoptosis, highlighting both intrinsic and extrinsic pathways. Additionally, the review explains the tumor microenvironment's influence on apoptosis and its implications for cancer therapy resistance. Understanding the complex interplay between apoptotic signaling and cellular responses is crucial for developing targeted therapies that can effectively manage diseases associated with apoptosis dysregulation. The effects of conventional therapeutics and alternative substances with natural sources such as herbal compounds, alongside vitamins, minerals, and trace elements on cellular homeostasis and disease pathogenesis have been thoroughly investigated. Moreover, recent advances in therapeutic strategies aimed at modulating apoptosis are discussed, with a focus on novel interventions such as nutrition bio shield dietary supplement. These emerging approaches offer potential benefits beyond conventional treatments by selectively targeting apoptotic pathways to inhibit cancer progression and metastasis. By integrating insights from recent studies, this review aims to enhance our understanding of apoptosis and guide future research in developing innovative therapeutic approaches.
Collapse
Affiliation(s)
- Mehrdad Mosadegh
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Narjes Noori Goodarzi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Erfani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Wu Y, Wang X, Zeng Y, Liu X. Exosomes are the mediators between the tumor microenvironment and prostate cancer (Review). Exp Ther Med 2024; 28:439. [PMID: 39355518 PMCID: PMC11443591 DOI: 10.3892/etm.2024.12728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/23/2024] [Indexed: 10/03/2024] Open
Abstract
Prostate cancer poses a serious threat to the well-being of men worldwide, with the leading cause of mortality being primarily through metastasis. Prostate cancer metastasis is dependent on cell communication, which is an essential component of this process; yet its exact mechanism remains obscure. Nonetheless, cell-to-cell communication plays a critical part in prostate cancer metastasis. Exosomes play an indispensable role in the development of metastatic growth by promoting intercellular communication. They are pivotal regulatory agents for both prostate cancer cells as well as their microenvironment. The present study investigated the makeup and function of exosomes in the tumor microenvironment, highlighting their significance to prostate cancer metastasis.
Collapse
Affiliation(s)
- Yiqi Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiao Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan Zeng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
5
|
Geng S, Yu X, Yu S. Efficacy and safety of natural killer cells injection combined with XELOX chemotherapy in postoperative patients with stage III colorectal cancer in China: a prospective randomised controlled clinical trial study protocol. BMJ Open 2024; 14:e080377. [PMID: 38531576 PMCID: PMC10966825 DOI: 10.1136/bmjopen-2023-080377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the second most frequently diagnosed cancer and the fifth leading cause of cancer-related death in China. However, resistance to multiple chemotherapeutics after surgery leads to failure of the main therapy to CRC. Natural killer (NK) cells are innate cytotoxic lymphocytes that exhibit strong cytotoxic activity against tumour cells. NK cell-based therapy, either alone or in combination with chemotherapy, has achieved favourable results and holds promise for addressing recurrence and metastasis in CRC patients after surgery. METHODS AND ANALYSIS This is a prospective, randomised controlled clinical trial to evaluate efficacy and safety of interleukin 2 activated NK cells injection combined with XELOX (capecitabine plus oxaliplatin)-based chemotherapy for postoperative CRC patients. Participants will be randomly divided into treatment group and control group, and every group includes 40 patients. The treatment group will also receive NK cells (5×109) with+XELOX-based chemotherapy, while the control group will receive only XELOX-based chemotherapy. This treatment will be repeated for eight cycles (6 months). The follow-up period lasts about 3 years, during which CEA, CA19-9, CA125, enhancement CT and colonoscopy will be conducted. The primary endpoints of this study are progression-free survival and overall survival, while the secondary endpoint is safety (number and severity of adverse events). Additionally, we aim to identify cancer stem cells in peripheral blood and predictive biomarkers (cytokines secreted by NK cells and activated markers of NK cells) that indicate patients who achieve an effective response. ETHICS AND DISSEMINATION The study has been approved by the Clinical Research Ethics Committee of our hospital (approval number 2023LLSC006) and the Chinese Clinical Trials. It will be conducted in accordance with the Declaration of Helsinki. Written informed consent will be obtained from all participants. The study findings will be submitted to peer-reviewed journals for publication. TRIAL REGISTRATION NUMBER Chinese Clinical Trials Registry (ChiCTR2300075861).
Collapse
Affiliation(s)
- Shan Geng
- Department of Endocrinology, The People's Hospital of Dazu Chongqing, Chongqing, China
| | - Xingrui Yu
- Department of Computer Science, Xiamen University, Xiamen, China
| | - Shaohong Yu
- Department of General Surgery, The People's Hospital of Dazu Chongqing, Chongqing, China
| |
Collapse
|
6
|
Di X, Pei Z, Pei Y, James TD. Tumor microenvironment-oriented MOFs for chemodynamic therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
7
|
Kitsios K, Sharifi S, Mahmoudi M. Nanomedicine Technologies for Diagnosis and Treatment of Breast Cancer. ACS Pharmacol Transl Sci 2023; 6:671-682. [PMID: 37200812 PMCID: PMC10186357 DOI: 10.1021/acsptsci.3c00044] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Indexed: 05/20/2023]
Abstract
Breast cancer is one of the most common cancers in women worldwide, yet conventional treatments have several shortcomings, including low specificity, systemic toxicity, and drug resistance. Nanomedicine technologies provide a promising alternative while also overcoming the limitations posed by conventional therapies. This mini-Review highlights important signaling pathways related to occurrence and development of breast cancer and current breast cancer therapies, followed by an analysis of various nanomedicine technologies developed for diagnosis and treatment of breast cancers.
Collapse
Affiliation(s)
- Katerina Kitsios
- Department of Radiology and
Precision Health Program, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Shahriar Sharifi
- Department of Radiology and
Precision Health Program, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Morteza Mahmoudi
- Department of Radiology and
Precision Health Program, Michigan State
University, East Lansing, Michigan 48824, United States
| |
Collapse
|
8
|
Lalani AR, Fakhari F, Radgoudarzi S, Rastegar-Pouyani N, Moloudi K, Khodamoradi E, Taeb S, Najafi M. Immunoregulation by resveratrol; implications for normal tissue protection and tumour suppression. Clin Exp Pharmacol Physiol 2023; 50:353-368. [PMID: 36786378 DOI: 10.1111/1440-1681.13760] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
Immune reactions are involved in both tumour and normal tissue in response to therapy. Elevated secretion of certain chemokines, exosomes and cytokines triggers inflammation, pain, fibrosis and ulceration among other normal tissue side effects. On the other hand, secretion of tumour-promoting molecules suppresses activity of anticancer immune cells and facilitates the proliferation of malignant cells. Novel anticancer drugs such as immune checkpoint inhibitors (ICIs) boost anticancer immunity via inducing the proliferation of anticancer cells such as natural killer (NK) cells and CD8+ T lymphocytes. Certain chemotherapy drugs and radiotherapy may induce anticancer immunity in the tumour, however, both have severe side effects for normal tissues through stimulation of several immune responses. Thus, administration of natural products with low side effects may be a promising approach to modulate the immune system in both tumour and normal organs. Resveratrol is a well-known phenol with diverse effects on normal tissues and tumours. To date, a large number of experiments have confirmed the potential of resveratrol as an anticancer adjuvant. This review focuses on ensuing stimulation or suppression of immune responses in both tumour and normal tissue after radiotherapy or anticancer drugs. Later on, the immunoregulatory effects of resveratrol in both tumour and normal tissue following exposure to anticancer agents will be discussed.
Collapse
Affiliation(s)
- Armineh Rezagholi Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fatemeh Fakhari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shakila Radgoudarzi
- I.M. Sechenov First Moscow State Medical University (Первый МГМУ им), Moscow, Russia
| | - Nima Rastegar-Pouyani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Kave Moloudi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran.,Medical Biotechnology Research Center, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
9
|
Ghazvinian Z, Abdolahi S, Tokhanbigli S, Tarzemani S, Piccin A, Reza Zali M, Verdi J, Baghaei K. Contribution of natural killer cells in innate immunity against colorectal cancer. Front Oncol 2023; 12:1077053. [PMID: 36686835 PMCID: PMC9846259 DOI: 10.3389/fonc.2022.1077053] [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: 10/22/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Natural killer cells are members of the innate immune system and promote cytotoxic activity against tumor or infected cells independently from MHC recognition. NK cells are modulated by the expression of activator/inhibitory receptors. The ratio of this activator/inhibitory receptors is responsible for the cytotoxic activity of NK cells toward the target cells. Owing to the potent anti-tumor properties of NK cells, they are considered as interesting approach in tumor treatment. Colorectal cancer (CRC) is the second most common cause of death in the world and the incidence is about 2 million new cases per year. Metastatic CRC is accompanied by a poor prognosis with less than three years of overall survival. Chemotherapy and surgery are the most adopted treatments. Besides, targeted therapy and immune checkpoint blockade are novel approach to CRC treatment. In these patients, circulating NK cells are a prognostic marker. The main target of CRC immune cell therapy is to improve the tumor cell's recognition and elimination by immune cells. Adaptive NK cell therapy is the milestone to achieve the purpose. Allogeneic NK cell therapy has been widely investigated within clinical trials. In this review, we focus on the NK related approaches including CAR NK cells, cell-based vaccines, monoclonal antibodies and immunomodulatory drugs against CRC tumoral cells.
Collapse
Affiliation(s)
- Zeinab Ghazvinian
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrokh Abdolahi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Tokhanbigli
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shadi Tarzemani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Andrea Piccin
- Northern Ireland Blood Transfusion Service, Belfast, United Kingdom
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
- Department of Industrial Engineering, University of Trento, Trento, Italy
| | - Mohammad Reza Zali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Xie Y, Wang M, Sun Q, Wang D, Li C. Recent Advances in Tetrakis (4‐Carboxyphenyl) Porphyrin‐Based Nanocomposites for Tumor Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Yulin Xie
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Man Wang
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Qianqian Sun
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Dongmei Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials College of Chemistry and Life Sciences Zhejiang Normal University Jinhua 321004 P.R. China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| |
Collapse
|
11
|
High Expression of E2F4 Is an Adverse Prognostic Factor and Related to Immune Infiltration in Oral Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4731364. [PMID: 36567912 PMCID: PMC9780755 DOI: 10.1155/2022/4731364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/01/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Background We aimed to evaluate the prognostic value of E2F4 expression in oral squamous cell carcinoma (OSCC) and clarify its influence on immune cell infiltration and biological functions. Methods The Cancer Genome Atlas (TCGA) database, the STRING database, and related online tools as well as single-sample gene set enrichment analysis (ssGSEA) were used for the analyses in our study. Results The E2F4 expression was elevated in OSCC tumor tissue compared with paracancerous tissues. The high expression of E2F4 was closely related to the poorer overall survival, disease-free survival, and progression-free interval of OSCC. In addition, pathway enrichment analyses revealed that the top 49 genes most closely related to E2F4 were strongly associated with the cell cycle. E2F4-related proteins were closely related to the following KEGG pathways: cell cycle, cellular senescence, PI3K-Akt signaling pathway, Wnt signaling pathway, and notch signaling pathway. It was also demonstrated that the E2F4 expression was negatively correlated with immune purity and strongly related to immune cell infiltration in OSCC. Conclusions E2F4 can be used as a novel biomarker for the diagnosis and prognosis of OSCC.
Collapse
|
12
|
Ali AA, Abuwatfa WH, Al-Sayah MH, Husseini GA. Gold-Nanoparticle Hybrid Nanostructures for Multimodal Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12203706. [PMID: 36296896 PMCID: PMC9608376 DOI: 10.3390/nano12203706] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 06/01/2023]
Abstract
With the urgent need for bio-nanomaterials to improve the currently available cancer treatments, gold nanoparticle (GNP) hybrid nanostructures are rapidly rising as promising multimodal candidates for cancer therapy. Gold nanoparticles (GNPs) have been hybridized with several nanocarriers, including liposomes and polymers, to achieve chemotherapy, photothermal therapy, radiotherapy, and imaging using a single composite. The GNP nanohybrids used for targeted chemotherapy can be designed to respond to external stimuli such as heat or internal stimuli such as intratumoral pH. Despite their promise for multimodal cancer therapy, there are currently no reviews summarizing the current status of GNP nanohybrid use for cancer theragnostics. Therefore, this review fulfills this gap in the literature by providing a critical analysis of the data available on the use of GNP nanohybrids for cancer treatment with a specific focus on synergistic approaches (i.e., triggered drug release, photothermal therapy, and radiotherapy). It also highlights some of the challenges that hinder the clinical translation of GNP hybrid nanostructures from bench to bedside. Future studies that could expedite the clinical progress of GNPs, as well as the future possibility of improving GNP nanohybrids for cancer theragnostics, are also summarized.
Collapse
Affiliation(s)
- Amaal Abdulraqeb Ali
- Biomedical Engineering Graduate Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Mohammad H. Al-Sayah
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
13
|
Ali AA, Al-Othman A, Al-Sayah MH. Multifunctional stimuli-responsive hybrid nanogels for cancer therapy: Current status and challenges. J Control Release 2022; 351:476-503. [PMID: 36170926 DOI: 10.1016/j.jconrel.2022.09.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022]
Abstract
With cancer research shifting focus to achieving multifunctionality in cancer treatment strategies, hybrid nanogels are making a rapid rise to the spotlight as novel, multifunctional, stimuli-responsive, and biocompatible cancer therapeutic strategies. They can possess cancer cell-specific cytotoxic effects themselves, carry drugs or enzymes that can produce cytotoxic effects, improve imaging modalities, and target tumor cells over normal cells. Hybrid nanogels bring together a wide range of desirable properties for cancer treatment such as stimuli-responsiveness, efficient loading and protection of molecules such as drugs or enzymes, and effective crossing of cellular barriers among other properties. Despite their promising abilities, hybrid nanogels are still far from being used in the clinic, and their available data remains relatively limited. However, many studies can be done to facilitate this clinical transition. This review is critically summarizing and analyzing the recent information and progress on the use of hybrid nanogels particularly inorganic nanoparticle-based and organic nanoparticle-based hybrid nanogels in the field of oncology and future directions to aid in transferring those results to the clinic. This work concludes that the future of hybrid nanogels is greatly impacted by therapeutic and non-therapeutic factors. Therapeutic factors include the lack of hemocompatibility studies, acute and chronic toxicological studies, and information on agglomeration capability and extent, tumor heterogeneity, interaction with proteins in physiological fluids, endocytosis-exocytosis, and toxicity of the nanogels' breakdown products. Non-therapeutic factors include the lack of clear regulatory guidelines and standardized assays, limitations of animal models, and difficulties associated with good manufacture practices (GMP).
Collapse
Affiliation(s)
- Amaal Abdulraqeb Ali
- Biomedical Engineering Graduate Program, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| | - Amani Al-Othman
- Department of Chemical Engineering, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates.
| | - Mohammad H Al-Sayah
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| |
Collapse
|
14
|
Charlestin V, Fulkerson D, Arias Matus CE, Walker ZT, Carthy K, Littlepage LE. Aquaporins: New players in breast cancer progression and treatment response. Front Oncol 2022; 12:988119. [PMID: 36212456 PMCID: PMC9532844 DOI: 10.3389/fonc.2022.988119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
Aquaporins (AQPs) are a family of small transmembrane proteins that selectively transport water and other small molecules and ions following an osmotic gradient across cell plasma membranes. This enables them to regulate numerous functions including water homeostasis, fat metabolism, proliferation, migration, and adhesion. Previous structural and functional studies highlight a strong biological relationship between AQP protein expression, localization, and key biological functions in normal and cancer tissues, where aberrant AQP expression correlates with tumorigenesis and metastasis. In this review, we discuss the roles of AQP1, AQP3, AQP4, AQP5, and AQP7 in breast cancer progression and metastasis, including the role of AQPs in the tumor microenvironment, to highlight potential contributions of stromal-derived to epithelial-derived AQPs to breast cancer. Emerging evidence identifies AQPs as predictors of response to cancer therapy and as targets for increasing their sensitivity to treatment. However, these studies have not evaluated the requirements for protein structure on AQP function within the context of breast cancer. We also examine how AQPs contribute to a patient's response to cancer treatment, existing AQP inhibitors and how AQPs could serve as novel predictive biomarkers of therapy response in breast cancer. Future studies also should evaluate AQP redundancy and compensation as mechanisms used to overcome aberrant AQP function. This review highlights the need for additional research into how AQPs contribute molecularly to therapeutic resistance and by altering the tumor microenvironment.
Collapse
Affiliation(s)
- Verodia Charlestin
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Daniel Fulkerson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Carlos E. Arias Matus
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
- Department of Biotechnology, Universidad Popular Autónoma del Estado de Puebla, Pue, Mexico
| | - Zachary T. Walker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Kevin Carthy
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Laurie E. Littlepage
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| |
Collapse
|
15
|
Gentile D, Esposito M, Grumati P. Metabolic adaption of cancer cells toward autophagy: Is there a role for ER-phagy? Front Mol Biosci 2022; 9:930223. [PMID: 35992272 PMCID: PMC9382244 DOI: 10.3389/fmolb.2022.930223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Autophagy is an evolutionary conserved catabolic pathway that uses a unique double-membrane vesicle, called autophagosome, to sequester cytosolic components, deliver them to lysosomes and recycle amino-acids. Essentially, autophagy acts as a cellular cleaning system that maintains metabolic balance under basal conditions and helps to ensure nutrient viability under stress conditions. It is also an important quality control mechanism that removes misfolded or aggregated proteins and mediates the turnover of damaged and obsolete organelles. In this regard, the idea that autophagy is a non-selective bulk process is outdated. It is now widely accepted that forms of selective autophagy are responsible for metabolic rewiring in response to cellular demand. Given its importance, autophagy plays an essential role during tumorigenesis as it sustains malignant cellular growth by acting as a coping-mechanisms for intracellular and environmental stress that occurs during malignant transformation. Cancer development is accompanied by the formation of a peculiar tumor microenvironment that is mainly characterized by hypoxia (oxygen < 2%) and low nutrient availability. Such conditions challenge cancer cells that must adapt their metabolism to survive. Here we review the regulation of autophagy and selective autophagy by hypoxia and the crosstalk with other stress response mechanisms, such as UPR. Finally, we discuss the emerging role of ER-phagy in sustaining cellular remodeling and quality control during stress conditions that drive tumorigenesis.
Collapse
Affiliation(s)
- Debora Gentile
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Marianna Esposito
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Scuola Superiore Meridionale, Naples, Italy
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
- *Correspondence: Paolo Grumati,
| |
Collapse
|
16
|
Wang Y, Liu X, Quan X, Qin X, Zhou Y, Liu Z, Chao Z, Jia C, Qin H, Zhang H. Pigment epithelium-derived factor and its role in microvascular-related diseases. Biochimie 2022; 200:153-171. [DOI: 10.1016/j.biochi.2022.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023]
|
17
|
Vieira AM, Silvestre OF, Silva BF, Ferreira CJ, Lopes I, Gomes AC, Espiña B, Sárria MP. pH-sensitive nanoliposomes for passive and CXCR-4-mediated marine yessotoxin delivery for cancer therapy. Nanomedicine (Lond) 2022; 17:717-739. [PMID: 35481356 DOI: 10.2217/nnm-2022-0010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Yessotoxin (YTX), a marine-derived drug, was encapsulated in PEGylated pH-sensitive nanoliposomes, covalently functionalized (strategy I) with SDF-1α and by nonspecific adsorption (strategy II), to actively target chemokine receptor CXCR-4. Methods: Cytotoxicity to normal human epithelial cells (HK-2) and prostate (PC-3) and breast (MCF-7) adenocarcinoma models, with different expression levels of CXCR-4, were tested. Results: Strategy II exerted the highest cytotoxicity toward cancer cells while protecting normal epithelia. Acid pH-induced fusion of nanoliposomes seemed to serve as a primary route of entry into MCF-7 cells but PC-3 data support an endocytic pathway for their internalization. Conclusion: This work describes an innovative hallmark in the current marine drug clinical pipeline, as the developed nanoliposomes are promising candidates in the design of groundbreaking marine flora-derived anticancer nanoagents.
Collapse
Affiliation(s)
- Ana Mg Vieira
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal.,Centre of Molecular & Environmental Biology (CBMA), University of Minho, Braga, 4710-057, Portugal
| | - Oscar F Silvestre
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| | - Bruno Fb Silva
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| | - Celso Jo Ferreira
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal.,Centro de Física das Universidades do Minho e do Porto (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
| | - Ivo Lopes
- Centre of Molecular & Environmental Biology (CBMA), University of Minho, Braga, 4710-057, Portugal
| | - Andreia C Gomes
- Centre of Molecular & Environmental Biology (CBMA), University of Minho, Braga, 4710-057, Portugal.,Institute of Science & Innovation for Biosustainability (IB-S), University of Minho, Braga, 4710-057, Portugal
| | - Begoña Espiña
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| | - Marisa P Sárria
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| |
Collapse
|
18
|
Lou H, Chu L, Zhou W, Dou J, Teng X, Tan W, Zhou B. Diselenium-bridged covalent organic framework with pH/GSH/photo-triple-responsiveness for highly controlled drug release toward joint chemo/photothermal/chemodynamic cancer therapy. J Mater Chem B 2022; 10:7955-7966. [PMID: 35792081 DOI: 10.1039/d2tb01015a] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Here, a novel joint chemo/photothermal/chemodynamic therapy was developed using a pH/GSH/photo triple-responsive 2D-covalent organic framework (COFs) drug carriers for passive target treatment of tumor with extraordinarily high efficiency. The well-designed...
Collapse
Affiliation(s)
- Han Lou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
- Department of Urology, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Shandong, P. R. China
| | - Lichao Chu
- Department of Anesthesiology, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Wenbin Zhou
- Department of Urology, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Shandong, P. R. China
| | - Jinli Dou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
| | - Xiaotong Teng
- Department of Respiratory Medicine, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Wei Tan
- Department of Respiratory Medicine, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
| |
Collapse
|
19
|
Blaszczak W, Swietach P. What do cellular responses to acidity tell us about cancer? Cancer Metastasis Rev 2021; 40:1159-1176. [PMID: 34850320 PMCID: PMC8825410 DOI: 10.1007/s10555-021-10005-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 12/20/2022]
Abstract
The notion that invasive cancer is a product of somatic evolution is a well-established theory that can be modelled mathematically and demonstrated empirically from therapeutic responses. Somatic evolution is by no means deterministic, and ample opportunities exist to steer its trajectory towards cancer cell extinction. One such strategy is to alter the chemical microenvironment shared between host and cancer cells in a way that no longer favours the latter. Ever since the first description of the Warburg effect, acidosis has been recognised as a key chemical signature of the tumour microenvironment. Recent findings have suggested that responses to acidosis, arising through a process of selection and adaptation, give cancer cells a competitive advantage over the host. A surge of research efforts has attempted to understand the basis of this advantage and seek ways of exploiting it therapeutically. Here, we review key findings and place these in the context of a mathematical framework. Looking ahead, we highlight areas relating to cellular adaptation, selection, and heterogeneity that merit more research efforts in order to close in on the goal of exploiting tumour acidity in future therapies.
Collapse
Affiliation(s)
- Wiktoria Blaszczak
- Department of Physiology, Anatomy & Genetics, Parks Road, Oxford, OX1 3PT, England
| | - Pawel Swietach
- Department of Physiology, Anatomy & Genetics, Parks Road, Oxford, OX1 3PT, England.
| |
Collapse
|
20
|
Corsale AM, Di Simone M, Lo Presti E, Picone C, Dieli F, Meraviglia S. Metabolic Changes in Tumor Microenvironment: How Could They Affect γδ T Cells Functions? Cells 2021; 10:2896. [PMID: 34831116 PMCID: PMC8616133 DOI: 10.3390/cells10112896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
The metabolic changes that occur in tumor microenvironment (TME) can influence not only the biological activity of tumor cells, which become more aggressive and auto sustained, but also the immune response against tumor cells, either producing ineffective responses or polarizing the response toward protumor activity. γδ T cells are a subset of T cells characterized by a plasticity that confers them the ability to differentiate towards different cell subsets according to the microenvironment conditions. On this basis, we here review the more recent studies focused on altered tumor metabolism and γδ T cells, considering their already known antitumor role and the possibility of manipulating their effector functions by in vitro and in vivo approaches. γδ T cells, thanks to their unique features, are themselves a valid alternative to overcome the limits associated with the use of conventional T cells, such as major histocompatibility complex (MHC) restriction, costimulatory signal and specific tumor-associated antigen recognition. Lipids, amino acids, hypoxia, prostaglandins and other metabolic changes inside the tumor microenvironment could reduce the efficacy of this important immune population and polarize γδ T cells toward IL17 producing cells that play a pro tumoral role. A deeper knowledge of this phenomenon could be helpful to formulate new immunotherapeutic approaches that target tumor metabolisms.
Collapse
Affiliation(s)
- Anna Maria Corsale
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90133 Palermo, Italy; (A.M.C.); (M.D.S.); (C.P.); (F.D.)
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy
| | - Marta Di Simone
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90133 Palermo, Italy; (A.M.C.); (M.D.S.); (C.P.); (F.D.)
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy
| | - Elena Lo Presti
- National Research Council (CNR), Institute for Biomedical Research and Innovation (IRIB), 90146 Palermo, Italy;
| | - Carmela Picone
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90133 Palermo, Italy; (A.M.C.); (M.D.S.); (C.P.); (F.D.)
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy
| | - Francesco Dieli
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90133 Palermo, Italy; (A.M.C.); (M.D.S.); (C.P.); (F.D.)
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy
| | - Serena Meraviglia
- Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, 90133 Palermo, Italy; (A.M.C.); (M.D.S.); (C.P.); (F.D.)
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
21
|
Multi-omics analysis of m 6A modification-related patterns based on m 6A regulators and tumor microenvironment infiltration in lung adenocarcinoma. Sci Rep 2021; 11:20921. [PMID: 34686691 PMCID: PMC8536683 DOI: 10.1038/s41598-021-00272-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/30/2021] [Indexed: 01/10/2023] Open
Abstract
Epigenetic modifications, especially N6-methyladenosine (m6A) modification, play a key role in tumor microenvironment (TME) infiltration. However, the regulatory role of m6A modification in the TME of lung adenocarcinoma (LUAD) remains unclear. A total of 2506 patients with LUAD were included in the analysis and divided into different groups according to distinct m6A modification-related patterns based on 23 m6A regulators. A comprehensive analysis was performed to explore TME infiltration in different m6A modification-related patterns. Principal component analysis was performed to obtain the m6Ascore and to quantify m6A modification-related patterns in different individuals. Three distinct m6A modification-related patterns were identified by 23 m6A regulators. The pathway enrichment analysis showed that m6Acluster-A was associated with immune activation; m6Acluster-B was associated with carcinogenic activation; m6Acluster-C was prominently related to substance metabolism. M6Acluster-A was remarkably rich in TME-infiltrating immune cells and patients with this pattern showed a survival advantage. The m6Ascore could predict TME infiltration, tumor mutation burden (TMB), the effect of tumor immunotherapy, and the prognosis of patients in LUAD. High m6Ascore was characterized by increased TME infiltration, reduced TMB, and survival advantage. Patients with a high m6Ascore exhibited significantly improved clinical response to anti-cytotoxic T lymphocyte antigen-4 (anti-CTLA4) immunotherapy. This study explored the regulatory mechanisms of TME infiltration in LUAD. The comprehensive analysis of m6A modification-related patterns may contribute to the development of individualized immunotherapy and the improvement of the overall effectiveness of immunotherapy for LUAD patients.
Collapse
|
22
|
Du Z, Cai S, Yan D, Li H, Zhang X, Yang W, Cao J, Yi N, Tang Z. Development and Validation of a Radiosensitivity Prediction Model for Lower Grade Glioma Based on Spike-and-Slab Lasso. Front Oncol 2021; 11:701500. [PMID: 34395274 PMCID: PMC8363254 DOI: 10.3389/fonc.2021.701500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/16/2021] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose Lower grade glioma (LGG) is one of the leading causes of death world worldwide. We attempted to develop and validate a radiosensitivity model for predicting the survival of lower grade glioma by using spike-and-slab lasso Cox model. Methods In this research, differentially expressed genes based on tumor microenvironment was obtained to further analysis. Log-rank test was used to identify genes in patients who received radiotherapy and patients who did not receive radiotherapy, respectively. Then, spike-and-slab lasso was performed to select genes in patients who received radiotherapy. Finally, three genes (INA, LEPREL1 and PTCRA) were included in the model. A radiosensitivity-related risk score model was established based on overall rate of TCGA dataset in patients who received radiotherapy. The model was validated in TCGA dataset that PFS as endpoint and two CGGA datasets that OS as endpoint. A novel nomogram integrated risk score with age and tumor grade was developed to predict the OS of LGG patients. Results We developed and verified a radiosensitivity-related risk score model. The radiosensitivity-related risk score is served as an independent prognostic indicator. This radiosensitivity-related risk score model has prognostic prediction ability. Moreover, the nomogram integrated risk score with age and tumor grade was established to perform better for predicting 1, 3, 5-year survival rate. Conclusions This model can be used by clinicians and researchers to predict patient’s survival rates and achieve personalized treatment of LGG.
Collapse
Affiliation(s)
- Zixuan Du
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Shang Cai
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Derui Yan
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Huijun Li
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Xinyan Zhang
- School of Data Science and Analytics, Kennesaw State University, Kennesaw, GA, United States
| | - Wei Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jianping Cao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Nengjun Yi
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zaixiang Tang
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
23
|
Mainini F, De Santis F, Fucà G, Di Nicola M, Rivoltini L, Eccles M. Nanobiotechnology and Immunotherapy: Two Powerful and Cooperative Allies against Cancer. Cancers (Basel) 2021; 13:3765. [PMID: 34359665 PMCID: PMC8345046 DOI: 10.3390/cancers13153765] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
A number of novel cancer therapies have recently emerged that have rapidly moved from the bench to the clinic. Onco-immunotherapies, such as immune checkpoint blockade inhibitors and adoptive cell therapies, have revolutionized the field, since they provide a way to induce strong anti-tumor immune responses, which are able to fight cancer effectively. However, despite showing great efficacy in hematological and some solid tumors, unresponsiveness, development of therapy resistance and the development of serious adverse effects, limit their capacity to impact the vast majority of tumors. Nanoparticle-based delivery systems are versatile vehicles for a wide variety of molecular cargoes and provide an innovative strategy to improve conventional onco-immunotherapies. They can be finely tuned to release their contents in the tumor microenvironment, or to deliver combinations of adjuvants and antigens in the case of nanovaccines. In this review, we summarize the recent advancements in the field of nanobiotechnology, to remodel the tumor microenvironment and to enhance immunotherapies.
Collapse
Affiliation(s)
- Francesco Mainini
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Francesca De Santis
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Giovanni Fucà
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Massimo Di Nicola
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Michael Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
24
|
Sun X, Peng Y, Zhao J, Xie Z, Lei X, Tang G. Discovery and development of tumor glycolysis rate-limiting enzyme inhibitors. Bioorg Chem 2021; 112:104891. [PMID: 33940446 DOI: 10.1016/j.bioorg.2021.104891] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/13/2022]
Abstract
Tumor cells mainly provide necessary energy and substances for rapid cell growth through aerobic perglycolysis rather than oxidative phosphorylation. This phenomenon is called the "Warburg effect". The mechanism of glycolysis in tumor cells is more complicated, which is caused by the comprehensive regulation of multiple factors. Abnormal enzyme metabolism is one of the main influencing factors and inhibiting the three main rate-limiting enzymes in glycolysis is thought to be important strategy for cancer treatment. Therefore, numerous inhibitors of glycolysis rate-limiting enzyme have been developed in recent years, such as the latest HKII inhibitor and PKM2 inhibitor Pachymic acid (PA) and N-(4-(3-(3-(methylamino)-3-oxopropyl)-5-(4'-(trifluoromethyl)-[1,1'-biphenyl]-4-yl)-1H-pyrazol-1-yl)phenyl)propiolamide. The review focuses on source, structure-activity relationship, bioecological activity and mechanism of the three main rate-limiting enzymes inhibitors, and hopes to guide the future research on the design and synthesis of rate-limiting enzyme inhibitors.
Collapse
Affiliation(s)
- Xueyan Sun
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China
| | - Yijiao Peng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China
| | - Jingduo Zhao
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China
| | - Zhizhong Xie
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hengyang City, Hunan Province, PR China
| | - Xiaoyong Lei
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hengyang City, Hunan Province, PR China
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China; Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hengyang City, Hunan Province, PR China.
| |
Collapse
|
25
|
Kim S, Lee M, Song Y, Lee SY, Choi I, Park IS, Kim J, Kim JS, Kim KM, Seo HR. Argininosuccinate synthase 1 suppresses tumor progression through activation of PERK/eIF2α/ATF4/CHOP axis in hepatocellular carcinoma. J Exp Clin Cancer Res 2021; 40:127. [PMID: 33838671 PMCID: PMC8035787 DOI: 10.1186/s13046-021-01912-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common malignant cancers worldwide, and liver cancer has increased in mortality due to liver cancer because it was detected at an advanced stages in patients with liver dysfunction, making HCC a lethal cancer. Accordingly, we aim to new targets for HCC drug discovery using HCC tumor spheroids. METHODS Our comparative proteomic analysis of HCC cells grown in culture as monolayers (2D) and spheroids (3D) revealed that argininosuccinate synthase 1 (ASS1) expression was higher in 3D cells than in 2D cells due to upregulated endoplasmic reticulum (ER) stress responses. We investigated the clinical value of ASS1 in Korean patients with HCC. The mechanism underlying ASS1-mediated tumor suppression was investigated in HCC spheroids. ASS1-mediated improvement of chemotherapy efficiency was observed using high content screening in an HCC xenograft mouse model. RESULTS Studies of tumor tissue from Korean HCC patients showed that, although ASS1 expression was low in most samples, high levels of ASS1 were associated with favorable overall survival of patients. Here, we found that bidirectional interactions between ASS1 ER stress responses in HCC-derived multicellular tumor spheroids can limit HCC progression. ASS1 overexpression effectively inhibited tumor growth and enhanced the efficacy of in vitro and in vivo anti-HCC combination chemotherapy via activation of the PERK/eIF2α/ATF4/CHOP axis, but was not dependent on the status of p53 and arginine metabolism. CONCLUSIONS These results demonstrate the critical functional roles for the arginine metabolism-independent tumor suppressor activity of ASS1 in HCC and suggest that upregulating ASS1 in these tumors is a potential strategy in HCC cells with low ASS1 expression.
Collapse
Affiliation(s)
- Sanghwa Kim
- Cancer Biology Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400 Republic of Korea
| | - Minji Lee
- Cancer Biology Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400 Republic of Korea
| | - Yeonhwa Song
- Cancer Biology Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400 Republic of Korea
| | - Su-Yeon Lee
- Cancer Biology Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400 Republic of Korea
| | - Inhee Choi
- Medicinal Chemistry, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 South Korea
| | - I-Seul Park
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 South Korea
| | - Jiho Kim
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488 South Korea
| | - Jin-sun Kim
- Department of Gastroenterology, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Kang mo Kim
- Department of Gastroenterology, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Haeng Ran Seo
- Cancer Biology Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400 Republic of Korea
| |
Collapse
|
26
|
刘 皓, 胡 颖. [A Review of Celluar Senescence and Tumor Treatment]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2021; 52:176-181. [PMID: 33829688 PMCID: PMC10408921 DOI: 10.12182/20210360503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 11/23/2022]
Abstract
Cellular senescence is a permanent state of cell cycle arrest, combined with the acquisition of a variety of secretory phenotypes. In addition to apoptosis, the induction of cellular senescence is an important mechanism that chemo- and radiotherapies and some targeted therapies rely on to produce an anti-tumor effect. However, being a self-protective mechanism of cells, cellular senescence can produce both positive and negative effects in tumor treatment. It remains a challenge to effectively utilize the anti-tumor effect of cellular senescence while averting the pro-tumor effect. How to improve the sensitivity of tumor treatment and to prevent tumor recurrence and metastasis has become the bottleneck in cellular senescence research. We summarize in this review the "double-edged-sword" effect of cellular senescence in tumor treatment. We summarize and discuss the cell autonomous and non-autonomous mechanisms that senescent cells use to affect tumor treatment, hoping to provide information that will help improve the outcome of tumor treatment and promote further research in basic and clinical application of cellular senescence in tumor treatment.
Collapse
Affiliation(s)
- 皓 刘
- 哈尔滨工业大学 生命科学与技术学院 (哈尔滨 150000)School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, China
| | - 颖 胡
- 哈尔滨工业大学 生命科学与技术学院 (哈尔滨 150000)School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, China
| |
Collapse
|
27
|
Peng Y, Xiong R, Li Z, Peng J, Xie ZZ, Lei XY, He D, Tang G. Design, synthesis, and biological evaluation of 3',4',5'-trimethoxy evodiamine derivatives as potential antitumor agents. Drug Dev Res 2021; 82:1021-1032. [PMID: 33600007 DOI: 10.1002/ddr.21806] [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: 11/30/2020] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 11/12/2022]
Abstract
A series of compounds bearing 3',4',5'-trimethoxy module into the core structure of evodiamine were designed and synthesized. The synthesized compounds were screened in vitro for their antitumor potential. MTT results showed that compounds 14a-14c and 14i-14j had significant effects, with compound 14h being the most prominent, with an IC50 value of 3.3 ± 1.5 μM, which was lower than evodiamine and 5-Fu. Subsequent experiments further confirmed that compound 14h could inhibit cell proliferation and migration, and induce G2/M phase arrest to inhibit the proliferation of HGC-27 cells, which is consistent with the results of the cytotoxicity experiment. Besides, 14h could inhibit microtubule assembly and might kill tumor cells by inhibiting VEGF and glycolysis. All experimental results indicate that compound 14h might be a potential drug candidate for the treatment of gastric cancer and was worthy of further study.
Collapse
Affiliation(s)
- Yijiao Peng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| | - Runde Xiong
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| | - Zhen Li
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| | - Junmei Peng
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| | - Zhi-Zhong Xie
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| | - Xiao-Yong Lei
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| | - Dongxiu He
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang City, Hunan Province, China
| |
Collapse
|
28
|
Roma-Rodrigues C, Raposo LR, Valente R, Fernandes AR, Baptista PV. Combined cancer therapeutics-Tackling the complexity of the tumor microenvironment. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1704. [PMID: 33565269 DOI: 10.1002/wnan.1704] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
Cancer treatment has yet to find a "silver bullet" capable of selectively and effectively kill tumor cells without damaging healthy cells. Nanomedicine is a promising field that can combine several moieties in one system to produce a multifaceted nanoplatform. The tumor microenvironment (TME) is considered responsible for the ineffectiveness of cancer therapeutics and the difficulty in the translation from the bench to bed side of novel nanomedicines. A promising approach is the use of combinatorial therapies targeting the TME with the use of stimuli-responsive nanomaterials which would increase tumor targeting. Contemporary combined strategies for TME-targeting nanoformulations are based on the application of external stimuli therapies, such as photothermy, hyperthermia or ultrasounds, in combination with stimuli-responsive nanoparticles containing a core, usually composed by metal oxides or graphene, and a biocompatible stimuli-responsive coating layer that could also contain tumor targeting moieties and a chemotherapeutic agent to enhance the therapeutic efficacy. The obstacles that nanotherapeutics must overcome in the TME to accomplish an effective therapeutic cargo delivery and the proposed strategies for improved nanotherapeutics will be reviewed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Catarina Roma-Rodrigues
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Luís R Raposo
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rúben Valente
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V Baptista
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
29
|
Prasad S, Chandra A, Cavo M, Parasido E, Fricke S, Lee Y, D'Amone E, Gigli G, Albanese C, Rodriguez O, Del Mercato LL. Optical and magnetic resonance imaging approaches for investigating the tumour microenvironment: state-of-the-art review and future trends. NANOTECHNOLOGY 2021; 32:062001. [PMID: 33065554 DOI: 10.1088/1361-6528/abc208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The tumour microenvironment (TME) strongly influences tumorigenesis and metastasis. Two of the most characterized properties of the TME are acidosis and hypoxia, both of which are considered hallmarks of tumours as well as critical factors in response to anticancer treatments. Currently, various imaging approaches exist to measure acidosis and hypoxia in the TME, including magnetic resonance imaging (MRI), positron emission tomography and optical imaging. In this review, we will focus on the latest fluorescent-based methods for optical sensing of cell metabolism and MRI as diagnostic imaging tools applied both in vitro and in vivo. The primary emphasis will be on describing the current and future uses of systems that can measure intra- and extra-cellular pH and oxygen changes at high spatial and temporal resolution. In addition, the suitability of these approaches for mapping tumour heterogeneity, and assessing response or failure to therapeutics will also be covered.
Collapse
Affiliation(s)
- Saumya Prasad
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Anil Chandra
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Marta Cavo
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Erika Parasido
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
| | - Stanley Fricke
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
- Department of Radiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Yichien Lee
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Eliana D'Amone
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
- Department of Mathematics and Physics 'Ennio De Giorgi', University of Salento, via Arnesano, 73100, Lecce, Italy
| | - Chris Albanese
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
- Department of Radiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Olga Rodriguez
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| |
Collapse
|
30
|
Shi Y, Cai Y, Cao Y, Hong Z, Chai Y. Recent advances in microfluidic technology and applications for anti-cancer drug screening. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2020.116118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
31
|
Wu ZY, Shen W, Yue JQ, Yao WY, Liu SL, Jin YP, Dong P, Ma F, Wu XS, Gong W. Combining Immunoscore with Clinicopathologic Features in Cholangiocarcinoma: An Influential Prognostic Nomogram. Onco Targets Ther 2020; 13:11359-11376. [PMID: 33192071 PMCID: PMC7654544 DOI: 10.2147/ott.s274754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/21/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE The aim of this study was to determine the Immunoscore as an independent prognostic factor for cholangiocarcinoma and establish a useful prognostic model for postoperative patients. METHODS This retrospective study was performed to assess the correlation between the clinicopathological features, tumor immune microenvironment, and prognosis of 76 patients with cholangiocarcinoma. Multivariate analysis was used to identify independent factors significantly associated with local recurrence-free survival (LRFS) and overall survival (OS). Finally, we constructed a nomogram combining the Immunoscore with clinicopathologic features to predict postoperative recurrence and OS. RESULTS The present study showed that immune cell infiltration was negatively correlated with tumor size, peripheral vascular invasion, lymph node metastasis, and tumor staging. Kaplan-Meier curves indicated that a decreased Immunoscore was associated with poor prognosis. Multivariate analysis demonstrated that resection type, number of tumors, lymph node metastasis, TNM staging, and the Immunoscore were significantly associated with LRFS. For OS, the significantly correlated factors included resection type, peripheral vascular invasion, TNM staging, and the Immunoscore. Immunoscore was superior to TNM staging in predicting both LRFS and OS according to the receiver operating characteristic analysis. Based on the results of the Cox regression analysis, a prognostic nomogram for the postoperative recurrence of cholangiocarcinoma and OS of patients was established. CONCLUSION The results of this study suggest that the Immunoscore may be used as an independent predictor of postoperative recurrence and OS of patients with cholangiocarcinoma. The Immunoscore appears to offer distinct advantages over the TNM staging system. By combining the Immunoscore and clinicopathological features, the proposed nomogram provides a more accurate predictive tool for postoperative patients with cholangiocarcinoma.
Collapse
Affiliation(s)
- Zi-You Wu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| | - Wei Shen
- Shanghai Colorectal Cancer Research Center, Shanghai, People’s Republic of China
| | - Juan-Qing Yue
- Department of Pathology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Wen-Yan Yao
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| | - Shi-Lei Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| | - Yun-Peng Jin
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| | - Ping Dong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| | - Fei Ma
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| | - Xiang-Song Wu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| | - Wei Gong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, People’s Republic of China
| |
Collapse
|
32
|
Zhang X, Dong Y, Zhao M, Ding L, Yang X, Jing Y, Song Y, Chen S, Hu Q, Ni Y. ITGB2-mediated metabolic switch in CAFs promotes OSCC proliferation by oxidation of NADH in mitochondrial oxidative phosphorylation system. Am J Cancer Res 2020; 10:12044-12059. [PMID: 33204328 PMCID: PMC7667693 DOI: 10.7150/thno.47901] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022] Open
Abstract
Objectives: Integrins, the coordinator of extracellular and intracellular signaling, are often found to be aberrant in tumors and can reshape the tumor microenvironment. Although previous studies showed that integrin beta 2 (ITGB2) is important for host defense, its expression profile and role in tumors, especially in cancer associated fibroblasts (CAFs) are still unknown. Methods: Immunofluorescence stain and fluorescence activated cell sorting were used to analyze the ITGB2 expression profile in oral squamous cell carcinoma (OSCC). RT-PCR and western blot were used to compare ITGB2 expression in normal fibroblasts (NFs) and cancer associated fibroblasts (CAFs). Clinical data and function-based experiments were used to investigate the promoting tumor growth ability of ITGB2 expressing CAFs. Enhanced glycolysis activity was identified by using bioinformatics analyses and GC/MS assays. MCT1 knockdown OSCC cell lines were constructed to explore the pro-proliferative mechanisms of ITGB2 expressing CAFs in multiple in vitro and in vivo assays. Results: We found that CAFs exhibited significantly higher ITGB2 expression than the matched NFs. In addition, higher ITGB2 expression in CAFs was correlated with higher TNM stages and more Ki67+ tumor cells, indicating its ability to promote OSCC proliferation. Further, co-culture assay demonstrated that ITGB2-mediated lactate release in CAFs promoted OSCC cell proliferation. Mechanically, ITGB2 regulated PI3K/AKT/mTOR pathways to enhance glycolysis activity in CAFs. Accordingly, lactate derived from ITGB2-expressing CAFs was absorbed and metabolized in OSCC to generate NADH, which was then oxidized in the mitochondrial oxidative phosphorylation system (OXPHOS) to produce ATP. Notably, inhibiting the OXPHOS system with metformin delayed the proliferative capacity of OSCC cells cultured in the ITGB2-expressing CAFs medium. Conclusions: Our study uncovered the ITGB2high pro-tumoral CAFs that activated the PI3K/AKT/mTOR axis to promote tumor proliferation in OSCC by NADH oxidation in the mitochondrial oxidative phosphorylation system.
Collapse
|
33
|
Abstract
The study of cancer metabolism has evolved vastly beyond the remit of tumour proliferation and survival with the identification of the role of 'oncometabolites' in tumorigenesis. Simply defined, oncometabolites are conventional metabolites that, when aberrantly accumulated, have pro-oncogenic functions. Their discovery has led researchers to revisit the Warburg hypothesis, first postulated in the 1950s, of aberrant metabolism as an aetiological determinant of cancer. As such, the identification of oncometabolites and their utilization in diagnostics and prognostics, as novel therapeutic targets and as biomarkers of disease, are areas of considerable interest in oncology. To date, fumarate, succinate, L-2-hydroxyglutarate (L-2-HG) and D-2-hydroxyglutarate (D-2-HG) have been characterized as bona fide oncometabolites. Extensive metabolic reprogramming occurs during tumour initiation and progression in renal cell carcinoma (RCC) and three oncometabolites - fumarate, succinate and L-2-HG - have been implicated in this disease process. All of these oncometabolites inhibit a superfamily of enzymes known as α-ketoglutarate-dependent dioxygenases, leading to epigenetic dysregulation and induction of pseudohypoxic phenotypes, and also have specific pro-oncogenic capabilities. Oncometabolites could potentially be exploited for the development of novel targeted therapies and as biomarkers of disease.
Collapse
Affiliation(s)
- Cissy Yong
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
34
|
Bonifácio VDB. Ovarian Cancer Biomarkers: Moving Forward in Early Detection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:355-363. [PMID: 32130708 DOI: 10.1007/978-3-030-34025-4_18] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ovarian cancer is a silent cancer which rate survival mainly relays in early stage detection. The discovery of reliable ovarian cancer biomarkers plays a crucial role in the disease management and strongly impact in patient's prognosis and survival. Although having many limitations CA125 is a classical ovarian cancer biomarker, but current research using proteomic or metabolomic methodologies struggles to find alternative biomarkers, using non-invasive our relatively non-invasive sources such as urine, serum, plasma, tissue, ascites or exosomes. Metabolism and metabolites are key players in cancer biology and its importance in biomarkers discovery cannot be neglected. In this chapter we overview the state of art and the challenges facing the use and discovery of biomarkers and focus on ovarian cancer early detection.
Collapse
Affiliation(s)
- Vasco D B Bonifácio
- IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|