1
|
Sprokkerieft J, van der Beek JN, Spreafico F, Selle B, Chowdhury T, Graf N, Verschuur AC, Dandis R, Bex A, Geller JI, Tytgat GAM, van den Heuvel-Eibrink MM. Outcome after treatment with axitinib in children, young adults, and adults with renal cell carcinoma: a narrative review. Crit Rev Oncol Hematol 2024; 204:104523. [PMID: 39326645 DOI: 10.1016/j.critrevonc.2024.104523] [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: 05/03/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024] Open
Abstract
Renal cell carcinoma (RCC) is a very rare type of renal cancer in children and young adults. When metastasized or recurrent, no standards of care are available, and outcome is still poor. The tyrosine kinase inhibitor axitinib is approved for treatment of RCC in adults, but its effects in children and young adults with RCC remains unclear. Due to the histological and biological differences between children and adults, it is difficult to extrapolate knowledge on treatments from the adult to the pediatric and young adult setting. This paper summarizes the clinical characteristics and outcomes of patients with RCC who were treated with axitinib, with the aim to gain insight in the clinical efficacy of this compound in this young patient group.
Collapse
Affiliation(s)
- Julia Sprokkerieft
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Justine N van der Beek
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Barbara Selle
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Tanzina Chowdhury
- Pediatric Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Arnauld C Verschuur
- Department of Pediatric Hematology-Oncology, Hôpital d'Enfants de la Timone, APHM, Marseille, France
| | - Rana Dandis
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Axel Bex
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam 1066CX, the Netherlands; Division of Surgical and Interventional Science, The Royal Free London NHS Foundation Trust and UCL, London, UK
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, USA
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | |
Collapse
|
2
|
Yue C, Lu W, Fan S, Huang Z, Yang J, Dong H, Zhang X, Shang Y, Lai W, Li D, Dong T, Yuan A, Wu J, Kang L, Hu Y. Nanoparticles for inducing Gaucher disease-like damage in cancer cells. NATURE NANOTECHNOLOGY 2024; 19:1203-1215. [PMID: 38740934 DOI: 10.1038/s41565-024-01668-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 03/27/2024] [Indexed: 05/16/2024]
Abstract
Nutrient avidity is one of the most distinctive features of tumours. However, nutrient deprivation has yielded limited clinical benefits. In Gaucher disease, an inherited metabolic disorder, cells produce cholesteryl-glucoside which accumulates in lysosomes and causes cell damage. Here we develop a nanoparticle (AbCholB) to emulate natural-lipoprotein-carried cholesterol and initiate Gaucher disease-like damage in cancer cells. AbCholB is composed of a phenylboronic-acid-modified cholesterol (CholB) and albumin. Cancer cells uptake the nanoparticles into lysosomes, where CholB reacts with glucose and generates a cholesteryl-glucoside-like structure that resists degradation and aggregates into microscale crystals, causing Gaucher disease-like damage in a glucose-dependent manner. In addition, the nutrient-sensing function of mTOR is suppressed. It is observed that normal cells escape severe damage due to their inferior ability to compete for nutrients compared with cancer cells. This work provides a bioinspired strategy to selectively impede the metabolic action of cancer cells by taking advantage of their nutrient avidity.
Collapse
Affiliation(s)
- Chunyan Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Wenjing Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Shuxin Fan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Zhusheng Huang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Jiaying Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Xiaojun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Yuxin Shang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Wenjia Lai
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, China
| | - Dandan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Tiejun Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Lifeng Kang
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China.
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China.
| |
Collapse
|
3
|
McDonald RC, Fischer AH, Rusckowski M. Oxygen Sensor-Guided Fine Needle Biopsy Studies of Human Cancer Xenografts in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.596060. [PMID: 38854036 PMCID: PMC11160627 DOI: 10.1101/2024.05.27.596060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
An oxygen sensor-mounted fine-needle biopsy tool was used for in vivo measurement of oxygen levels in tumor xenografts. The system provides a means of measuring the oxygen content in harvested tumor tissue from specific locations. Oxygen in human tumor xenografts in a murine model was observed for over 1 min. Tissues were mapped in relation to oxygen tension (pO2) readings and sampled for conventional cytological examination. Careful modeling of the pO2 readings over 60 seconds yielded a diffusion coefficient for oxygen at the sensor tip, providing additional diagnostic information about the tissue before sampling. Oxygen level measurement may provide a useful adjunct to the use of biomarkers in tumor diagnosis.
Collapse
Affiliation(s)
| | | | - Mary Rusckowski
- University of Massachusetts Medical School, Associate Professor, Department of Radiology
| |
Collapse
|
4
|
Zhang SW, Wang H, Ding XH, Xiao YL, Shao ZM, You C, Gu YJ, Jiang YZ. Bidirectional crosstalk between therapeutic cancer vaccines and the tumor microenvironment: Beyond tumor antigens. FUNDAMENTAL RESEARCH 2023; 3:1005-1024. [PMID: 38933006 PMCID: PMC11197801 DOI: 10.1016/j.fmre.2022.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 11/20/2022] Open
Abstract
Immunotherapy has rejuvenated cancer therapy, especially after anti-PD-(L)1 came onto the scene. Among the many therapeutic options, therapeutic cancer vaccines are one of the most essential players. Although great progress has been made in research on tumor antigen vaccines, few phase III trials have shown clinical benefits. One of the reasons lies in obstruction from the tumor microenvironment (TME). Meanwhile, the therapeutic cancer vaccine reshapes the TME in an ambivalent way, leading to immune stimulation or immune escape. In this review, we summarize recent progress on the interaction between therapeutic cancer vaccines and the TME. With respect to vaccine resistance, innate immunosuppressive TME components and acquired resistance caused by vaccination are both involved. Understanding the underlying mechanism of this crosstalk provides insight into the treatment of cancer by directly targeting the TME or synergizing with other therapeutics.
Collapse
Affiliation(s)
- Si-Wei Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Han Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiao-Hong Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Ya-Jia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
5
|
Khadka S, Lin YH, Ackroyd J, Chen YA, Sheng Y, Qian W, Guo S, Chen Y, Behr E, Barekatain Y, Uddin N, Arthur K, Yan V, Hsu WH, Chang Q, Poral A, Tran T, Chaurasia S, Georgiou DK, Asara JM, Barthel FP, Millward SW, DePinho RA, Muller FL. Anaplerotic nutrient stress drives synergy of angiogenesis inhibitors with therapeutics targeting tumor metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.07.539744. [PMID: 37214825 PMCID: PMC10197573 DOI: 10.1101/2023.05.07.539744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Tumor angiogenesis is a cancer hallmark, and its therapeutic inhibition has provided meaningful, albeit limited, clinical benefit. While anti-angiogenesis inhibitors deprive the tumor of oxygen and essential nutrients, cancer cells activate metabolic adaptations to diminish therapeutic response. Despite these adaptations, angiogenesis inhibition incurs extensive metabolic stress, prompting us to consider such metabolic stress as an induced vulnerability to therapies targeting cancer metabolism. Metabolomic profiling of angiogenesis-inhibited intracranial xenografts showed universal decrease in tricarboxylic acid cycle intermediates, corroborating a state of anaplerotic nutrient deficit or stress. Accordingly, we show strong synergy between angiogenesis inhibitors (Avastin, Tivozanib) and inhibitors of glycolysis or oxidative phosphorylation through exacerbation of anaplerotic nutrient stress in intracranial orthotopic xenografted gliomas. Our findings were recapitulated in GBM xenografts that do not have genetically predisposed metabolic vulnerabilities at baseline. Thus, our findings cement the central importance of the tricarboxylic acid cycle as the nexus of metabolic vulnerabilities and suggest clinical path hypothesis combining angiogenesis inhibitors with pharmacological cancer interventions targeting tumor metabolism for GBM tumors.
Collapse
Affiliation(s)
- Sunada Khadka
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Yu-Hsi Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey Ackroyd
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Yi-An Chen
- Cancer and Cell Biology Division, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Yanghui Sheng
- Crown Bioscience Inc., Suzhou Industrial Park, 218 Xinghu Rd, Jiangsu, China
| | - Wubin Qian
- Crown Bioscience Inc., Suzhou Industrial Park, 218 Xinghu Rd, Jiangsu, China
| | - Sheng Guo
- Crown Bioscience Inc., Suzhou Industrial Park, 218 Xinghu Rd, Jiangsu, China
| | - Yining Chen
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eliot Behr
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasaman Barekatain
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Nasir Uddin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenisha Arthur
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victoria Yan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Wen-Hao Hsu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anton Poral
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Theresa Tran
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Surendra Chaurasia
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dimitra K Georgiou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Asara
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Floris P Barthel
- Cancer and Cell Biology Division, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Steve W Millward
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florian L Muller
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
- Present address: Sporos Bioventures, Houston, TX, USA
| |
Collapse
|
6
|
Tan KF, In LLA, Vijayaraj Kumar P. Surface Functionalization of Gold Nanoparticles for Targeting the Tumor Microenvironment to Improve Antitumor Efficiency. ACS APPLIED BIO MATERIALS 2023; 6:2944-2981. [PMID: 37435615 DOI: 10.1021/acsabm.3c00202] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Gold nanoparticles (AuNPs) have undergone significant research for their use in the treatment of cancer. Numerous researchers have established their potent antitumor properties, which have greatly impacted the treatment of cancer. AuNPs have been used in four primary anticancer treatment modalities, namely radiation, photothermal therapy, photodynamic therapy, and chemotherapy. However, the ability of AuNPs to destroy cancer is lacking and can even harm healthy cells without the right direction to transport them to the tumor microenvironment. Consequently, a suitable targeting technique is needed. Based on the distinct features of the human tumor microenvironment, this review discusses four different targeting strategies that target the four key features of the tumor microenvironment, including abnormal vasculature, overexpression of specific receptors, an acidic microenvironment, and a hypoxic microenvironment, to direct surface-functionalized AuNPs to the tumor microenvironment and increase antitumor efficacies. In addition, some current completed or ongoing clinical trials of AuNPs will also be discussed below to further reinforce the concept of using AuNPs in anticancer therapy.
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
7
|
Pringels L, Cook JL, Witvrouw E, Burssens A, Vanden Bossche L, Wezenbeek E. Exploring the role of intratendinous pressure in the pathogenesis of tendon pathology: a narrative review and conceptual framework. Br J Sports Med 2023; 57:1042-1048. [PMID: 36323498 PMCID: PMC10423488 DOI: 10.1136/bjsports-2022-106066] [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] [Accepted: 10/13/2022] [Indexed: 02/07/2023]
Abstract
Despite the high prevalence of tendon pathology in athletes, the underlying pathogenesis is still poorly understood. Various aetiological theories have been presented and rejected in the past, but the tendon cell response model still holds true. This model describes how the tendon cell is the key regulator of the extracellular matrix and how pathology is induced by a failed adaptation to a disturbance of tissue homeostasis. Such failure has been attributed to various kinds of stressors (eg, mechanical, thermal and ischaemic), but crucial elements seem to be missing to fully understand the pathogenesis. Importantly, a disturbance of tissue pressure homeostasis has not yet been considered a possible factor, despite it being associated with numerous pathologies. Therefore, we conducted an extensive narrative literature review on the possible role of intratendinous pressure in the pathogenesis of tendon pathology. This review explores the current understanding of pressure dynamics and the role of tissue pressure in the pathogenesis of other disorders with structural similarities to tendons. By bridging these insights with known structural changes that occur in tendon pathology, a conceptual model was constituted. This model provides an overview of the possible mechanism of how an increase in intratendinous pressure might be involved in the development and progression of tendon pathology and contribute to tendon pain. In addition, some therapies that could reduce intratendinous pressure and accelerate tendon healing are proposed. Further experimental research is encouraged to investigate our hypotheses and to initiate debate on the relevance of intratendinous pressure in tendon pathology.
Collapse
Affiliation(s)
- Lauren Pringels
- Department of Physical and Rehabilitation Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
| | - Jill L Cook
- La Trobe Sport and Exercise Medicine Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Erik Witvrouw
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
| | - Arne Burssens
- Department of Orthopaedic Surgery, Ghent University Hospital, Ghent, Belgium
| | - Luc Vanden Bossche
- Department of Physical and Rehabilitation Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
| | - Evi Wezenbeek
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
| |
Collapse
|
8
|
Zhou C, Huang YQ, Da MX, Jin WL, Zhou FH. Adipocyte-derived extracellular vesicles: bridging the communications between obesity and tumor microenvironment. Discov Oncol 2023; 14:92. [PMID: 37289328 PMCID: PMC10250291 DOI: 10.1007/s12672-023-00704-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023] Open
Abstract
By the year 2035 more than 4 billion people might be affected by obesity and being overweight. Adipocyte-derived Extracellular Vesicles (ADEVs/ADEV-singular) are essential for communication between the tumor microenvironment (TME) and obesity, emerging as a prominent mechanism of tumor progression. Adipose tissue (AT) becomes hypertrophic and hyperplastic in an obese state resulting in insulin resistance in the body. This modifies the energy supply to tumor cells and simultaneously stimulates the production of pro-inflammatory adipokines. In addition, obese AT has a dysregulated cargo content of discharged ADEVs, leading to elevated amounts of pro-inflammatory proteins, fatty acids, and carcinogenic microRNAs. ADEVs are strongly associated with hallmarks of cancer (proliferation and resistance to cell death, angiogenesis, invasion, metastasis, immunological response) and may be useful as biomarkers and antitumor therapy strategy. Given the present developments in obesity and cancer-related research, we conclude by outlining significant challenges and significant advances that must be addressed expeditiously to promote ADEVs research and clinical applications.
Collapse
Affiliation(s)
- Chuan Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| | - Yu-Qian Huang
- Department of Center of Medical Cosmetology, Chengdu Second People’s Hospital, Chengdu, 610017 People’s Republic of China
| | - Ming-Xu Da
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| | - Wei-Lin Jin
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Feng-Hai Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Department of Urology, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| |
Collapse
|
9
|
Bobiński R, Dutka M, Pizon M, Waksmańska W, Pielesz A. Ferroptosis, Acyl Starvation, and Breast Cancer. Mol Pharmacol 2023; 103:132-144. [PMID: 36750321 DOI: 10.1124/molpharm.122.000607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/31/2022] [Indexed: 12/14/2022] Open
Abstract
To maintain their growth rate, cancer cells must secure a supply of fatty acids, which are necessary for building cell membranes and maintaining energy processes. This is one of the reasons why tissues with intensive fatty acid metabolism, such as the mammary gland, are more likely to develop tumors. One natural or induced defense process against cancer is ferroptosis, which interferes with normal fatty acid metabolism. This leads to the oxidation of polyunsaturated fatty acids, which causes a rearrangement of the metabolism and damages cell membranes. As a consequence of this oxidation, there is a shortage of normal polyunsaturated fatty acids, which disturbs the complicated metabolism of fatty acids. This imbalance in metabolism, resulting from the deficiency of properly structured fatty acids, is called, by these authors, "acyl starvation." When cancer cells are exposed to alternating hypoxia and reoxygenation, they often develop resistance to neoadjuvant therapies. Blocking the stearoyl-CoA desaturase - fatty acid-binding protein 4 - fatty acid translocase axis appears to be a promising pathway in the treatment of breast cancer. On the one hand, the inhibition of desaturase leads to the formation of toxic phospholipid hydroperoxides in ferroptosis, whereas on the other hand, the inhibition of fatty acid-binding protein 4 and translocase leads to a reduced uptake of fatty acids and disruption of the cellular transport of fatty acids, resulting in intracellular acyl starvation. The disruption in the metabolism of fatty acids in cancer cells may augment the effectiveness of neoadjuvant therapy. SIGNIFICANCE STATEMENT: Regulation of the metabolism of fatty acids in cancer cells seems to be a promising therapeutic direction. Studies show that the induction of ferroptosis in cancer cells, combined with use of neoadjuvant therapies, effectively inhibits the proliferation of these cells. We link the process of ferroptosis with apoptosis and SCD1-FABP4-CD36 axis and propose the term "acyl starvation" for the processes leading to FA deficiency, dysregulation of FA metabolism in cancer cells, and, most importantly, the appearance of incorrect proportions FAs.
Collapse
Affiliation(s)
- Rafał Bobiński
- Department of Biochemistry and Molecular Biology (R.B., M.D., W.W.) and Department of Microbiology and Environmental Technology, Institute of Engineering and Environmental Protection (A.P.), University of Bielsko-Biala, Bielsko-Biala, Poland; and Department of Research and Development, Transfusion Center Bayreuth, Bayreuth, Germany (M.P.)
| | - Mieczysław Dutka
- Department of Biochemistry and Molecular Biology (R.B., M.D., W.W.) and Department of Microbiology and Environmental Technology, Institute of Engineering and Environmental Protection (A.P.), University of Bielsko-Biala, Bielsko-Biala, Poland; and Department of Research and Development, Transfusion Center Bayreuth, Bayreuth, Germany (M.P.)
| | - Monika Pizon
- Department of Biochemistry and Molecular Biology (R.B., M.D., W.W.) and Department of Microbiology and Environmental Technology, Institute of Engineering and Environmental Protection (A.P.), University of Bielsko-Biala, Bielsko-Biala, Poland; and Department of Research and Development, Transfusion Center Bayreuth, Bayreuth, Germany (M.P.)
| | - Wioletta Waksmańska
- Department of Biochemistry and Molecular Biology (R.B., M.D., W.W.) and Department of Microbiology and Environmental Technology, Institute of Engineering and Environmental Protection (A.P.), University of Bielsko-Biala, Bielsko-Biala, Poland; and Department of Research and Development, Transfusion Center Bayreuth, Bayreuth, Germany (M.P.)
| | - Anna Pielesz
- Department of Biochemistry and Molecular Biology (R.B., M.D., W.W.) and Department of Microbiology and Environmental Technology, Institute of Engineering and Environmental Protection (A.P.), University of Bielsko-Biala, Bielsko-Biala, Poland; and Department of Research and Development, Transfusion Center Bayreuth, Bayreuth, Germany (M.P.)
| |
Collapse
|
10
|
Tu X, Zhang J, Yuan W, Wu X, Xu Z, Qing C. Simvastatin Enhanced Anti-tumor Effects of Bevacizumab against Lung Adenocarcinoma A549 Cells via Abating HIF-1α-Wnt/β-Catenin Signaling Pathway. Anticancer Agents Med Chem 2023; 23:2083-2094. [PMID: 37587804 DOI: 10.2174/1871520623666230816090914] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Bevacizumab increased hypoxia-inducible factor (HIF-1α) expression attenuates its antitumor effect. Simvastatin can reduce the expression of HIF-1α to exert a tumor-suppressive effect in many in vitro experiments. Therefore, this study aimed to determine whether simvastatin could strengthen the anti-tumor activity of bevacizumab in lung adenocarcinoma. OBJECTIVE To determine whether simvastatin could strengthen the anti-tumor activity of bevacizumab in lung adenocarcinoma. METHODS The changes in the biological behavior of A549 cells treated with different drugs were determined through colony forming assay, Cell Counting Assay-8 (CCK-8), transwell assay, wound healing assay, and flow cytometry. The expressions of pathway-related factors HIF-1α and β-Catenin were determined via qRT-PCR and western blotting. The expressions of proliferation-related proteins, invasion-related proteins, and apoptosis-related proteins were detected by western blotting. In addition, a xenograft non-small cell lung cancer model in nude mice was used to explore in vivo tumor growth. RESULTS We found that simvastatin combined with bevacizumab synergistically suppressed the proliferation, migration, and invasion of A549 cells while promoting their apoptosis. As demonstrated by qRT-PCR and western blotting experiments, the bevacizumab group displayed a higher expression of pathway-related factors HIF-1α and β-Catenin than the control groups, however simvastatin group showed the opposite trend. Its combination with bevacizumab induced elevation of HIF-1α and β-catenin expressions. During in vivo experiments, simvastatin inhibited tumor growth, and in comparison, the inhibitory effects of its combination with bevacizumab were stronger. CONCLUSION Based on our findings, simvastatin may affect the biological responses of bevacizumab on A549 cells by restraining the HIF-1α-Wnt/β-catenin signaling pathway, thus representing a novel and effective combination therapy that can be potentially applied in a clinical therapy for lung adenocarcinoma.
Collapse
Affiliation(s)
- Xin Tu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jian Zhang
- Department of Gastroenterology, The Second People's Hospital of Yibin, Yibin, Sichuan, People's Republic of China
| | - Wei Yuan
- Department of Neurology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Xia Wu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Zhi Xu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Cuo Qing
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
11
|
Liu Q, Guan C, Liu C, Li H, Wu J, Sun C. Targeting hypoxia-inducible factor-1alpha: A new strategy for triple-negative breast cancer therapy. Biomed Pharmacother 2022; 156:113861. [DOI: 10.1016/j.biopha.2022.113861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/02/2022] Open
|
12
|
Zalpoor H, Aziziyan F, Liaghat M, Bakhtiyari M, Akbari A, Nabi-Afjadi M, Forghaniesfidvajani R, Rezaei N. The roles of metabolic profiles and intracellular signaling pathways of tumor microenvironment cells in angiogenesis of solid tumors. Cell Commun Signal 2022; 20:186. [PMID: 36419156 PMCID: PMC9684800 DOI: 10.1186/s12964-022-00951-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/06/2022] [Indexed: 11/27/2022] Open
Abstract
Innate and adaptive immune cells patrol and survey throughout the human body and sometimes reside in the tumor microenvironment (TME) with a variety of cell types and nutrients that may differ from those in which they developed. The metabolic pathways and metabolites of immune cells are rooted in cell physiology, and not only provide nutrients and energy for cell growth and survival but also influencing cell differentiation and effector functions. Nowadays, there is a growing awareness that metabolic processes occurring in cancer cells can affect immune cell function and lead to tumor immune evasion and angiogenesis. In order to safely treat cancer patients and prevent immune checkpoint blockade-induced toxicities and autoimmunity, we suggest using anti-angiogenic drugs solely or combined with Immune checkpoint blockers (ICBs) to boost the safety and effectiveness of cancer therapy. As a consequence, there is significant and escalating attention to discovering techniques that target metabolism as a new method of cancer therapy. In this review, a summary of immune-metabolic processes and their potential role in the stimulation of intracellular signaling in TME cells that lead to tumor angiogenesis, and therapeutic applications is provided. Video abstract.
Collapse
Affiliation(s)
- Hamidreza Zalpoor
- grid.412571.40000 0000 8819 4698Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Fatemeh Aziziyan
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran ,grid.412266.50000 0001 1781 3962Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahsa Liaghat
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran ,Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Maryam Bakhtiyari
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran ,grid.412606.70000 0004 0405 433XDepartment of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Abdullatif Akbari
- grid.412571.40000 0000 8819 4698Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mohsen Nabi-Afjadi
- grid.412266.50000 0001 1781 3962Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Razieh Forghaniesfidvajani
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran ,grid.411705.60000 0001 0166 0922Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran ,grid.411705.60000 0001 0166 0922Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Charbonneau M, Harper K, Brochu-Gaudreau K, Perreault A, McDonald PP, Ekindi-Ndongo N, Jeldres C, Dubois CM. Establishment of a ccRCC patient-derived chick chorioallantoic membrane model for drug testing. Front Med (Lausanne) 2022; 9:1003914. [PMID: 36275794 PMCID: PMC9582329 DOI: 10.3389/fmed.2022.1003914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is an aggressive subtype of renal cell carcinoma accounting for the majority of deaths in kidney cancer patients. Advanced ccRCC has a high mortality rate as most patients progress and develop resistance to currently approved targeted therapies, highlighting the ongoing need for adequate drug testing models to develop novel therapies. Current animal models are expensive and time-consuming. In this study, we investigated the use of the chick chorioallantoic membrane (CAM), a rapid and cost-effective model, as a complementary drug testing model for ccRCC. Our results indicated that tumor samples from ccRCC patients can be successfully cultivated on the chick chorioallantoic membrane (CAM) within 7 days while retaining their histopathological characteristics. Furthermore, treatment of ccRCC xenografts with sunitinib, a tyrosine kinase inhibitor used for the treatment of metastatic RCC, allowed us to evaluate differential responses of individual patients. Our results indicate that the CAM model is a complementary in vivo model that allows for rapid and cost-effective evaluation of ccRCC patient response to drug therapy. Therefore, this model has the potential to become a useful platform for preclinical evaluation of new targeted therapies for the treatment of ccRCC.
Collapse
Affiliation(s)
- Martine Charbonneau
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kelly Harper
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Karine Brochu-Gaudreau
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Alexis Perreault
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | - Claudio Jeldres
- Division of Urology, Department of Surgery, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Claire M. Dubois
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada,*Correspondence: Claire M. Dubois
| |
Collapse
|
14
|
Ivan M, Fishel ML, Tudoran OM, Pollok KE, Wu X, Smith PJ. Hypoxia signaling: Challenges and opportunities for cancer therapy. Semin Cancer Biol 2022; 85:185-195. [PMID: 34628029 PMCID: PMC8986888 DOI: 10.1016/j.semcancer.2021.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/26/2022]
Abstract
Hypoxia is arguably the first recognized cancer microenvironment hallmark and affects virtually all cellular populations present in tumors. During the past decades the complex adaptive cellular responses to oxygen deprivation have been largely elucidated, raising hope for new anti cancer agents. Despite undeniable preclinical progress, therapeutic targeting of tumor hypoxia is yet to transition from bench to bedside. This review focuses on new pharmacological agents that exploit tumor hypoxia or interfere with hypoxia signaling and discusses strategies to maximize their therapeutic impact.
Collapse
Affiliation(s)
- Mircea Ivan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Melissa L Fishel
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology and Toxicology, IU Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Oana M Tudoran
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Cluj, Romania
| | - Karen E Pollok
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xue Wu
- Ohio State University, Columbus, OH, USA
| | - Paul J Smith
- School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
15
|
Wang S, Li L, Zuo S, Kong L, Wei J, Dong J. Metabolic-related gene pairs signature analysis identifies ABCA1 expression levels on tumor-associated macrophages as a prognostic biomarker in primary IDHWT glioblastoma. Front Immunol 2022; 13:869061. [PMID: 36248907 PMCID: PMC9561761 DOI: 10.3389/fimmu.2022.869061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/14/2022] [Indexed: 11/26/2022] Open
Abstract
Background Although isocitrate dehydrogenase (IDH) mutation serves as a prognostic signature for routine clinical management of glioma, nearly 90% of glioblastomas (GBM) patients have a wild-type IDH genotype (IDHWT) and lack reliable signatures to identify distinct entities. Methods To develop a robust prognostic signature for IDHWT GBM patients, we retrospectively analyzed 4 public datasets of 377 primary frozen tumor tissue transcriptome profiling and clinical follow-up data. Samples were divided into a training dataset (204 samples) and a validation (173 samples) dataset. A prognostic signature consisting of 21 metabolism-related gene pairs (MRGPs) was developed based on the relative ranking of single-sample gene expression levels. GSEA and immune subtype analyses were performed to reveal differences in biological processes between MRGP risk groups. The single-cell RNA-seq dataset was used to examine the expression distribution of each MRG constituting the signature in tumor tissue subsets. Finally, the association of MRGs with tumor progression was biologically validated in orthotopic GBM models. Results The metabolic signature remained an independent prognostic factor (hazard ratio, 5.71 [3.542-9.218], P < 0.001) for stratifying patients into high- and low-risk levels in terms of overall survival across subgroups with MGMTp methylation statuses, expression subtypes, and chemo/ratio therapies. Immune-related biological processes were significantly different between MRGP risk groups. Compared with the low-risk group, the high-risk group was significantly enriched in humoral immune responses and phagocytosis processes, and had more monocyte infiltration and less activated DC, NK, and γδ T cell infiltration. scRNA-seq dataset analysis identified that the expression levels of 5 MRGs (ABCA1, HMOX1, MTHFD2, PIM1, and PTPRE) in TAMs increased with metabolic risk. With tumor progression, the expression level of ABCA1 in TAMs was positively correlated with the population of TAMs in tumor tissue. Downregulation of ABCA1 levels can promote TAM polarization towards an inflammatory phenotype and control tumor growth. Conclusions The metabolic signature is expected to be used in the individualized management of primary IDHWT GBM patients.
Collapse
Affiliation(s)
- Shiqun Wang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Lu Li
- Department of Nephrology, Affiliated Children’s Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuguang Zuo
- Liuzhou Key Laboratory of Molecular Diagnosis, Guangxi Key Laboratory of Molecular Diagnosis and Application, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Lingkai Kong
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jiwu Wei
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- *Correspondence: Jie Dong, ; Jiwu Wei,
| | - Jie Dong
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- *Correspondence: Jie Dong, ; Jiwu Wei,
| |
Collapse
|
16
|
Nakayama H, Ohnuki H, Nakahara M, Nishida-Fukuda H, Sakaue T, Fukuda S, Higashiyama S, Doi Y, Mitsuyoshi M, Okimoto T, Tosato G, Kusumoto C. Inactivation of axon guidance molecule netrin-1 in human colorectal cancer by an epigenetic mechanism. Biochem Biophys Res Commun 2022; 611:146-150. [PMID: 35489200 DOI: 10.1016/j.bbrc.2022.04.069] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 01/16/2023]
Abstract
Netrin-1, the protein product of the NTN1 gene, is an axon guidance molecule implicated in regulation of cell survival and tumorigenesis. Expression of the netrin-1 receptors deleted in colorectal cancer (DCC) and uncoordinated 5 homolog (UNC5H) is frequently silenced in colorectal cancer (CRC) by either loss of heterozygosity or epigenetic mechanisms. However, netrin-1 expression and regulation in CRC are mostly unknown. Here, we report that NTN1 expression is significantly reduced in most CRC tissues compared to the adjacent normal intestinal mucosa, and that NTN1 DNA methylation is significantly higher in CRCs (24.6%) than in the adjacent normal intestinal mucosa (4.0%). In 6 CRC cell lines, NTN1 expression is low. Treatment with 5-Aza-2'-deoxycytidine increased expression of NTN1 in CRC cell lines, indicating that DNA methylation represses NTN1 transcription in CRCs. NTN1 DNA hypermethylation was significantly associated with advanced CRC disease. Median netrin-1 serum levels were significantly decreased in CRC patients (330.1 pg/mL) compared with normal individuals (438.6 pg/mL). Our results suggest that netrin-1 is a candidate biomarker for CRC.
Collapse
Affiliation(s)
- Hironao Nakayama
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, Hiroshima, 739-2695, Japan.
| | - Hidetaka Ohnuki
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Masako Nakahara
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, Hiroshima, 739-2695, Japan
| | - Hisayo Nishida-Fukuda
- Department of Genome Editing, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, 5731010, Japan
| | - Tomohisa Sakaue
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Shitsukawa, Ehime, 791-0295, Japan; Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Shitsukawa, Ehime, 791-0295, Japan
| | - Shinji Fukuda
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Shitsukawa, Ehime, 791-0295, Japan; Department of Biochemistry, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, 464-8650, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Shitsukawa, Ehime, 791-0295, Japan; Department of Molecular and Cellular Biology, Osaka International Cancer Institute, Chuo-ku, Osaka, 541-8567, Japan
| | - Yuki Doi
- Department of Surgery, Nippon Kokan Fukuyama Hospital, Fukuyama, Hiroshima, 721-0927, Japan
| | - Masahiro Mitsuyoshi
- Department of Surgery, Nippon Kokan Fukuyama Hospital, Fukuyama, Hiroshima, 721-0927, Japan; Department of Surgery I, School of Medicine, University of Occupational and Environmental Health, Kita-kyushu, Fukuoka, 807-0804, Japan
| | - Takashi Okimoto
- Department of Surgery, Nippon Kokan Fukuyama Hospital, Fukuyama, Hiroshima, 721-0927, Japan; Department of Surgery, Kyushu Rosai Hospital, Kita-kyushu, Fukuoka, 800-0296, Japan
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chiaki Kusumoto
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, Hiroshima, 739-2695, Japan; Department of Gastroenterology, Nippon Kokan Fukuyama Hospital, Fukuyama, Hiroshima, 721-0927, Japan.
| |
Collapse
|
17
|
O'Connor MN, Kallenberg DM, Camilli C, Pilotti C, Dritsoula A, Jackstadt R, Bowers CE, Watson HA, Alatsatianos M, Ohme J, Dowsett L, George J, Blackburn JWD, Wang X, Singhal M, Augustin HG, Ager A, Sansom OJ, Moss SE, Greenwood J. LRG1 destabilizes tumor vessels and restricts immunotherapeutic potency. MED 2021; 2:1231-1252.e10. [PMID: 35590198 PMCID: PMC7614757 DOI: 10.1016/j.medj.2021.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 09/02/2021] [Accepted: 10/05/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND A poorly functioning tumor vasculature is pro-oncogenic and may impede the delivery of therapeutics. Normalizing the vasculature, therefore, may be beneficial. We previously reported that the secreted glycoprotein leucine-rich α-2-glycoprotein 1 (LRG1) contributes to pathogenic neovascularization. Here, we investigate whether LRG1 in tumors is vasculopathic and whether its inhibition has therapeutic utility. METHODS Tumor growth and vascular structure were analyzed in subcutaneous and genetically engineered mouse models in wild-type and Lrg1 knockout mice. The effects of LRG1 antibody blockade as monotherapy, or in combination with co-therapies, on vascular function, tumor growth, and infiltrated lymphocytes were investigated. FINDINGS In mouse models of cancer, Lrg1 expression was induced in tumor endothelial cells, consistent with an increase in protein expression in human cancers. The expression of LRG1 affected tumor progression as Lrg1 gene deletion, or treatment with a LRG1 function-blocking antibody, inhibited tumor growth and improved survival. Inhibition of LRG1 increased endothelial cell pericyte coverage and improved vascular function, resulting in enhanced efficacy of cisplatin chemotherapy, adoptive T cell therapy, and immune checkpoint inhibition (anti-PD1) therapy. With immunotherapy, LRG1 inhibition led to a significant shift in the tumor microenvironment from being predominantly immune silent to immune active. CONCLUSIONS LRG1 drives vascular abnormalization, and its inhibition represents a novel and effective means of improving the efficacy of cancer therapeutics. FUNDING Wellcome Trust (206413/B/17/Z), UKRI/MRC (G1000466, MR/N006410/1, MC/PC/14118, and MR/L008742/1), BHF (PG/16/50/32182), Health and Care Research Wales (CA05), CRUK (C42412/A24416 and A17196), ERC (ColonCan 311301 and AngioMature 787181), and DFG (CRC1366).
Collapse
Affiliation(s)
- Marie N O'Connor
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - David M Kallenberg
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Carlotta Camilli
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Camilla Pilotti
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Athina Dritsoula
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Rene Jackstadt
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Chantelle E Bowers
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - H Angharad Watson
- Division of Infection and Immunity, School of Medicine and Systems Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Markella Alatsatianos
- Division of Infection and Immunity, School of Medicine and Systems Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Julia Ohme
- Division of Infection and Immunity, School of Medicine and Systems Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Laura Dowsett
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Jestin George
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Jack W D Blackburn
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Xiaomeng Wang
- Institute of Ophthalmology, University College London, London SE5 8BN, UK
| | - Mahak Singhal
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ann Ager
- Division of Infection and Immunity, School of Medicine and Systems Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Stephen E Moss
- Institute of Ophthalmology, University College London, London SE5 8BN, UK.
| | - John Greenwood
- Institute of Ophthalmology, University College London, London SE5 8BN, UK.
| |
Collapse
|
18
|
Kumar D, Patel SA, Khan R, Chawla S, Mohapatra N, Dixit M. IQ Motif-Containing GTPase-Activating Protein 2 Inhibits Breast Cancer Angiogenesis By Suppressing VEGFR2-AKT Signaling. Mol Cancer Res 2021; 20:77-91. [PMID: 34615693 DOI: 10.1158/1541-7786.mcr-20-1044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/17/2021] [Accepted: 10/01/2021] [Indexed: 12/24/2022]
Abstract
Antiangiogenesis cancer therapies are facing setbacks due to side effects and resistance. Parallel targeting of multiple pathways can help in the development of more effective therapies. This requires the discovery of new molecules that can regulate multiple cellular processes. Our study has recently established the association of reduced IQGAP2 expression in breast cancer with EMT and poor prognosis of the patient. Existing literature indirectly suggests the role of IQGAP2 in angiogenesis that is still unexplored. In this study, we searched the role of IQGAP2 in tumor angiogenesis in a comprehensive manner using cell culture, patients, and animal models. Depletion of IQGAP2 in breast cancer cells increased proliferation, migration, and tubulogenesis of HUVECs. Findings were validated in ex ovo CAM, Matrigel plug and skin wound-healing assays in mouse model, showing that the reduction of IQGAP2 significantly increased angiogenesis. As a confirmation, IHC analysis of the patient's tissues showed a negative correlation of IQGAP2 expression with the microvessel density. Mechanistically, loss of IQGAP2 appeared to activate VEGF-A via ERK activation in tumor cells, which activated the VEGFR2-AKT axis in HUVECs. IMPLICATIONS: The findings of this study suggest the antiangiogenic properties of IQGAP2 in breast cancer. The Dual effect of IQGAP2 on EMT and angiogenesis makes it a potential target for anticancer therapy.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, HBNI, Khurda, Odisha, India
| | - Saket Awadhesbhai Patel
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, HBNI, Khurda, Odisha, India
| | - Rehan Khan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, HBNI, Khurda, Odisha, India
| | - Saurabh Chawla
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, HBNI, Khurda, Odisha, India
| | | | - Manjusha Dixit
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, HBNI, Khurda, Odisha, India.
| |
Collapse
|
19
|
Rationally designed drug delivery systems for the local treatment of resected glioblastoma. Adv Drug Deliv Rev 2021; 177:113951. [PMID: 34461201 DOI: 10.1016/j.addr.2021.113951] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023]
Abstract
Glioblastoma (GBM) is a particularly aggressive brain cancer associated with high recurrence and poor prognosis. The standard of care, surgical resection followed by concomitant radio- and chemotherapy, leads to low survival rates. The local delivery of active agents within the tumor resection cavity has emerged as an attractive means to initiate oncological treatment immediately post-surgery. This complementary approach bypasses the blood-brain barrier, increases the local concentration at the tumor site while reducing or avoiding systemic side effects. This review will provide a global overview on the local treatment for GBM with an emphasis on the lessons learned from past clinical trials. The main parameters to be considered to rationally design fit-of-purpose biomaterials and develop drug delivery systems for local administration in the GBM resection cavity to prevent the tumor recurrence will be described. The intracavitary local treatment of GBM should i) use materials that facilitate translation to the clinic; ii) be characterized by easy GMP effective scaling up and easy-handling application by the neurosurgeons; iii) be adaptable to fill the tumor-resected niche, mold to the resection cavity or adhere to the exposed brain parenchyma; iv) be biocompatible and possess mechanical properties compatible with the brain; v) deliver a therapeutic dose of rationally-designed or repurposed drug compound(s) into the GBM infiltrative margin. Proof of concept with high translational potential will be provided. Finally, future perspectives to facilitate the clinical translation of the local perisurgical treatment of GBM will be discussed.
Collapse
|
20
|
Turkez H, Tozlu OO, Arslan ME, Mardinoglu A. Safety and Efficacy Assessments to Take Antioxidants in Glioblastoma Therapy: From In Vitro Experiences to Animal and Clinical Studies. Neurochem Int 2021; 150:105168. [PMID: 34450218 DOI: 10.1016/j.neuint.2021.105168] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM) is considered one of the most common malignant brain tumors, occurring as over 15% of all primary central nervous system and brain neoplasms. The unique and standard treatment option towards GBM involves the combination of surgical resection followed by radiotherapy (RT) and chemotherapy (CT). However, due to the aggressive nature and heterogeneity of GBMs, they remained difficult to treat. Recent findings from preclinical studies have revealed that disruption of the redox balance via using either oxidative or anti-oxidative agents in GBM presented an effective and promising therapeutic approach. A limited number of clinical trials substantially encouraged their concomitant use with RT or CT. Thus, treatment of GBMs may benefit from natural or synthetic antioxidative compounds as novel therapeutics. Despite the presence of variegated in vitro and in vivo studies focusing on safety and efficacy issues of these promising therapeutics, nowadays their translation to clinics is far from applicability due to several challenges. In this review, we briefly introduce the enzymatic and non-enzymatic antioxidant defense systems as well as potential signaling pathways related to the pathogenesis of GBM with a special interest in antioxidant mechanisms. In addition, we describe the advantages and limitations of antioxidant supplementation in GBM cases or disease models as well as growing challenges for GBM therapies with antioxidants in the future.
Collapse
Affiliation(s)
- Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Ataturk University, 25240, Erzurum, Turkey
| | - Ozlem Ozdemir Tozlu
- Department of Molecular Biology and Genetics, Faculty of Science, 25250; Erzurum Technical University, Erzurum, Turkey
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Faculty of Science, 25250; Erzurum Technical University, Erzurum, Turkey
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK; Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, SE-17121, Sweden.
| |
Collapse
|
21
|
Hirschmüller A, Morath O. [Tendinopathies of the Achilles tendon]. Z Rheumatol 2021; 80:629-640. [PMID: 34287670 DOI: 10.1007/s00393-021-01006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2021] [Indexed: 10/20/2022]
Abstract
Disorders of the Achilles tendon are among the most frequent musculoskeletal injuries in athletes as well as in the general population. It is very important to differentiate the different clinical pictures summarized under the general term achillodynia and to understand the pathogenesis in order to undertake the correct therapeutic measures. In the case of insertional tendinopathies in particular, a rheumatological origin should be clarified. Doppler ultrasound is the most important diagnostic tool. Evidence-based treatment methods include various training programs, shock wave treatment, diverse injection and surgical procedures, each of which are discussed in detail in this article.
Collapse
Affiliation(s)
- Anja Hirschmüller
- Altius Swiss Sportmed Center Ag, Habich-Dietschy-Str. 5a, 4310, Rheinfelden, Schweiz. .,Klinik für Orthopädie und Unfallchirurgie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Deutschland.
| | - Oliver Morath
- Institut Bewegungs- und Arbeitsmedizin, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Deutschland
| |
Collapse
|
22
|
Moraes JA, Encarnação C, Franco VA, Xavier Botelho LG, Rodrigues GP, Ramos-Andrade I, Barja-Fidalgo C, Renovato-Martins M. Adipose Tissue-Derived Extracellular Vesicles and the Tumor Microenvironment: Revisiting the Hallmarks of Cancer. Cancers (Basel) 2021; 13:3328. [PMID: 34283044 PMCID: PMC8268128 DOI: 10.3390/cancers13133328] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are crucial elements that sustain the communication between tumor cells and their microenvironment, and have emerged as a widespread mechanism of tumor formation and metastasis. In obesity, the adipose tissue becomes hypertrophic and hyperplastic, triggering increased production of pro-inflammatory adipokines, such as tumor necrosis factor α, interleukin 6, interleukin 1, and leptin. Furthermore, obese adipose tissue undergoes dysregulation in the cargo content of the released EVs, resulting in an increased content of pro-inflammatory proteins, fatty acids, and oncogenic microRNAs. These alterations drive obesity-associated inflammatory responses both locally and systemically. After being ignored for a long time, adipose tissues have recently received considerable attention as a major player in tumor microenvironment-linked obesity and cancer. The role of adipose tissue in the establishment and progression of cancer is reinforced by its high plasticity and inflammatory content. Such a relationship may be established by direct contact between adipocytes and cancer cells within the microenvironment or systemically, via EV-mediated cell-to-cell communication. Here, we highlight cues evidencing the influence of adipose tissue-derived EVs on the hallmarks of cancer, which are critical for tumor malignancy.
Collapse
Affiliation(s)
- João Alfredo Moraes
- Redox Biology Laboratory, Programa de Pesquisa em Farmacologia e Inflamação, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil;
| | - Carol Encarnação
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Victor Aguiar Franco
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Luiz Gabriel Xavier Botelho
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Gabriella Pacheco Rodrigues
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Isadora Ramos-Andrade
- Laboratory of Cellular and Molecular Pharmacology, Departamento de Biologia Celular, IBRAG, Universidade do Estado do Rio de Janeiro, 20550-170 Rio de Janeiro, Brazil; (I.R.-A.); (C.B.-F.)
| | - Christina Barja-Fidalgo
- Laboratory of Cellular and Molecular Pharmacology, Departamento de Biologia Celular, IBRAG, Universidade do Estado do Rio de Janeiro, 20550-170 Rio de Janeiro, Brazil; (I.R.-A.); (C.B.-F.)
| | - Mariana Renovato-Martins
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| |
Collapse
|
23
|
Grande E, Rodriguez-Antona C, López C, Alonso-Gordoa T, Benavent M, Capdevila J, Teulé A, Custodio A, Sevilla I, Hernando J, Gajate P, Molina-Cerrillo J, Díez JJ, Santos M, Lanillos J, García-Carbonero R. Sunitinib and Evofosfamide (TH-302) in Systemic Treatment-Naïve Patients with Grade 1/2 Metastatic Pancreatic Neuroendocrine Tumors: The GETNE-1408 Trial. Oncologist 2021; 26:941-949. [PMID: 34190375 DOI: 10.1002/onco.13885] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/23/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Sunitinib (SUN)-induced hypoxia within the tumor could promote the activation of the prodrug evofosfamide (EVO), locally releasing the cytotoxic DNA alkylator bromo-isophosphoramide mustard. SUNEVO, a phase II, open-label, single-arm trial, investigated the potential synergy of SUN plus EVO in advanced progressive pancreatic neuroendocrine tumors (panNETs). METHODS Systemic treatment-naïve patients with advanced or metastatic, unresectable, grade 1/2 panNETs with a Ki67 ≤20%, received EVO 340 mg/m2 on days 8, 15, and 22 every 4 weeks and sunitinib 37.5 mg/day continuously. The primary endpoint was objective response rate, measured every 8 weeks by RECIST version 1.1. RESULTS From 2015 to 2018, 17 patients were enrolled. The median age was 62.4 years, 47% had a Ki67 >10%, and 70.6% had liver metastasis. Patients received a median of five and four cycles of SUN and EVO, respectively. After a median follow-up of 15.7 months, 17.6% of patients achieved a complete (n = 1) or partial response (n = 2), and 11 patients had stable disease (64.7%). The median progression-free survival was 10.4 months (95% confidence interval, 2.6-18.0). Treatment-related adverse events (grade ≥3) were observed in 64.7% of the patients, the most frequent being neutropenia (35.3%), fatigue (17.6%), and thrombopenia (11.8%). Treatment discontinuation due to toxicity was reported in 88.2% of the patients. No correlation was found between treatment response and DAXX, ATRX, MEN1, SETD2, and PTEN gene mutations. CONCLUSION SUN plus EVO had a negative toxicity profile that should be taken into account for further clinical research in advanced panNETs. The combination showed moderate activity in terms of treatment response that did not correlate with somatic mutations. (Clinical trial identification number: NCT02402062) IMPLICATIONS FOR PRACTICE: Addition of hypoxia-activated prodrugs has been proposed as a potential mechanism to overcome tumor resistance to antiangiogenic agents. Sunitinib and evofosfamide, which were widely proposed as a potential synergistic option, showed modest efficacy in pancreatic neuroendocrine tumors (panNETs), reaching a median objective response rate of 17.6% and median progression-free survival of 10.4 months. Treatment response does not correlate with the biomarkers analyzed. The high systemic toxicity, with 88.2% of patients discontinuing the treatment, makes this therapeutic approach unfeasible and encourages future research to overcome panNETs' resistance to antiangiogenic agents with other therapies with a safer profile.
Collapse
Affiliation(s)
- Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Cristina Rodriguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Carlos López
- Medical Oncology, Hospital Universitario Marqués de Valdecilla, Instituto de investigación sanitaria Valdecilla (IDIVAL), Santander, Spain
| | | | - Marta Benavent
- Medical Oncology Department, Hospital Virgen del Rocío/Instituto de Biomedicina de Sevilla (IBIS), Seville, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Vall Hebron University Hospital, Vall Hebron Institute of Oncology, Barcelona, Spain
| | - Alex Teulé
- Institut Català d'Oncologia (ICO)-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Ana Custodio
- Medical Oncology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Isabel Sevilla
- Investigación Clínica y Traslacional en Cáncer/Instituto de Investigaciones Biomédicas de Málaga (IBIMA)/Hospitales Universitarios Regional y Virgen de la Victoria, Málaga, Spain
| | - Jorge Hernando
- Medical Oncology Department, Vall Hebron University Hospital, Vall Hebron Institute of Oncology, Barcelona, Spain
| | - Pablo Gajate
- Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Juan José Díez
- Department of Endocrinology, Hospital Universitario Puerta de Hierro Majadahonda, Instituto de Investigación Sanitaria Puerta de Hierro Segovia de Arana, Madrid, Spain
| | - María Santos
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Javier Lanillos
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Rocío García-Carbonero
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación 12 de Octubre (i+12), Universidad Complutense de Madrid (UCM), Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
24
|
Tumor resistance to ferroptosis driven by Stearoyl-CoA Desaturase-1 (SCD1) in cancer cells and Fatty Acid Biding Protein-4 (FABP4) in tumor microenvironment promote tumor recurrence. Redox Biol 2021; 43:102006. [PMID: 34030117 PMCID: PMC8163990 DOI: 10.1016/j.redox.2021.102006] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Problem Tumor recurrence is a major clinical issue that represents the principal cause of cancer-related deaths, with few targetable common pathways. Mechanisms by which residual tumors persist and progress under a continuous shift between hypoxia-reoxygenation after neoadjuvent-therapy are unknown. In this study, we investigated the role of lipid metabolism and tumor redox balance in tumor recurrence. Methods Lipidomics, proteomics and mass spectrometry imaging approaches where applied to mouse tumor models of recurrence. Genetic and pharmacological inhibitions of lipid mediators in tumors were used in vivo and in functional assays in vitro. Results We found that stearoyl-CoA desaturase-1 (SCD1) expressed by cancer cells and fatty acid binding protein-4 (FABP4) produced by tumor endothelial cells (TECs) and adipocytes in the tumor microenvironment (TME) are essential for tumor relapse in response to tyrosine kinase inhibitors (TKI) and chemotherapy. SCD1 and FABP4 were also found upregulated in recurrent human breast cancer samples and correlated with worse prognosis of cancer patients with different types of tumors. Mechanistically, SCD1 leads to fatty acid (FA) desaturation and FABP4 derived from TEM enhances lipid droplet (LD) in cancer cells, which cooperatively protect from oxidative stress-induced ferroptosis. We revealed that lipid mobilization and desaturation elicit tumor intrinsic antioxidant and anti-ferroptotic resources for survival and regrowth in a harsh TME. Inhibition of lipid transport from TME by FABP4 inhibitor reduced tumor regrowth and by genetic — or by pharmacological — targeting SCD1 in vivo, tumor regrowth was abolished completely. Conclusion This finding unveils that it is worth taking advantage of tumor lipid addiction, as a tumor vulnerability to design novel treatment strategy to prevent cancer recurrence. Increased oxidative stress markers and lipid metabolism in residual tumors. Expression of SCD1 in cancer cells and FABP4 in the tumor microenvironment drive tumor recurrence. Fatty acid desaturation by SCD1 and lipid transport by FABP4 confer resistance to ROS and ferroptosis. Blocking SCD1 and FABP4 sensitized cancer cells to ROS-induced ferroptosis and reduced tumor recurrence.
Collapse
|
25
|
Lee ST, Muralidharan V, Tebbutt N, Wong P, Fang C, Liu Z, Gan H, Sachinidis J, Pathmaraj K, Christophi C, Scott AM. Prevalence of hypoxia and correlation with glycolytic metabolism and angiogenic biomarkers in metastatic colorectal carcinoma. Eur J Nucl Med Mol Imaging 2021; 48:1585-1592. [PMID: 33125527 DOI: 10.1007/s00259-020-05074-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Hypoxia is associated with aggressive tumour behaviour and can influence response to systemic therapy and radiotherapy. The prevalence of hypoxia in metastatic colorectal cancer is poorly understood, and the relationship of hypoxia to patient outcomes has not been clearly established. The aims of the study were to evaluate hypoxia in metastatic colorectal cancer with [18F]Fluoromisonidazole ([18F]FMISO PET) and correlate these findings with glycolytic metabolism ([18F]FDG PET) and angiogenic blood biomarkers and patient outcomes. METHODS Patients with metastatic colorectal cancer received routine staging investigations and both [18F] FMISO PET and [18F] FDG PET scans. Correlative blood specimens were also obtained at the time of the [18F] FMISO PET scan. Patient follow-up was performed to establish progression-free survival. RESULTS A total of 40 patients were recruited into the trial. [18F]FMISO and [18F]FDG PET scans showed a significant correlation of SUVmax (p = 0.003). A significant correlation of progression-free survival and [18F] FMISO TNR (p = 0.02) and overall survival with [18F]FMISO TNR (p = 0.003) and [18F]FDG TGV (p = 0.02) was observed. Serum levels of osteopontin, but not VEGF, correlated with [18F] FMISO and [18F]FDG PET scan parameters. CONCLUSION [18F]FMISO PET uptake in metastatic colorectal cancer significantly correlates with glycolytic metabolism and is predictive of progression-free and overall survival. These findings have implications for the assessment and treatment of metastatic colorectal cancer patients with novel therapies which affect tumour angiogenesis and hypoxia.
Collapse
Affiliation(s)
- S T Lee
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, Australia.
- Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia.
| | - V Muralidharan
- Department of Surgery, Austin Health, Melbourne, Australia
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, Australia
| | - N Tebbutt
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, Australia
- Department of Medical Oncology, Austin Health, Melbourne, Australia
| | - P Wong
- Department of Surgery, Austin Health, Melbourne, Australia
| | - C Fang
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Australia
| | - Z Liu
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Australia
| | - H Gan
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Department of Medical Oncology, Austin Health, Melbourne, Australia
| | - J Sachinidis
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - K Pathmaraj
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - C Christophi
- Department of Surgery, Austin Health, Melbourne, Australia
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, Australia
| | - A M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia
| |
Collapse
|
26
|
Cignarella A, Fadini GP, Bolego C, Trevisi L, Boscaro C, Sanga V, Seccia TM, Rosato A, Rossi GP, Barton M. Clinical Efficacy and Safety of Angiogenesis Inhibitors: Sex Differences and Current Challenges. Cardiovasc Res 2021; 118:988-1003. [PMID: 33739385 DOI: 10.1093/cvr/cvab096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Vasoactive molecules, such as vascular endothelial growth factor (VEGF) and endothelins, share cytokine-like activities and regulate endothelial cell (EC) growth, migration and inflammation. Some endothelial mediators and their receptors are targets for currently approved angiogenesis inhibitors, drugs that are either monoclonal antibodies raised towards VEGF, or inhibitors of vascular receptor protein kinases and signaling pathways. Pharmacological interference with the protective functions of ECs results in a similar spectrum of adverse effects. Clinically, the most common side effects of VEGF signaling pathway inhibition include an increase in arterial pressure, left ventricular (LV) dysfunction ultimately causing heart failure, and thromboembolic events, including pulmonary embolism, stroke, and myocardial infarction. Sex steroids such as androgens, progestins, and estrogen and their receptors (ERα, ERβ, GPER; PR-A, PR-B; AR) have been identified as important modifiers of angiogenesis, and sex differences have been reported for anti-angiogenic drugs. This review article discusses the current challenges clinicians are facing with regard to angiogenesis inhibitor treatments, including the need to consider sex differences affecting clinical efficacy and safety. We also propose areas for future research taking into account the role of sex hormone receptors and sex chromosomes. Development of new sex-specific drugs with improved target and cell-type selectivity likely will open the way personalized medicine in men and women requiring antiangiogenic therapy and result in reduced adverse effects and improved therapeutic efficacy.
Collapse
Affiliation(s)
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Viola Sanga
- Department of Medicine, University of Padova, Italy
| | | | - Antonio Rosato
- Venetian Cancer Institute IOV - IRCCS, Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
| | | | - Matthias Barton
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy.,Molecular Internal Medicine, University of Zürich, Switzerland.,Andreas Grüntzig Foundation, Zürich, Switzerland
| |
Collapse
|
27
|
Hypoxia-Driven Effects in Cancer: Characterization, Mechanisms, and Therapeutic Implications. Cells 2021; 10:cells10030678. [PMID: 33808542 PMCID: PMC8003323 DOI: 10.3390/cells10030678] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Hypoxia, a common feature of solid tumors, greatly hinders the efficacy of conventional cancer treatments such as chemo-, radio-, and immunotherapy. The depletion of oxygen in proliferating and advanced tumors causes an array of genetic, transcriptional, and metabolic adaptations that promote survival, metastasis, and a clinically malignant phenotype. At the nexus of these interconnected pathways are hypoxia-inducible factors (HIFs) which orchestrate transcriptional responses under hypoxia. The following review summarizes current literature regarding effects of hypoxia on DNA repair, metastasis, epithelial-to-mesenchymal transition, the cancer stem cell phenotype, and therapy resistance. We also discuss mechanisms and pathways, such as HIF signaling, mitochondrial dynamics, exosomes, and the unfolded protein response, that contribute to hypoxia-induced phenotypic changes. Finally, novel therapeutics that target the hypoxic tumor microenvironment or interfere with hypoxia-induced pathways are reviewed.
Collapse
|
28
|
Bernauer C, Man YKS, Chisholm JC, Lepicard EY, Robinson SP, Shipley JM. Hypoxia and its therapeutic possibilities in paediatric cancers. Br J Cancer 2021; 124:539-551. [PMID: 33106581 PMCID: PMC7851391 DOI: 10.1038/s41416-020-01107-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
In tumours, hypoxia-a condition in which the demand for oxygen is higher than its availability-is well known to be associated with reduced sensitivity to radiotherapy and chemotherapy, and with immunosuppression. The consequences of hypoxia on tumour biology and patient outcomes have therefore led to the investigation of strategies that can alleviate hypoxia in cancer cells, with the aim of sensitising cells to treatments. An alternative therapeutic approach involves the design of prodrugs that are activated by hypoxic cells. Increasing evidence indicates that hypoxia is not just clinically significant in adult cancers but also in paediatric cancers. We evaluate relevant methods to assess the levels and extent of hypoxia in childhood cancers, including novel imaging strategies such as oxygen-enhanced magnetic resonance imaging (MRI). Preclinical and clinical evidence largely supports the use of hypoxia-targeting drugs in children, and we describe the critical need to identify robust predictive biomarkers for the use of such drugs in future paediatric clinical trials. Ultimately, a more personalised approach to treatment that includes targeting hypoxic tumour cells might improve outcomes in subgroups of paediatric cancer patients.
Collapse
Affiliation(s)
- Carolina Bernauer
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK
| | - Y K Stella Man
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK
| | - Julia C Chisholm
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Surrey, UK
- Sarcoma Clinical Trials in Children and Young People Team, The Institute of Cancer Research, London, UK
| | - Elise Y Lepicard
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Janet M Shipley
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK.
| |
Collapse
|
29
|
Emami Nejad A, Najafgholian S, Rostami A, Sistani A, Shojaeifar S, Esparvarinha M, Nedaeinia R, Haghjooy Javanmard S, Taherian M, Ahmadlou M, Salehi R, Sadeghi B, Manian M. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int 2021; 21:62. [PMID: 33472628 PMCID: PMC7816485 DOI: 10.1186/s12935-020-01719-5] [Citation(s) in RCA: 309] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Hypoxia is a common feature of solid tumors, and develops because of the rapid growth of the tumor that outstrips the oxygen supply, and impaired blood flow due to the formation of abnormal blood vessels supplying the tumor. It has been reported that tumor hypoxia can: activate angiogenesis, thereby enhancing invasiveness and risk of metastasis; increase survival of tumor, as well as suppress anti-tumor immunity and hamper the therapeutic response. Hypoxia mediates these effects by several potential mechanisms: altering gene expression, the activation of oncogenes, inactivation of suppressor genes, reducing genomic stability and clonal selection. We have reviewed the effects of hypoxia on tumor biology and the possible strategiesto manage the hypoxic tumor microenvironment (TME), highlighting the potential use of cancer stem cells in tumor treatment.
Collapse
Affiliation(s)
- Asieh Emami Nejad
- Department of Biology, Payame Noor University (PNU), P.O.Box 19395-3697, Tehran, Iran
| | - Simin Najafgholian
- Department of Emergency Medicine, School of Medicine , Arak University of Medical Sciences, Arak, Iran
| | - Alireza Rostami
- Department of Surgery, School of Medicine Amiralmomenin Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Alireza Sistani
- Department of Emergency Medicine, School of Medicine Valiasr Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Samaneh Shojaeifar
- Department of Midwifery, Faculty of Nursing and Midwifery , Arak University of Medical Sciences , Arak, Iran
| | - Mojgan Esparvarinha
- Department of Immunology, School of Medicine , Tabriz University of Medical Sciences , Tabriz, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease , Isfahan University of Medical Sciences , Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Marjan Taherian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Ahmadlou
- Sciences Medical of University Arak, Hospital Amiralmomenin, Center Development Research Clinical, Arak, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease , Isfahan University of Medical Sciences , Isfahan, Iran.,Department of Genetics and Molecular Biology, School of Medicine , Isfahan University of Medical Sciences , Isfahan, Iran
| | - Bahman Sadeghi
- Department of Health and Community Medicine, School of Medicine, Arak University of Medical Sciences, Arak, 3848176341, Iran.
| | - Mostafa Manian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Department of Medical Laboratory Science, Faculty of Medical Science Kermanshah Branch, Islamic Azad University, Imam Khomeini Campus, Farhikhtegan Bld., Shahid J'afari St., Kermanshah, 3848176341, Iran.
| |
Collapse
|
30
|
Xie W, Zhao H, Wang F, Wang Y, He Y, Wang T, Zhang K, Yang H, Zhou Z, Shi H, Wang J, Huang G. A novel humanized Frizzled-7-targeting antibody enhances antitumor effects of Bevacizumab against triple-negative breast cancer via blocking Wnt/β-catenin signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:30. [PMID: 33436039 PMCID: PMC7802198 DOI: 10.1186/s13046-020-01800-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/03/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Anti-angiogenic therapy has been widely applied to the clinical treatment of malignant tumors. However, the efficacy of such treatments has been called into question, especially in triple-negative breast cancer (TNBC). Bevacizumab, the first anti-angiogenic agent approved by FDA, actually increases invasive and metastatic properties of TNBC cells, resulting from the activation of Wnt/β-catenin signaling in response to hypoxia. As a critical receptor of Wnt/β-catenin signaling, Frizzled-7 (Fzd7) is aberrantly expressed in TNBC, indicating Fzd7 a potential target for developing drugs to be combined with anti-angiogenic agents. METHODS Hybridoma technique and antibody humanization technique were utilized to generate a Fzd7-targeting antibody (SHH002-hu1). Biolayer interferometry (BLI) assay and near infrared (NIR) imaging were conducted to detect the affinity and targeting ability of SHH002-hu1. Next, whether SHH002-hu1 could suppress the invasion and migration of TNBC cells induced by Bevacizumab were validated, and the underlying molecular mechanisms were elucidated by luciferase reporter and western blot assays. The nude-mice transplanted TNBC models were established to assess the anti-TNBC activities of SHH002-hu1 when combined with Bevacizumab. Then, the effects on putative TNBC stem-like cells and Wnt/β-catenin signaling were evaluated by immunofluorescence (IF). Further, the tumor-initiating and self-renew capacity of TNBC cells were studied by secondary nude mouse xenograft model and sphere formation assay. In addition, the effects of SHH002-hu1 on the adaptation of TNBC cells to hypoxia were evaluated by the detection of vasculogenic mimicry (VM) and hypoxia-inducible factor-1α (HIF-1α) transcriptional activity. RESULTS The novel humanized antibody targeting Fzd7 (SHH002-hu1) exhibited extremely high affinity with Fzd7, and specifically targeted to Fzd7+ cells and tumor tissues. SHH002-hu1 repressed invasion, migration and epithelial-mesenchymal cell transformation (EMT) of TNBC cells induced by Bevacizumab through abating Wnt/β-catenin signaling. SHH002-hu1 significantly enhanced the capacity of Bevacizumab to inhibit the growth of TNBC via reducing the subpopulation of putative TNBC stem-like cells, further attenuating Bevacizumab-enhanced tumor-initiating and self-renew capacity of TNBC cells. Moreover, SHH002-hu1 effectively restrained the adaptation of TNBC cells to hypoxia via disrupting Wnt/β-catenin signaling. CONCLUSION SHH002-hu1 significantly enhances the anti-TNBC capacity of Bevacizumab, and shows the potential of preventing TNBC recurrence, suggesting SHH002-hu1 a good candidate for the synergistic therapy together with Bevacizumab.
Collapse
Affiliation(s)
- Wei Xie
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China. .,School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China.
| | - Huijie Zhao
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| | - Fengxian Wang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| | - Yiyun Wang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| | - Yuan He
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Tong Wang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China
| | - Kunchi Zhang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China
| | - Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China
| | - Zhaoli Zhou
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China
| | - Haibin Shi
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China.,State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, People's Republic of China
| | - Jin Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China.
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Pudong New Area, Shanghai, China.
| |
Collapse
|
31
|
Fendt SM, Frezza C, Erez A. Targeting Metabolic Plasticity and Flexibility Dynamics for Cancer Therapy. Cancer Discov 2020; 10:1797-1807. [PMID: 33139243 PMCID: PMC7710573 DOI: 10.1158/2159-8290.cd-20-0844] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/06/2020] [Accepted: 09/02/2020] [Indexed: 11/16/2022]
Abstract
Cancer cells continuously rewire their metabolism to fulfill their need for rapid growth and survival while subject to changes in environmental cues. Thus, a vital component of a cancer cell lies in its metabolic adaptability. The constant demand for metabolic alterations requires flexibility, that is, the ability to utilize different metabolic substrates; as well as plasticity, that is, the ability to process metabolic substrates in different ways. In this review, we discuss how dynamic changes in cancer metabolism affect tumor progression and the consequential implications for cancer therapy. SIGNIFICANCE: Recognizing cancer dynamic metabolic adaptability as an entity can lead to targeted therapy that is expected to decrease drug resistance.
Collapse
Affiliation(s)
- Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
32
|
Li T, Mao C, Wang X, Shi Y, Tao Y. Epigenetic crosstalk between hypoxia and tumor driven by HIF regulation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:224. [PMID: 33109235 PMCID: PMC7592369 DOI: 10.1186/s13046-020-01733-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Hypoxia is the major influence factor in physiological and pathological courses which are mainly mediated by hypoxia-inducible factors (HIFs) in response to low oxygen tensions within solid tumors. Under normoxia, HIF signaling pathway is inhibited due to HIF-α subunits degradation. However, in hypoxic conditions, HIF-α is activated and stabilized, and HIF target genes are successively activated, resulting in a series of tumour-specific activities. The activation of HIFs, including HIF-1α, HIF-2α and HIF-3α, subsequently induce downstream target genes which leads to series of responses, the resulting abnormal processes or metabolites in turn affect HIFs stability. Given its functions in tumors progression, HIFs have been regarded as therapeutic targets for improved treatment efficacy. Epigenetics refers to alterations in gene expression that are stable between cell divisions, and sometimes between generations, but do not involve changes in the underlying DNA sequence of the organism. And with the development of research, epigenetic regulation has been found to play an important role in the development of tumors, which providing accumulating basic or clinical evidences for tumor treatments. Here, given how little has been reported about the overall association between hypoxic tumors and epigenetics, we made a more systematic review from epigenetic perspective in hope of helping others better understand hypoxia or HIF pathway, and providing more established and potential therapeutic strategies in tumors to facilitate epigenetic studies of tumors.
Collapse
Affiliation(s)
- Tiansheng Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chao Mao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ying Shi
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
33
|
Chang CC, Dinh TK, Lee YA, Wang FN, Sung YC, Yu PL, Chiu SC, Shih YC, Wu CY, Huang YD, Wang J, Lu TT, Wan D, Chen Y. Nanoparticle Delivery of MnO 2 and Antiangiogenic Therapy to Overcome Hypoxia-Driven Tumor Escape and Suppress Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44407-44419. [PMID: 32865389 DOI: 10.1021/acsami.0c08473] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Antiangiogenic therapy is widely administered in many cancers, and the antiangiogenic drug sorafenib offers moderate benefits in advanced hepatocellular carcinoma (HCC). However, antiangiogenic therapy can also lead to hypoxia-driven angiogenesis and immunosuppression in the tumor microenvironment (TME) and metastasis. Here, we report the synthesis and evaluation of NanoMnSor, a tumor-targeted, nanoparticle drug carrier that efficiently codelivers oxygen-generating MnO2 and sorafenib into HCC. We found that MnO2 not only alleviates hypoxia by catalyzing the decomposition of H2O2 to oxygen but also enhances pH/redox-responsive T1-weighted magnetic resonance imaging and drug-release properties upon decomposition into Mn2+ ions in the TME. Moreover, macrophages exposed to MnO2 displayed increased mRNA associated with the immunostimulatory M1 phenotype. We further show that NanoMnSor treatment leads to sorafenib-induced decrease in tumor vascularization and significantly suppresses primary tumor growth and distal metastasis, resulting in improved overall survival in a mouse orthotopic HCC model. Furthermore, NanoMnSor reprograms the immunosuppressive TME by reducing the hypoxia-induced tumor infiltration of tumor-associated macrophages, promoting macrophage polarization toward the immunostimulatory M1 phenotype, and increasing the number of CD8+ cytotoxic T cells in tumors, thereby augmenting the efficacy of anti-PD-1 antibody and whole-cell cancer vaccine immunotherapies. Our study demonstrates the potential of oxygen-generating nanoparticles to deliver antiangiogenic agents, efficiently modulate the hypoxic TME, and overcome hypoxia-driven drug resistance, thereby providing therapeutic benefit in cancer.
Collapse
Affiliation(s)
- Chih-Chun Chang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Trinh Kieu Dinh
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-An Lee
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Fu-Nien Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Pei-Lun Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Shao-Chieh Chiu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yu-Chuan Shih
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Cheng-Yun Wu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-Da Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Dehui Wan
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
34
|
Montemagno C, Pagès G. Resistance to Anti-angiogenic Therapies: A Mechanism Depending on the Time of Exposure to the Drugs. Front Cell Dev Biol 2020; 8:584. [PMID: 32775327 PMCID: PMC7381352 DOI: 10.3389/fcell.2020.00584] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from preexisting one, represents a critical process for oxygen and nutrient supply to proliferating cells, therefore promoting tumor growth and metastasis. The Vascular Endothelial Growth Factor (VEGF) pathway is one of the key mediators of angiogenesis in cancer. Therefore, several therapies including monoclonal antibodies or tyrosine kinase inhibitors target this axis. Although preclinical studies demonstrated strong antitumor activity, clinical studies were disappointing. Antiangiogenic drugs, used to treat metastatic patients suffering of different types of cancers, prolonged survival to different extents but are not curative. In this review, we focused on different mechanisms involved in resistance to antiangiogenic therapies from early stage resistance involving mainly tumor cells to late stages related to the adaptation of the microenvironment.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| |
Collapse
|
35
|
Oxidized ATM promotes breast cancer stem cell enrichment through energy metabolism reprogram-mediated acetyl-CoA accumulation. Cell Death Dis 2020; 11:508. [PMID: 32641713 PMCID: PMC7343870 DOI: 10.1038/s41419-020-2714-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
Cancer stem cell (CSC) is a challenge in the therapy of triple-negative breast cancer (TNBC). Intratumoral hypoxia is a common feature of solid tumor. Hypoxia may contribute to the maintenance of CSC, resulting in a poor efficacy of traditional treatment and recurrence of TNBC cases. However, the underlying molecular mechanism involved in hypoxia-induced CSC stemness maintenance remains unclear. Here, we report that hypoxia stimulated DNA double-strand breaks independent of ATM kinase activation (called oxidized ATM in this paper) play a crucial role in TNBC mammosphere formation and stemness maintenance by governing a specific energy metabolism reprogramming (EMR). Oxidized ATM up-regulates GLUT1, PKM2, and PDHa expressions to enhance the uptake of glucose and production of pyruvate rather than lactate products, which facilitates glycolytic flux to mitochondrial pyruvate and citrate, thus resulting in accumulation of cytoplasmic acetyl-CoA instead of the tricarboxylic acid (TCA) cycle by regulating ATP-citrate lyase (ACLY) activity. Our findings unravel a novel model of TNBC-CSC glucose metabolism and its functional role in maintenance of hypoxic TNBC-CSC stemness. This work may help us to develop new therapeutic strategies for TNBC treatment.
Collapse
|
36
|
Zhang J, Xue W, Xu K, Yi L, Guo Y, Xie T, Tong H, Zhou B, Wang S, Li Q, Liu H, Chen X, Fang J, Zhang W. Dual inhibition of PFKFB3 and VEGF normalizes tumor vasculature, reduces lactate production, and improves chemotherapy in glioblastoma: insights from protein expression profiling and MRI. Theranostics 2020; 10:7245-7259. [PMID: 32641990 PMCID: PMC7330843 DOI: 10.7150/thno.44427] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Tumor vascular normalization (TVN) is emerging to enhance the efficacy of anticancer treatment in many cancers including glioblastoma (GBM). However, a common and severe challenge being currently faced is the transient TVN effect, hampering the sustained administration of anticancer therapy during TVN window. Additionally, the lack of non-contrast agent-based imaging biomarkers to monitor TVN process postpones the clinical translation of TVN strategy. In this study, we investigated whether dual inhibition of VEGF and the glycolytic activator PFKFB3 could reinforce the TVN effect in GBM. Dynamic contrast-enhanced-magnetic resonance imaging (DCE-MRI) and intravoxel incoherent motion (IVIM)-MRI were performed to monitor TVN process and to identify whether IVIM-MRI is a candidate or complementary imaging biomarker for monitoring TVN window without exogenous contrast agent administration. Methods: Patient-derived orthotopic GBM xenografts in mice were established and treated with bevacizumab (BEV), 3PO (PFKFB3 inhibitor), BEV+3PO dual therapy, or saline. The vascular morphology, tumor hypoxia, and lactate level were evaluated before and at different time points after treatments. Doxorubicin was used to evaluate chemotherapeutic efficacy and drug delivery. Microarray of angiogenesis cytokines and western blotting were conducted to characterize post-treatment molecular profiling. TVN process was monitored by DCE- and IVIM-MRI. Correlation analysis of pathological indicators and MRI parameters was further analyzed. Results: Dual therapy extended survival and delayed tumor growth over each therapy alone, concomitant with a decrease of cell proliferation and an increase of cell apoptosis. The dual therapy reinforces TVN effect, thereby alleviating tumor hypoxia, reducing lactate production, and improving the efficacy and delivery of doxorubicin. Mechanistically, several angiogenic cytokines and pathways were downregulated after dual therapy. Notably, dual therapy inhibited Tie1 expression, the key regulator of TVN, in both endothelial cells and tumor cells. DCE- and IVIM-MRI data showed that dual therapy induced a more homogenous and prominent TVN effect characterized by improved vascular function in tumor core and tumor rim. Correlation analysis revealed that IVIM-MRI parameter D* had better correlations with TVN pathological indicators compared with the DCE-MRI parameter Ktrans. Conclusions: Our results propose a rationale to overcome the current limitation of BEV monotherapy by integrating the synergistic effects of VEGF and PFKFB3 blockade to enhance chemotherapy efficacy through a sustained TVN effect. Moreover, we unveil IVIM-MRI parameter D* has much potential as a complementary imaging biomarker to monitor TVN window more precisely without exogenous contrast agent injection.
Collapse
Affiliation(s)
- Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wei Xue
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Kai Xu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liang Yi
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Tian Xie
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Haipeng Tong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Bo Zhou
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qing Li
- Department of Oncology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, 400042, China
| |
Collapse
|
37
|
Pellicani R, Poletto E, Andreuzzi E, Paulitti A, Doliana R, Bizzotto D, Braghetta P, Colladel R, Tarticchio G, Sabatelli P, Bucciotti F, Bressan G, Iozzo RV, Colombatti A, Bonaldo P, Mongiat M. Multimerin-2 maintains vascular stability and permeability. Matrix Biol 2020; 87:11-25. [DOI: 10.1016/j.matbio.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022]
|
38
|
Li X, Li Y, Lu W, Chen M, Ye W, Zhang D. The Tumor Vessel Targeting Strategy: A Double-Edged Sword in Tumor Metastasis. Cells 2019; 8:E1602. [PMID: 31835465 PMCID: PMC6952935 DOI: 10.3390/cells8121602] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
Tumor vessels provide essential paths for tumor cells to escape from the primary tumor and form metastatic foci in distant organs. The vessel targeting strategy has been widely used as an important clinical cancer chemotherapeutic strategy for patients with metastatic tumors. Our review introduces the contribution of angiogenesis to tumor metastasis and summarizes the application of Food and Drug Administration (FDA)-approved vessel targeting drugs for metastatic tumors. We recommend the application and mechanisms of vascular targeting drugs for inhibiting tumor metastasis and discuss the risk and corresponding countermeasures after vessel targeting treatment.
Collapse
Affiliation(s)
- Xiaobo Li
- College of Pharmacy, Jinan University, No. 601, Huangpu Road West, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Yong Li
- College of Pharmacy, Jinan University, No. 601, Huangpu Road West, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Weijin Lu
- College of Pharmacy, Jinan University, No. 601, Huangpu Road West, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, No. 601, Huangpu Road West, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Wencai Ye
- College of Pharmacy, Jinan University, No. 601, Huangpu Road West, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, No. 601, Huangpu Road West, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| |
Collapse
|
39
|
Kinetic modelling of quantitative proteome data predicts metabolic reprogramming of liver cancer. Br J Cancer 2019; 122:233-244. [PMID: 31819186 PMCID: PMC7052204 DOI: 10.1038/s41416-019-0659-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
Background Metabolic alterations can serve as targets for diagnosis and cancer therapy. Due to the highly complex regulation of cellular metabolism, definite identification of metabolic pathway alterations remains challenging and requires sophisticated experimentation. Methods We applied a comprehensive kinetic model of the central carbon metabolism (CCM) to characterise metabolic reprogramming in murine liver cancer. Results We show that relative differences of protein abundances of metabolic enzymes obtained by mass spectrometry can be used to assess their maximal velocity values. Model simulations predicted tumour-specific alterations of various components of the CCM, a selected number of which were subsequently verified by in vitro and in vivo experiments. Furthermore, we demonstrate the ability of the kinetic model to identify metabolic pathways whose inhibition results in selective tumour cell killing. Conclusions Our systems biology approach establishes that combining cellular experimentation with computer simulations of physiology-based metabolic models enables a comprehensive understanding of deregulated energetics in cancer. We propose that modelling proteomics data from human HCC with our approach will enable an individualised metabolic profiling of tumours and predictions of the efficacy of drug therapies targeting specific metabolic pathways.
Collapse
|
40
|
Rewiring of Lipid Metabolism and Storage in Ovarian Cancer Cells after Anti-VEGF Therapy. Cells 2019; 8:cells8121601. [PMID: 31835444 PMCID: PMC6953010 DOI: 10.3390/cells8121601] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022] Open
Abstract
Anti-angiogenic therapy triggers metabolic alterations in experimental and human tumors, the best characterized being exacerbated glycolysis and lactate production. By using both Liquid Chromatography-Mass Spectrometry (LC-MS) and Nuclear Magnetic Resonance (NMR) analysis, we found that treatment of ovarian cancer xenografts with the anti-Vascular Endothelial Growth Factor (VEGF) neutralizing antibody bevacizumab caused marked alterations of the tumor lipidomic profile, including increased levels of triacylglycerols and reduced saturation of lipid chains. Moreover, transcriptome analysis uncovered up-regulation of pathways involved in lipid metabolism. These alterations were accompanied by increased accumulation of lipid droplets in tumors. This phenomenon was reproduced under hypoxic conditions in vitro, where it mainly depended from uptake of exogenous lipids and was counteracted by treatment with the Liver X Receptor (LXR)-agonist GW3965, which inhibited cancer cell viability selectively under reduced serum conditions. This multi-level analysis indicates alterations of lipid metabolism following anti-VEGF therapy in ovarian cancer xenografts and suggests that LXR-agonists might empower anti-tumor effects of bevacizumab.
Collapse
|
41
|
Brossa A, Buono L, Fallo S, Fiorio Pla A, Munaron L, Bussolati B. Alternative Strategies to Inhibit Tumor Vascularization. Int J Mol Sci 2019; 20:E6180. [PMID: 31817884 PMCID: PMC6940973 DOI: 10.3390/ijms20246180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells present in tumors show different origin, phenotype, and genotype with respect to the normal counterpart. Various mechanisms of intra-tumor vasculogenesis sustain the complexity of tumor vasculature, which can be further modified by signals deriving from the tumor microenvironment. As a result, resistance to anti-VEGF therapy and activation of compensatory pathways remain a challenge in the treatment of cancer patients, revealing the need to explore alternative strategies to the classical anti-angiogenic drugs. In this review, we will describe some alternative strategies to inhibit tumor vascularization, including targeting of antigens and signaling pathways overexpressed by tumor endothelial cells, the development of endothelial vaccinations, and the use of extracellular vesicles. In addition, anti-angiogenic drugs with normalizing effects on tumor vessels will be discussed. Finally, we will present the concept of endothelial demesenchymalization as an alternative approach to restore normal endothelial cell phenotype.
Collapse
Affiliation(s)
- Alessia Brossa
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Lola Buono
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Sofia Fallo
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Alessandra Fiorio Pla
- Department of Life Science and Systems Biology, University of Torino, 10126 Torino, Italy; (A.F.P.); (L.M.)
| | - Luca Munaron
- Department of Life Science and Systems Biology, University of Torino, 10126 Torino, Italy; (A.F.P.); (L.M.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| |
Collapse
|
42
|
Long Y, Tao H, Karachi A, Grippin AJ, Jin L, Chang YE, Zhang W, Dyson KA, Hou AY, Na M, Deleyrolle LP, Sayour EJ, Rahman M, Mitchell DA, Lin Z, Huang J. Dysregulation of Glutamate Transport Enhances Treg Function That Promotes VEGF Blockade Resistance in Glioblastoma. Cancer Res 2019; 80:499-509. [PMID: 31723000 DOI: 10.1158/0008-5472.can-19-1577] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/15/2019] [Accepted: 11/08/2019] [Indexed: 11/16/2022]
Abstract
Anti-VEGF therapy prolongs recurrence-free survival in patients with glioblastoma but does not improve overall survival. To address this discrepancy, we investigated immunologic resistance mechanisms to anti-VEGF therapy in glioma models. A screening of immune-associated alterations in tumors after anti-VEGF treatment revealed a dose-dependent upregulation of regulatory T-cell (Treg) signature genes. Enhanced numbers of Tregs were observed in spleens of tumor-bearing mice and later in tumors after anti-VEGF treatment. Elimination of Tregs with CD25 blockade before anti-VEGF treatment restored IFNγ production from CD8+ T cells and improved antitumor response from anti-VEGF therapy. The treated tumors overexpressed the glutamate/cystine antiporter SLC7A11/xCT that led to elevated extracellular glutamate in these tumors. Glutamate promoted Treg proliferation, activation, suppressive function, and metabotropic glutamate receptor 1 (mGlutR1) expression. We propose that VEGF blockade coupled with glioma-derived glutamate induces systemic and intratumoral immunosuppression by promoting Treg overrepresentation and function, which can be pre-emptively overcome through Treg depletion for enhanced antitumor effects. SIGNIFICANCE: Resistance to VEGF therapy in glioblastoma is driven by upregulation of Tregs, combined blockade of VEGF, and Tregs may provide an additive antitumor effect for treating glioblastoma.
Collapse
Affiliation(s)
- Yu Long
- The Fourth Section of Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Haipeng Tao
- The Fourth Section of Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Aida Karachi
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Adam J Grippin
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Linchun Jin
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Yifan Emily Chang
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Wang Zhang
- The Fourth Section of Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Kyle A Dyson
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Alicia Y Hou
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Meng Na
- The Fourth Section of Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Loic P Deleyrolle
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, Florida
| | - Elias J Sayour
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, Florida
| | - Maryam Rahman
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, Florida
| | - Duane A Mitchell
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, Florida
| | - Zhiguo Lin
- The Fourth Section of Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Jianping Huang
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida.
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, Florida
| |
Collapse
|
43
|
Yun JA, Kim J, Baek YY, Park W, Park M, Kim S, Kim T, Choi S, Jeoung D, Lee H, Won MH, Kim JY, Ha KS, Kwon YG, Kim YM. N-Terminal Modification of the Tetrapeptide Arg-Leu-Tyr-Glu, a Vascular Endothelial Growth Factor Receptor-2 (VEGFR-2) Antagonist, Improves Antitumor Activity by Increasing its Stability against Serum Peptidases. Mol Pharmacol 2019; 96:692-701. [DOI: 10.1124/mol.119.117234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
|
44
|
Affiliation(s)
- Tero Ah Järvinen
- Faculty of Medicine and Health Technologies, Tampere University and Department of Orthopedics & Traumatology, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
45
|
Xie W, Zhang Y, Zhang S, Wang F, Zhang K, Huang Y, Zhou Z, Huang G, Wang J. Oxymatrine enhanced anti-tumor effects of Bevacizumab against triple-negative breast cancer via abating Wnt/β-Catenin signaling pathway. Am J Cancer Res 2019; 9:1796-1814. [PMID: 31497360 PMCID: PMC6726986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023] Open
Abstract
Bevacizumab, a monoclonal antibody targeting vascular endothelial growth factor A (VEGF-A), was used in combination with traditional chemotherapy as the first line treatment for metastatic colorectal cancer (mCRC), non-small cell lung cancer (NSCLC) and advanced ovarian cancer. However, it shows limited efficacy for human triple-negative breast cancer (TNBC). Bevacizumab shows potent anti-angiogenesis activity, meanwhile, it also increases invasive and metastatic properties of TNBC cells by activiting Wnt/β-Catenin pathway. To overcome this problem, and fully utilize its potency against cancer, further synergistic strategy is recommended to be developed, especially the concurrent use with those Wnt-targeting agents. Here, by screening a small library of traditional Chinese medicine, we identified a Chinese herb derived Oxymatrine, which could target Wnt/β-Catenin signaling and compromise the oncogenic effects of Bevacizumab. Bevacizumab was validated to induce epithelial-mesenchymal cell transformation (EMT) and cancer stem-like properties of TNBC cells in hypoxia/nutritional stress environment. On the contrary, Oxymatrine reversed the EMT phenotype and depleted the subpopulation of TNBC stem cells induced by Bevacizumab. Oxymatrine enhanced the anti-tumor effects of Bevacizumab in vivo, and holded the potential of reducing the risk of relapse and metastasis by impairing the self-renewal ability of TNBC stem cells. The underlying mechanism was elucidated: Bevacizumab stimulated Wnt/β-Catenin signaling pathway, and Oxymatrine could compromise this effect. On this foundation, factoring into the satisfactory anti-angiogenic activity and low toxicity, Oxymatrine is a good candidate for the synergistic therapy together with Bevacizumab for the treatment of TNBC.
Collapse
Affiliation(s)
- Wei Xie
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Yan Zhang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai University of Traditional Chinese Medicine Graduate SchoolShanghai 201203, P. R. China
| | - Shiwei Zhang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Fengxian Wang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai University of Traditional Chinese Medicine Graduate SchoolShanghai 201203, P. R. China
| | - Kunchi Zhang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Yanjuan Huang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Zhaoli Zhou
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Gang Huang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| | - Jin Wang
- School of Pharmacy, Shanghai University of Medicine and Health SciencesShanghai 201318, P. R. China
| |
Collapse
|
46
|
Harnessing Induced Essentiality: Targeting Carbonic Anhydrase IX and Angiogenesis Reduces Lung Metastasis of Triple Negative Breast Cancer Xenografts. Cancers (Basel) 2019; 11:cancers11071002. [PMID: 31319613 PMCID: PMC6678951 DOI: 10.3390/cancers11071002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/28/2019] [Accepted: 07/15/2019] [Indexed: 01/05/2023] Open
Abstract
Triple Negative Breast Cancer (TNBC) is aggressive, metastatic and drug-resistant, limiting the spectrum of effective therapeutic options for breast cancer patients. To date, anti-angiogenic agents have had limited success in the treatment of systemic breast cancer, possibly due to the exacerbation of tumor hypoxia and increased metastasis. Hypoxia drives increased expression of downstream effectors, including Carbonic Anhydrase IX (CAIX), a critical functional component of the pro-survival machinery required by hypoxic tumor cells. Here, we used the highly metastatic, CAIX-positive MDA-MB-231 LM2-4 orthotopic model of TNBC to investigate whether combinatorial targeting of CAIX and angiogenesis impacts tumor growth and metastasis in vivo to improve efficacy. The administration of a small molecule inhibitor of CAIX, SLC-0111, significantly reduced overall metastatic burden, whereas exposure to sunitinib increased hypoxia and CAIX expression in primary tumors, and failed to inhibit metastasis. The administration of SLC-0111 significantly decreased primary tumor vascular density and permeability, and reduced metastasis to the lung and liver. Furthermore, combining sunitinib and SLC-0111 significantly reduced both primary tumor growth and sunitinib-induced metastasis to the lung. Our findings suggest that targeting angiogenesis and hypoxia effectors in combination holds promise as a novel rational strategy for the effective treatment of patients with TNBC.
Collapse
|
47
|
Gupta R, Bhatt LK, Johnston TP, Prabhavalkar KS. Colon cancer stem cells: Potential target for the treatment of colorectal cancer. Cancer Biol Ther 2019; 20:1068-1082. [PMID: 31050577 DOI: 10.1080/15384047.2019.1599660] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite incessant research, colon cancer still is one of the most common causes of fatalities in both men and women worldwide. Also, nearly 50% of patients with colorectal cancer show tumor recurrence. Recent investigations have highlighted the involvement of colon cancer stem cells (CCSCs) in cancer relapse and chemoresistance. CCSCs deliver a significant protumorigenic niche through persistent overexpression of self-renewal capabilities. Moreover, CSCs cross network with stromal cells, immune infiltrates, and cyotokine-chemokine, which potentiate their aggressive proliferative potential. Targeting CCSCs through small molecule inhibitors, miRNAs, and monoclonal antibodies (mAbs) in in vivo studies has generated compelling evidence for the effectiveness of these various treatments. This review effectively compiles the role of CCSC surface markers and dysregulated and/or upregulated pathways in the pathogenesis of colorectal cancer that can be used to target CCSCs for effective colorectal cancer treatment.
Collapse
Affiliation(s)
- Riya Gupta
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Lokesh Kumar Bhatt
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Thomas P Johnston
- b Division of Pharmacology and Pharmaceutical Sciences , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Kedar S Prabhavalkar
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| |
Collapse
|
48
|
Hoshi T, Watanabe Miyano S, Watanabe H, Sonobe RMK, Seki Y, Ohta E, Nomoto K, Matsui J, Funahashi Y. Lenvatinib induces death of human hepatocellular carcinoma cells harboring an activated FGF signaling pathway through inhibition of FGFR-MAPK cascades. Biochem Biophys Res Commun 2019; 513:1-7. [PMID: 30944079 DOI: 10.1016/j.bbrc.2019.02.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/03/2019] [Indexed: 01/16/2023]
Abstract
Lenvatinib inhibits VEGF- and FGF-driven angiogenesis, and proliferation of tumor cells with activated FGF signaling pathways in preclinical models, and we previously demonstrated antitumor activity in human HCC xenograft tumor models. Here, we examined the inhibitory activity of lenvatinib against FGF-driven survival of human HCC cell lines. First, we conducted a histological analysis of FGF19-overexpressing Hep3B2.1-7 xenograft tumors collected from mice treated with lenvatinib. Second, we examined the effects of pharmacological inhibition on survival of cultured HCC cells with an activated FGF signaling pathway under nutrient-starved culture condition to mimic tumor microenvironments induced by angiogenesis inhibition. In the first analysis, area of histological focal necrosis was greater in Hep3B2.1-7 xenograft tumors with the lenvatinib treatment than that after the treatment with sorafenib, which does not inhibit FGFRs. Lenvatinib and E7090 (a selective FGFR1-3 inhibitor), but not sorafenib, induced death of Hep3B2.1-7, and another FGF19 overexpressing HuH-7 cells. Lenvatinib and E7090 decreased phosphorylation of downstream molecules of the FGF signaling pathway (such as FRS2, Erk, and p38 MAPK), and induced PARP cleavage, even under limited nutrients. PD0325901, MEK inhibitor, caused the same changes in HCC cells as those described above for lenvatinib and E7090. These results reveal that the FGF signaling pathway through MAPK cascades plays an important role in survival of HCC cell lines with an activated FGF signaling pathway under limited nutrients, and FGFR-MAPK cascades likely contribute to survival of HCC cells with an activated FGF signaling pathway under tumor microenvironments with limited nutrients, where tumor angiogenesis is inhibited.
Collapse
Affiliation(s)
- Taisuke Hoshi
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Hideki Watanabe
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Yuki Seki
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki, Japan
| | - Etsuko Ohta
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki, Japan
| | - Kenichi Nomoto
- Oncology Business Group, Eisai Inc., Woodcliff Lake, NJ, USA
| | - Junji Matsui
- Oncology Business Group, Eisai Inc., Woodcliff Lake, NJ, USA
| | | |
Collapse
|
49
|
Daubon T, Léon C, Clarke K, Andrique L, Salabert L, Darbo E, Pineau R, Guérit S, Maitre M, Dedieu S, Jeanne A, Bailly S, Feige JJ, Miletic H, Rossi M, Bello L, Falciani F, Bjerkvig R, Bikfalvi A. Deciphering the complex role of thrombospondin-1 in glioblastoma development. Nat Commun 2019; 10:1146. [PMID: 30850588 PMCID: PMC6408502 DOI: 10.1038/s41467-019-08480-y] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 01/09/2019] [Indexed: 12/17/2022] Open
Abstract
We undertook a systematic study focused on the matricellular protein Thrombospondin-1 (THBS1) to uncover molecular mechanisms underlying the role of THBS1 in glioblastoma (GBM) development. THBS1 was found to be increased with glioma grades. Mechanistically, we show that the TGFβ canonical pathway transcriptionally regulates THBS1, through SMAD3 binding to the THBS1 gene promoter. THBS1 silencing inhibits tumour cell invasion and growth, alone and in combination with anti-angiogenic therapy. Specific inhibition of the THBS1/CD47 interaction using an antagonist peptide decreases cell invasion. This is confirmed by CD47 knock-down experiments. RNA sequencing of patient-derived xenograft tissue from laser capture micro-dissected peripheral and central tumour areas demonstrates that THBS1 is one of the gene with the highest connectivity at the tumour borders. All in all, these data show that TGFβ1 induces THBS1 expression via Smad3 which contributes to the invasive behaviour during GBM expansion. Furthermore, tumour cell-bound CD47 is implicated in this process. Thrombospondin-1 (THSB1) is a component of the ECM with a role in regulating cancer development and tumour vasculature. Here, the authors show that TGF-beta-induced THBS1 expression contributes to the invasive behaviour of GBM cells and promotes resistance to antiangiogenic therapy partially through interaction with CD47.
Collapse
Affiliation(s)
- Thomas Daubon
- INSERM U1029, Institut Nationale de la Santé et de la Recherche Médicale, 33615, Pessac, France. .,University Bordeaux, 33615, Pessac, France. .,KG Jebsen Brain Tumor Research Center, University of Bergen, 5020, Bergen, Norway. .,Norlux Beuro-Oncology, Department of Biomedicine, University of Bergen, 5020, Bergen, Norway.
| | - Céline Léon
- INSERM U1029, Institut Nationale de la Santé et de la Recherche Médicale, 33615, Pessac, France.,University Bordeaux, 33615, Pessac, France
| | - Kim Clarke
- Computational Biology Facility, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Laetitia Andrique
- INSERM U1029, Institut Nationale de la Santé et de la Recherche Médicale, 33615, Pessac, France.,University Bordeaux, 33615, Pessac, France
| | - Laura Salabert
- INSERM U1029, Institut Nationale de la Santé et de la Recherche Médicale, 33615, Pessac, France.,University Bordeaux, 33615, Pessac, France
| | - Elodie Darbo
- UMR1218 ACTION, Bioinformatic Center CBiB, University of Bordeaux, 33076, Bordeaux, France
| | - Raphael Pineau
- Animal Facility, University Bordeaux, 33615, Pessac, France
| | - Sylvaine Guérit
- INSERM U1029, Institut Nationale de la Santé et de la Recherche Médicale, 33615, Pessac, France.,University Bordeaux, 33615, Pessac, France
| | - Marlène Maitre
- INSERM U1215, Neurocenter Magendie, Pathophysiology of Addiction Group, 33076, Bordeaux, France
| | | | - Albin Jeanne
- CNRS UMR 7369, MEDyC, 51687, Reims, France.,SATT Nord, 59800, Lille, France
| | | | | | - Hrvoje Miletic
- KG Jebsen Brain Tumor Research Center, University of Bergen, 5020, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, 5020, Bergen, Norway
| | - Marco Rossi
- Neurosurgical Oncology Unit, Department of Oncology and Hemato-Oncology, Humanitas Research Hospital, Universita Degli Studi di Milano, 20089, Rozzano, Milan, Italy
| | - Lorenzo Bello
- Neurosurgical Oncology Unit, Department of Oncology and Hemato-Oncology, Humanitas Research Hospital, Universita Degli Studi di Milano, 20089, Rozzano, Milan, Italy
| | - Francesco Falciani
- Computational Biology Facility, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Rolf Bjerkvig
- KG Jebsen Brain Tumor Research Center, University of Bergen, 5020, Bergen, Norway.,Norlux Beuro-Oncology, Department of Biomedicine, University of Bergen, 5020, Bergen, Norway.,Oncology Department, Luxembourg Institute of Health, 84, Val Fleuri, 1526, Luxembourg
| | - Andréas Bikfalvi
- INSERM U1029, Institut Nationale de la Santé et de la Recherche Médicale, 33615, Pessac, France. .,University Bordeaux, 33615, Pessac, France.
| |
Collapse
|
50
|
Bousseau S, Marchand M, Soleti R, Vergori L, Hilairet G, Recoquillon S, Le Mao M, Gueguen N, Khiati S, Clarion L, Bakalara N, Martinez MC, Germain S, Lenaers G, Andriantsitohaina R. Phostine 3.1a as a pharmacological compound with antiangiogenic properties against diseases with excess vascularization. FASEB J 2019; 33:5864-5875. [PMID: 30817178 DOI: 10.1096/fj.201801450rrr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Angiogenesis is a complex process leading to the growth of new blood vessels from existing vasculature, triggered by local proangiogenic factors such as VEGF. An excess of angiogenesis is a recurrent feature of various pathologic conditions such as tumor growth. Phostines are a family of synthetic glycomimetic compounds that exhibit anticancer properties, and the lead compound 3-hydroxy-4,5-bis-benzyloxy-6-benzyloxymethyl-2-phenyl2-oxo-2λ5-[1,2]oxaphosphinane (PST 3.1a) shows antiglioblastoma properties both in vitro and in vivo. In the present study, we assessed the effect of PST 3.1a on angiogenesis and endothelial metabolism. In vitro, PST 3.1a (10 µM) inhibited all steps that regulate angiogenesis, including migration, proliferation, adhesion, and tube formation. In vivo, PST 3.1a reduced intersegmental vessel formation and vascularization of the subintestinal plexus in zebrafish embryos and also altered pathologic angiogenesis and glioblastoma progression in vivo. Mechanistically, PST 3.1a altered interaction of VEGF receptor 2 and glycosylation-regulating protein galectin-1, a key component regulating angiogenesis associated with tumor resistance. Thus, these data show that use of PST 3.1a is an innovative approach to target angiogenesis.-Bousseau, S., Marchand, M., Soleti, R., Vergori, L., Hilairet, G., Recoquillon, S., Le Mao, M., Gueguen, N., Khiati, S., Clarion, L., Bakalara, N., Martinez, M. C., Germain, S., Lenaers, G., Andriantsitohaina, R. Phostine 3.1a as a pharmacological compound with antiangiogenic properties against diseases with excess vascularization.
Collapse
Affiliation(s)
- Simon Bousseau
- INSERM Unité Mixte de Recherche (UMR) 1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Angers, France.,MitoLab, Centre National de la Recherche (CNRS) Unité Mixte de Recherche (UMR) 6015, INSERM Unité 1083, Institut MitoVasc, Université d'Angers, Angers, France
| | - Marion Marchand
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Centre National de la Recherche (CNRS), INSERM, Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Raffaella Soleti
- INSERM Unité Mixte de Recherche (UMR) 1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Angers, France
| | - Luisa Vergori
- INSERM Unité Mixte de Recherche (UMR) 1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Angers, France
| | - Grégory Hilairet
- INSERM Unité Mixte de Recherche (UMR) 1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Angers, France
| | - Sylvain Recoquillon
- INSERM Unité Mixte de Recherche (UMR) 1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Angers, France
| | - Morgane Le Mao
- MitoLab, Centre National de la Recherche (CNRS) Unité Mixte de Recherche (UMR) 6015, INSERM Unité 1083, Institut MitoVasc, Université d'Angers, Angers, France
| | - Naig Gueguen
- MitoLab, Centre National de la Recherche (CNRS) Unité Mixte de Recherche (UMR) 6015, INSERM Unité 1083, Institut MitoVasc, Université d'Angers, Angers, France
| | - Salim Khiati
- MitoLab, Centre National de la Recherche (CNRS) Unité Mixte de Recherche (UMR) 6015, INSERM Unité 1083, Institut MitoVasc, Université d'Angers, Angers, France
| | - Ludovic Clarion
- Phost'in SAS (société par actions simplifiée), Montpellier, France
| | - Norbert Bakalara
- INSERM Unité 1051, Institut des Neurosciences de Montpellier, Montpellier, France
| | - M Carmen Martinez
- INSERM Unité Mixte de Recherche (UMR) 1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Angers, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Centre National de la Recherche (CNRS), INSERM, Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Guy Lenaers
- MitoLab, Centre National de la Recherche (CNRS) Unité Mixte de Recherche (UMR) 6015, INSERM Unité 1083, Institut MitoVasc, Université d'Angers, Angers, France
| | - Ramaroson Andriantsitohaina
- INSERM Unité Mixte de Recherche (UMR) 1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Angers, France
| |
Collapse
|