1
|
Suzuki S, Yashiro M, Izumi N, Tsukioka T, Inoue H, Hara K, Ito R, Tanimura T, Nishiyama N. Impact of CA9 expression in the diagnosis of lymph-node metastases in non-small cell lung cancer based on [18F]FDG PET/CT. PLoS One 2024; 19:e0312846. [PMID: 39471162 PMCID: PMC11521239 DOI: 10.1371/journal.pone.0312846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND Lung cancer is the leading cause of the global cancer incidence and mortality. It is important to obtain an accurate diagnosis of lymph-node metastasis before surgery to select the therapeutic strategy for non-small cell lung cancer (NSCLC) patients. Carbonic anhydrase 9 (CA9) is considered a marker of hypoxia and it has reported that CA9 is associated with tumor invasion and metastasis. In this study, the correlation between the CA9 expression for lymph-node metastases in NSCLC and [18F]FDG PET/CT results was investigated in order to clarify the efficacy of [18F]FDG PET/CT for detecting lymph-node metastases of NSCLC patients. METHODS Among the 564 patients who underwent surgical treatment for NSCLC between 2010 and 2016 at our hospital, a total of 338 patients who underwent preoperative [18F]FDG PET/CT were included in this study. CA9 expression was evaluated by immunochemistry. A lymph node with maximum standardized uptake value (SUVmax) ≥2.5 on [18F]FDG PET/CT was preoperatively defined as a metastatic lymph node. RESULT CA9 positivity was detected in 122 patients; the other 216 patients were CA9-negative. The CA9-positive NSCLC cases significantly associated with pleural invasion (p = 0.0063), pT-factor (p = 0.0080), pN-factor (p = 0.036) and pStage (p = 0.043). CA9-positive patients presented significantly poorer survival rate for OS than that of the CA9-negative patients (p = 0.0024). In the multivariable analysis, histological SCC and CA9 positivity were independent poor-prognosis factors for OS. For the total patient population, the sensitivity and specificity of [18F]FDG PET/CT for lymph-node metastases were 54% and 89%, respectively. In contrast, the sensitivity and specificity were particularly low in the CA9-positive SCC cases (36% and 69%, respectively). CONCLUSION [18F]FDG PET/CT might not be useful for diagnosing lymph-node metastases of CA9-positive SCC cases of NSCLC.
Collapse
Affiliation(s)
- Satoshi Suzuki
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Nobuhiro Izumi
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Takuma Tsukioka
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Inoue
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kantaro Hara
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Ryuichi Ito
- Department of Thoracic Surgery, Kansai Rosai Hospital, Hyogo, Japan
| | - Takuya Tanimura
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Noritoshi Nishiyama
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
2
|
Riaz F, Zhang J, Pan F. Forces at play: exploring factors affecting the cancer metastasis. Front Immunol 2024; 15:1274474. [PMID: 38361941 PMCID: PMC10867181 DOI: 10.3389/fimmu.2024.1274474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Metastatic disease, a leading and lethal indication of deaths associated with tumors, results from the dissemination of metastatic tumor cells from the site of primary origin to a distant organ. Dispersion of metastatic cells during the development of tumors at distant organs leads to failure to comply with conventional treatments, ultimately instigating abrupt tissue homeostasis and organ failure. Increasing evidence indicates that the tumor microenvironment (TME) is a crucial factor in cancer progression and the process of metastatic tumor development at secondary sites. TME comprises several factors contributing to the initiation and progression of the metastatic cascade. Among these, various cell types in TME, such as mesenchymal stem cells (MSCs), lymphatic endothelial cells (LECs), cancer-associated fibroblasts (CAFs), myeloid-derived suppressor cells (MDSCs), T cells, and tumor-associated macrophages (TAMs), are significant players participating in cancer metastasis. Besides, various other factors, such as extracellular matrix (ECM), gut microbiota, circadian rhythm, and hypoxia, also shape the TME and impact the metastatic cascade. A thorough understanding of the functions of TME components in tumor progression and metastasis is necessary to discover new therapeutic strategies targeting the metastatic tumor cells and TME. Therefore, we reviewed these pivotal TME components and highlighted the background knowledge on how these cell types and disrupted components of TME influence the metastatic cascade and establish the premetastatic niche. This review will help researchers identify these altered components' molecular patterns and design an optimized, targeted therapy to treat solid tumors and restrict metastatic cascade.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Jing Zhang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
3
|
Pang X, Li TJ, Shi RJ, Wan ZX, Tang YY, Tang YL, Liang XH. IRF2BP2 drives lymphatic metastasis in OSCC cells by elevating mitochondrial fission-dependent fatty acid oxidation. Mol Carcinog 2024; 63:45-60. [PMID: 37737489 DOI: 10.1002/mc.23635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Abstract
Lymph node metastasis (LNM) is a major determinant for the poor outcome of oral squamous cell carcinoma (OSCC). Interferon regulatory factor 2 binding protein 2 (IRF2BP2) has been reported to modulate the development and progression of several types of cancers, while its role in OSCC with LNM has not been reported yet. The expression of IRF2BP2 and its association with LNM were evaluated by immunohistochemistry and qualitative reverse transcription polymerase chain reaction in clinically collected OSCC tissues. Then, loss-of-function and rescue assays were conducted to identify the role of IRF2BP2-mediated fatty acid oxidation (FAO) in the invasion, lymphoinvasion, and epithelial-mesenchymal transition (EMT) in OSCC cells. Importantly, confocal microscope, transmission electron microscope, immunofluorescence, and Western blot were applied to identify the involvement of mitochondrial fission in IRF2BP2-regulated FAO. Lastly, the in vivo models were established to evaluate the role of IRF2BP2 in OSCC. IRF2BP2 overexpression has been associated with LNM in OSCC, whose knockdown inhibited invasion, lymphoinvasion, and EMT of OSCC cells, as well as retarded FAO rate with CPT1A downregulation. And CPT1A overexpression rescued invasion, lymphoinvasion, and induced EMT in IRF2BP2-silenced OSCC cells. Mechanically, IRF2BP2 accelerated mitochondrial fission by contributing to Drp1 S616 phosphorylation and mitochondrial localization, resulting in the upregulation of CPT1A. In addition, IRF2BP2 knockdown significantly inhibited tumor growth and LNM in vivo. The highly expressed IRF2BP2 may induce the phosphorylation and mitochondrial translocation of Drp1 to activate mitochondrial fission, which upregulated CPT1A expression and FAO rate, resulting in LNM in OSCC. This highlighted a potential therapeutic vulnerability for the treatment of LNM+ OSCC via targeting IRF2BP2-FAO.
Collapse
Affiliation(s)
- Xin Pang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tian-Jiao Li
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rong-Jia Shi
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zi-Xin Wan
- Department of Oral Pathology, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Yang Tang
- Department of Oral Pathology, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Ling Tang
- Department of Oral Pathology, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin-Hua Liang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
5
|
Wang L, Yi S, Teng Y, Li W, Cai J. Role of the tumor microenvironment in the lymphatic metastasis of cervical cancer (Review). Exp Ther Med 2023; 26:486. [PMID: 37753293 PMCID: PMC10518654 DOI: 10.3892/etm.2023.12185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/15/2023] [Indexed: 09/28/2023] Open
Abstract
Lymphatic metastasis is the primary type of cervical cancer metastasis and is associated with an extremely poor prognosis in patients. The tumor microenvironment primarily includes cancer-associated fibroblasts, tumor-associated macrophages, myeloid-derived suppressor cells, immune and inflammatory cells, and blood and lymphatic vascular networks, which can promote the establishment of lymphatic metastatic sites within immunosuppressive microenvironments or promote lymphatic metastasis by stimulating lymphangiogenesis and epithelial-mesenchymal transformation. As the most important feature of the tumor microenvironment, hypoxia plays an essential role in lymph node metastasis. In this review, the known mechanisms of hypoxia, and the involvement of stromal components and immune inflammatory cells in the tumor microenvironment of lymphatic metastasis of cervical cancer are discussed. Additionally, a summary of the clinical trials targeting the tumor microenvironment for the treatment of cervical cancer is provided, emphasizing the potential and challenges of immunotherapy.
Collapse
Affiliation(s)
- Lufang Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shuyan Yi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yun Teng
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine; Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province; Institute of Laboratory Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Wenhan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
6
|
Chen Z, Han F, Du Y, Shi H, Zhou W. Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:70. [PMID: 36797231 PMCID: PMC9935926 DOI: 10.1038/s41392-023-01332-8] [Citation(s) in RCA: 424] [Impact Index Per Article: 212.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/20/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Having a hypoxic microenvironment is a common and salient feature of most solid tumors. Hypoxia has a profound effect on the biological behavior and malignant phenotype of cancer cells, mediates the effects of cancer chemotherapy, radiotherapy, and immunotherapy through complex mechanisms, and is closely associated with poor prognosis in various cancer patients. Accumulating studies have demonstrated that through normalization of the tumor vasculature, nanoparticle carriers and biocarriers can effectively increase the oxygen concentration in the tumor microenvironment, improve drug delivery and the efficacy of radiotherapy. They also increase infiltration of innate and adaptive anti-tumor immune cells to enhance the efficacy of immunotherapy. Furthermore, drugs targeting key genes associated with hypoxia, including hypoxia tracers, hypoxia-activated prodrugs, and drugs targeting hypoxia-inducible factors and downstream targets, can be used for visualization and quantitative analysis of tumor hypoxia and antitumor activity. However, the relationship between hypoxia and cancer is an area of research that requires further exploration. Here, we investigated the potential factors in the development of hypoxia in cancer, changes in signaling pathways that occur in cancer cells to adapt to hypoxic environments, the mechanisms of hypoxia-induced cancer immune tolerance, chemotherapeutic tolerance, and enhanced radiation tolerance, as well as the insights and applications of hypoxia in cancer therapy.
Collapse
Affiliation(s)
- Zhou Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Fangfang Han
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yan Du
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Huaqing Shi
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China. .,Lanzhou University Sencond Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
7
|
The Anti-Tubercular Aminolipopeptide Trichoderin A Displays Selective Toxicity against Human Pancreatic Ductal Adenocarcinoma Cells Cultured under Glucose Starvation. Pharmaceutics 2023; 15:pharmaceutics15010287. [PMID: 36678914 PMCID: PMC9866366 DOI: 10.3390/pharmaceutics15010287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma remains a highly debilitating condition with no effective disease-modifying interventions. In our search for natural products with promising anticancer activity, we identified the aminolipopeptide trichoderin A as a potential candidate. While it was initially isolated as an antitubercular peptide, we provide evidence that it is also selectively toxic against BxPC-3 and PANC-1 human pancreatic ductal adenocarcinoma cells cultured under glucose deprivation. This has critical implications for the pancreatic ductal adenocarcinoma, which is characterized by nutrient deprivation due to its hypovascularized network. We have also successfully simplified the trichoderin A peptide backbone, allowing greater accessibility to the peptide for further biological testing. In addition, we also conducted a preliminary investigation into the role of peptide lipidation at the N-terminus. This showed that analogues with longer fatty acyl chains exhibited superior cytotoxicity than those with shorter acyl chains. Further structural optimization of trichoderin A is anticipated to improve its biological activity, whilst ongoing mechanistic studies to elucidate its intracellular mechanism of action are conducted in parallel.
Collapse
|
8
|
Ray SK, Mukherjee S. Interaction Among Noncoding RNAs, DNA Damage Reactions, and Genomic Instability in the Hypoxic Tumor: Is it Therapeutically Exploitable Practice? Curr Mol Med 2023; 23:200-215. [PMID: 35048804 DOI: 10.2174/1566524022666220120123557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 02/08/2023]
Abstract
Hypoxia is a classical function of the tumor's microenvironment with a substantial effect on the development and therapeutic response of cancer. When put in hypoxic environments, cells undergo several biological reactions, including activation of signaling pathways that control proliferation, angiogenesis, and death. These pathways have been adapted by cancer cells to allow tumors to survive and even develop in hypoxic conditions, and poor prognosis is associated with tumor hypoxia. The most relevant transcriptional regulator in response to hypoxia, Hypoxia-inducible factor-1 alpha (HIF-1α), has been shown to modulate hypoxic gene expression and signaling transduction networks significantly. The significance of non-coding RNAs in hypoxic tumor regions has been revealed in an increasing number of studies over the past few decades. In regulating hypoxic gene expression, these hypoxia-responsive ncRNAs play pivotal roles. Hypoxia, a general characteristic of the tumor's microenvironment, significantly affects the expression of genes and is closely associated with the development of cancer. Indeed, the number of known hypoxia-associated lncRNAs has increased dramatically, demonstrating the growing role of lncRNAs in cascades and responses to hypoxia signaling. Decades of research have helped us create an image of the shift in hypoxic cancer cells' DNA repair capabilities. Emerging evidence suggests that hypoxia can trigger genetic instability in cancer cells because of microenvironmental tumor stress. Researchers have found that critical genes' expression is coordinately repressed by hypoxia within the DNA damage and repair pathways. In this study, we include an update of current knowledge on the presentation, participation, and potential clinical effect of ncRNAs in tumor hypoxia, DNA damage reactions, and genomic instability, with a specific emphasis on their unusual cascade of molecular regulation and malignant progression induced by hypoxia.
Collapse
Affiliation(s)
| | - Sukhes Mukherjee
- Department of Biochemistry All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020. India
| |
Collapse
|
9
|
Lomakin A, Svedlund J, Strell C, Gataric M, Shmatko A, Rukhovich G, Park JS, Ju YS, Dentro S, Kleshchevnikov V, Vaskivskyi V, Li T, Bayraktar OA, Pinder S, Richardson AL, Santagata S, Campbell PJ, Russnes H, Gerstung M, Nilsson M, Yates LR. Spatial genomics maps the structure, nature and evolution of cancer clones. Nature 2022; 611:594-602. [PMID: 36352222 PMCID: PMC9668746 DOI: 10.1038/s41586-022-05425-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/07/2022] [Indexed: 11/10/2022]
Abstract
Genome sequencing of cancers often reveals mosaics of different subclones present in the same tumour1-3. Although these are believed to arise according to the principles of somatic evolution, the exact spatial growth patterns and underlying mechanisms remain elusive4,5. Here, to address this need, we developed a workflow that generates detailed quantitative maps of genetic subclone composition across whole-tumour sections. These provide the basis for studying clonal growth patterns, and the histological characteristics, microanatomy and microenvironmental composition of each clone. The approach rests on whole-genome sequencing, followed by highly multiplexed base-specific in situ sequencing, single-cell resolved transcriptomics and dedicated algorithms to link these layers. Applying the base-specific in situ sequencing workflow to eight tissue sections from two multifocal primary breast cancers revealed intricate subclonal growth patterns that were validated by microdissection. In a case of ductal carcinoma in situ, polyclonal neoplastic expansions occurred at the macroscopic scale but segregated within microanatomical structures. Across the stages of ductal carcinoma in situ, invasive cancer and lymph node metastasis, subclone territories are shown to exhibit distinct transcriptional and histological features and cellular microenvironments. These results provide examples of the benefits afforded by spatial genomics for deciphering the mechanisms underlying cancer evolution and microenvironmental ecology.
Collapse
Affiliation(s)
- Artem Lomakin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
- Wellcome Sanger Institute, Hinxton, UK
- Division of AI in Oncology, German Cancer Research Centre DKFZ, Heidelberg, Germany
| | - Jessica Svedlund
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Carina Strell
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Milana Gataric
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
- Wellcome Sanger Institute, Hinxton, UK
| | - Artem Shmatko
- Division of AI in Oncology, German Cancer Research Centre DKFZ, Heidelberg, Germany
| | - Gleb Rukhovich
- Wellcome Sanger Institute, Hinxton, UK
- Division of AI in Oncology, German Cancer Research Centre DKFZ, Heidelberg, Germany
| | - Jun Sung Park
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
- Wellcome Sanger Institute, Hinxton, UK
- Division of AI in Oncology, German Cancer Research Centre DKFZ, Heidelberg, Germany
| | - Young Seok Ju
- Laboratory of Cancer Genomics, GSMSE, KAIST, Daejeon, Korea
| | - Stefan Dentro
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
- Wellcome Sanger Institute, Hinxton, UK
- Division of AI in Oncology, German Cancer Research Centre DKFZ, Heidelberg, Germany
| | | | | | - Tong Li
- Wellcome Sanger Institute, Hinxton, UK
| | | | - Sarah Pinder
- Guys and St Thomas' NHS Trust, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | | | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | | | - Hege Russnes
- Department of Pathology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK.
- Division of AI in Oncology, German Cancer Research Centre DKFZ, Heidelberg, Germany.
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden.
| | | |
Collapse
|
10
|
Kawai T, Matsuo M, Takakusagi Y, Saito K, Hyodo F, Devasahayam N, Matsumoto S, Kishimoto S, Yasui H, Yamamoto K, Krishna MC. Continuous monitoring of postirradiation reoxygenation and cycling hypoxia using electron paramagnetic resonance imaging. NMR IN BIOMEDICINE 2022; 35:e4783. [PMID: 35661282 PMCID: PMC9482554 DOI: 10.1002/nbm.4783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/17/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Reoxygenation has a significant impact on the tumor response to radiotherapy. With developments in radiotherapy technology, the relevance of the reoxygenation phenomenon in treatment efficacy has been a topic of interest. Evaluating the reoxygenation in the tumor microenvironment throughout the course of radiation therapy is important in developing effective treatment strategies. In the current study, we used electron paramagnetic resonance imaging (EPRI) to directly map and quantify the partial oxygen pressure (pO2 ) in tumor tissues. Human colorectal cancer cell lines, HT29 and HCT116, were used to induce tumor growth in female athymic nude mice. Tumors were irradiated with 3, 10, or 20 Gy using an x-ray irradiator. Prior to each EPRI scan, magnetic resonance imaging (MRI) was performed to obtain T2-weighted anatomical images for reference. The differences in the mean pO2 were determined through two-tailed Student's t-test and one-way analysis of variance. The median pO2 60 min after irradiation was found to be lower in HCT116 than in HT29 (9.1 ± 1.5 vs. 14.0 ± 1.0 mmHg, p = 0.045). There was a tendency for delayed and incomplete recovery of pO2 in the HT29 tumor when a higher dose of irradiation (10 and 20 Gy) was applied. Moreover, there was a dose-dependent increase in the hypoxic areas (pO2 < 10 mmHg) 2 and 24 h after irradiation in all groups. In addition, an area that showed pO2 fluctuation between hypoxia and normoxia (pO2 > 10 mmHg) was also identified surrounding the region with stable hypoxia, and it slightly enlarged after recovery from acute hypoxia. In conclusion, we demonstrated the reoxygenation phenomenon in an in vivo xenograft model study using EPRI. These findings may lead to new knowledge regarding the reoxygenation process and possibilities of a new radiation therapy concept, namely, reoxygenation-based radiation therapy.
Collapse
Affiliation(s)
- Tatsuya Kawai
- Radiation Oncology BranchNational Cancer InstituteBethesdaMarylandUSA
- Department of RadiologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Masayuki Matsuo
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
- Department of RadiologyGifu UniversityGifuJapan
| | - Yoichi Takakusagi
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
- Institute for Quantum Life ScienceNational Institutes for Quantum Science and TechnologyChiba‐cityJapan
| | - Keita Saito
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
| | - Fuminori Hyodo
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
- Department of Radiology, Frontier Science for ImagingGifu UniversityGifuJapan
| | | | - Shingo Matsumoto
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
- Division of Bioengineering and Bioinformatics, Graduate School of Information Science and TechnologyHokkaido UniversityHokkaidoJapan
| | - Shun Kishimoto
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
| | - Hironobu Yasui
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary MedicineHokkaido UniversityHokkaidoJapan
| | | | - Murali C. Krishna
- Radiation Biology BranchNational Cancer InstituteBethesdaMarylandUSA
| |
Collapse
|
11
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
12
|
Takei J, Fukasawa N, Tanaka T, Yamamoto Y, Tamura R, Sasaki H, Akasaki Y, Kamata Y, Murahashi M, Shimoda M, Murayama Y. Impact of Neoadjuvant Bevacizumab on Neuroradiographic Response and Histological Findings Related to Tumor Stemness and the Hypoxic Tumor Microenvironment in Glioblastoma: Paired Comparison Between Newly Diagnosed and Recurrent Glioblastomas. Front Oncol 2022; 12:898614. [PMID: 35785200 PMCID: PMC9247463 DOI: 10.3389/fonc.2022.898614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/17/2022] [Indexed: 12/03/2022] Open
Abstract
Background Previously, we reported that bevacizumab (Bev) produces histological and neuroradiographic alterations including changes in tumor oxygenation, induction of an immunosupportive tumor microenvironment, and inhibition of stemness. To confirm how those effects vary during Bev therapy, paired samples from the same patients with newly diagnosed glioblastoma (GBM) who received preoperative neoadjuvant Bev (neoBev) were investigated with immunohistochemistry before and after recurrence. Methods Eighteen samples from nine patients with newly diagnosed GBM who received preoperative neoBev followed by surgery and chemoradiotherapy and then autopsy or salvage surgery after recurrence were investigated. The expression of carbonic anhydrase 9 (CA9), hypoxia-inducible factor-1 alpha (HIF-1α), nestin, and Forkhead box M1 (FOXM1) was evaluated with immunohistochemistry. For comparison between neoBev and recurrent tumors, we divided the present cohort into two groups based on neuroradiographic response: good and poor responders (GR and PR, respectively) to Bev were defined by the tumor regression rate on T1-weighted images with gadolinium enhancement (T1Gd) and fluid-attenuated inversion recovery images. Patterns of recurrence after Bev therapy were classified as cT1 flare-up and T2-diffuse/T2-circumscribed. Furthermore, we explored the possibility of utilizing FOXM1 as a biomarker of survival in this cohort. Results A characteristic “pseudo-papillary”-like structure containing round-shaped tumor cells clustered adjacent to blood vessels surrounded by spindle-shaped tumor cells was seen only in recurrent tumors. Tumor cells at the outer part of the “pseudo-papillary” structure were CA9-positive (CA9+)/HIF-1α+, whereas cells at the inner part of this structure were CA9−/HIF-1α+ and nestin+/FOXM1+. CA9 and HIF-1α expression was lower in T1Gd-GR and decreased in the “T2-circumscribed/T2-diffuse” pattern compared with the “T1 flare-up” pattern, suggesting that tumor oxygenation was frequently observed in T1Gd-GR in initial tumors and in the “T2-circumscribed/T2-diffuse” pattern in recurrent tumors. FOXM1 low-expression tumors tended to have a better prognosis than that of FOXM1 high-expression tumors. Conclusion A “pseudo-papillary” structure was seen in recurrent GBM after anti-vascular endothelial growth factor therapy. Bev may contribute to tumor oxygenation, leading to inhibition of stemness and correlation with a neuroimaging response during Bev therapy. FOXM1 may play a role as a biomarker of survival during Bev therapy.
Collapse
Affiliation(s)
- Jun Takei
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Nei Fukasawa
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
- Department of Neurosurgery, Jikei University School of Medicine Kashiwa Hospital, Kashiwa, Japan
- *Correspondence: Toshihide Tanaka,
| | - Yohei Yamamoto
- Department of Neurosurgery, Jikei University School of Medicine Daisan Hospital, Tokyo, Japan
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Hikaru Sasaki
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Yuko Kamata
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Mutsunori Murahashi
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Hypoxia Selectively Increases a SMAD3 Signaling Axis to Promote Cancer Cell Invasion. Cancers (Basel) 2022; 14:cancers14112751. [PMID: 35681731 PMCID: PMC9179584 DOI: 10.3390/cancers14112751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 01/27/2023] Open
Abstract
Transforming growth factor β (TGFβ) plays a paradoxical role in cancer, first inhibiting then promoting its progression, a duality that poses a real challenge for the development of effective TGFβ-targeted therapies. The major TGFβ downstream effectors, SMAD2 and SMAD3, display both distinct and overlapping functions and accumulating evidence suggests that their activation ratio may contribute to the dual effect of TGFβ. However, the mechanisms responsible for their selective activation remain poorly understood. Here, we provide experimental evidence that hypoxia induces the pro-invasive arm of TGFβ signaling through a selective increase in SMAD3 interaction with SMAD-Anchor for Receptor Activation (SARA). This event relies on HDAC6-dependent SMAD3 bioavailability, as well as increased SARA recruitment to EEA1+ endosomes. A motility gene expression study indicated that SMAD3 selectively increased the expression of ITGB2 and VIM, two genes that were found to be implicated in hypoxia-induced cell invasion and associated with tumor progression and metastasis in cohorts of cancer patients. Furthermore, CAM xenograft assays show the significant benefit of selective inhibition of the SMAD3 signaling pathway as opposed to global TGFβ inhibition in preventing tumor progression. Overall, these results suggest that fine-tuning of the pro-invasive HDAC6-SARA-SMAD3 axis could be a better strategy towards effective cancer treatments.
Collapse
|
14
|
Wadsworth BJ, Lee CM, Bennewith KL. Transiently hypoxic tumour cell turnover and radiation sensitivity in human tumour xenografts. Br J Cancer 2022; 126:1616-1626. [PMID: 35031765 PMCID: PMC9130130 DOI: 10.1038/s41416-021-01691-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/24/2021] [Accepted: 12/23/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Solid tumour perfusion can be unstable, creating transiently hypoxic cells that can contribute to radiation resistance. We investigated the in vivo lifetime of transiently hypoxic tumour cells and chronically hypoxic tumour cells during tumour growth and following irradiation. METHODS Hypoxic cells in SiHa and WiDr human tumour xenografts were labelled using pimonidazole and EF5, and turnover was quantified as the loss of labelled cells over time. The perfusion-modifying drug pentoxifylline was used to reoxygenate transiently hypoxic cells prior to hypoxia marker administration or irradiation. RESULTS Chronically hypoxic cells constantly turnover in SiHa and WiDr tumours, with half-lives ranging from 42-82 h and significant numbers surviving >96 h. Transiently hypoxic cells constitute 26% of the total hypoxic cells in WiDr tumours. These transiently hypoxic cells survive at least 24 h, but then rapidly turnover with a half-life of 34 h and are undetectable 72 h after labelling. Transiently hypoxic cells are radiation-resistant, although vascular dysfunction induced by 10 Gy of ionising radiation preferentially kills transiently hypoxic cells. CONCLUSIONS Transiently hypoxic tumour cells survive up to 72 h in WiDr tumours and are radiation-resistant, although transiently hypoxic cells are sensitive to vascular dysfunction induced by high doses of ionising radiation.
Collapse
Affiliation(s)
- Brennan J. Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Che-Min Lee
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| | - Kevin L. Bennewith
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
15
|
Shang C, Li Y, He T, Liao Y, Du Q, Wang P, Qiao J, Guo H. The prognostic miR-532-5p-correlated ceRNA-mediated lipid droplet accumulation drives nodal metastasis of cervical cancer. J Adv Res 2021; 37:169-184. [PMID: 35499057 PMCID: PMC9040090 DOI: 10.1016/j.jare.2021.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/15/2022] Open
Abstract
The prognostic miR-532-5p inhibited epithelial-mesenchymal transition and lymphangiogenesis by regulating lipid droplets accumulation. miR-532-5p-correlated ceRNA network in which LINC01410 directly bound to miR-532-5p effectively functioned as a sponge for miR-532-5p to disinhibit its target gene-FASN. Combined therapy with miR-532-5p and FASN inhibitor-orlistat blocked lymph node metastasis and tumor growth.
Introduction The prognosis for cervical cancer (CC) patients with lymph node metastasis (LNM) is extremely poor. Lipid droplets (LDs) have a pivotal role in promoting tumor metastasis. The crosstalk mechanism between LDs and LNM modulated in CC remains largely unknown. Objectives This study aimed to construct a miRNA-dependent progonostic model for CC patients and investigate whether miR-532-5p has a biological impact on LNM by regualting LDs accumulation. Methods LASSO-Cox regression was applied to establish a prognostic prediction model. miR-532-5p had the lowest P-value in RNA expression (P < 0.001) and prognostic prediction (P < 0.0001) and was selected for further study. The functional role of the prognostic miR-532-5p-correlated competing endogenous RNA (ceRNA) network was investigated to clarify the crosstalk between LDs and LNM. The underlying mechanism was determined using site-directed mutagenesis, dual luciferase reporter assays, RNA immunoprecipitation assays, and rescue experiments. A xenograft LNM model was established to evaluate the effect of miR-532-5p and orlistat combination therapy on tumor growth and LNM. Results A novel 5-miRNAs prognostic signature was constructed to better predict the prognosis of CC patient. Further study demonstrated that miR-532-5p inhibited epithelial-mesenchymal transition and lymphangiogenesis by regulating LDs accumulation. Interestingly, we also found that LDs accumulation promoted cell metastasis in vitro. Mechanistically, we demonstrated a miR-532-5p-correlated ceRNA network in which LINC01410 was bound directly to miR-532-5p and effectively functioned as miR-532-5p sponge to disinhibit its target gene-fatty acid synthase (FASN). Combined therapy with miR-532-5p and FASN inhibitor-orlistat further inhibited tumor growth and LNM in vivo. Conclusion Our findings highlight a LD accumulation-dependent mechanism of miR-532-5p-modulated LNM and support treatment with miR-532-5p/orlistat as novel strategy for treating patients with LNM in CC.
Collapse
Affiliation(s)
- Chunliang Shang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, 100191 Beijing, China
| | - Yuan Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, 100191 Beijing, China
| | - Tianhui He
- Department of Obstetrics and Gynecology, Peking University Third Hospital, 100191 Beijing, China
| | - Yuandong Liao
- Department of Obstetrics and Gynecology, The First Affliated Hospital, Sun Yat-sen University, 510080 Guangzhou, China
| | - Qiqiao Du
- Department of Obstetrics and Gynecology, The First Affliated Hospital, Sun Yat-sen University, 510080 Guangzhou, China
| | - Pan Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, 100191 Beijing, China
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, 100191 Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, 100191 Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, 100191 Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, 100191 Beijing, China
- Corresponding authors at: Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Key Laboratory of Assisted, Beijing 100191, China (J. Qiao).
| | - Hongyan Guo
- Department of Obstetrics and Gynecology, Peking University Third Hospital, 100191 Beijing, China
- Corresponding authors at: Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Key Laboratory of Assisted, Beijing 100191, China (J. Qiao).
| |
Collapse
|
16
|
Vaginal drug delivery approaches for localized management of cervical cancer. Adv Drug Deliv Rev 2021; 174:114-126. [PMID: 33857555 DOI: 10.1016/j.addr.2021.04.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/10/2021] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
Cervical cancer or cervical intraepithelial neoplasia (CIN) remain a major public health problem among women globally. Traditional methods such as surgery are often associated with possible complications which may impact future pregnancies and childbirth especially for young female patients. Vagina with a high contact surface is a suitable route for the local and systemic delivery of drugs but its abundant mucus in continuous exchange presents a barrier for the popularization of conventional vaginal formulations including suppositories, gel, patch, creams and so on. So the development of new pharmaceutical forms based on nanotechnology became appealing owing to its several advantages such as mucosa penetration, bioadhesion, controlled drug release, and decreased adverse effects. This review provided an overview of the development of topical treatment of cervical cancer or CIN through vaginal drug delivery ranging from conventional vaginal formulations to new nanocarriers to the newly developed phototherapy and gene therapy, analyzing the problems faced by current methods used, and advising the developing trend in future. The methods of establishing preclinical animal model are also discussed.
Collapse
|
17
|
Sznurkowska MK, Aceto N. The gate to metastasis: key players in cancer cell intravasation. FEBS J 2021; 289:4336-4354. [PMID: 34077633 PMCID: PMC9546053 DOI: 10.1111/febs.16046] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/19/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Metastasis is a leading cause of cancer‐related death and consists of a sequence of events including tumor expansion, intravasation of cancer cells into the circulation, survival in the bloodstream, extravasation at distant sites, and subsequent organ colonization. Particularly, intravasation is a process whereby cancer cells transverse the endothelium and leave the primary tumor site, pioneering the metastatic cascade. The identification of those mechanisms that trigger the entry of cancer cells into the bloodstream may reveal fundamentally novel ways to block metastasis at its start. Multiple factors have been implicated in cancer progression, yet, signals that unequivocally provoke the detachment of cancer cells from the primary tumor are still under investigation. Here, we discuss the role of intrinsic properties of cancer cells, tumor microenvironment, and mechanical cues in the intravasation process, outlining studies that suggest the involvement of various factors and highlighting current understanding and open questions in the field.
Collapse
Affiliation(s)
- Magdalena K Sznurkowska
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel and University Hospital Basel, Switzerland
| | - Nicola Aceto
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel and University Hospital Basel, Switzerland.,Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Switzerland
| |
Collapse
|
18
|
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: 69] [Impact Index Per Article: 17.3] [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
|
19
|
Vedenko A, Panara K, Goldstein G, Ramasamy R, Arora H. Tumor Microenvironment and Nitric Oxide: Concepts and Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1277:143-158. [PMID: 33119871 DOI: 10.1007/978-3-030-50224-9_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cancer tissue exists not as a single entity, but as a combination of different cellular phenotypes which, taken together, dramatically contribute to the entirety of their ecosystem, collectively termed as the tumor microenvironment (TME). The TME is composed of both immune and nonimmune cell types, stromal components, and vasculature-all of which cooperate to promote cancer progression. Not all immune cells, however, are immune-suppressive; some of them can promote the immune microenvironment to fight the invading and uncontrollably dividing cell populations at the initial stages of tumor growth. Yet, many of these processes and cellular phenotypes fall short, and the immune ecosystem more often than not ends up stabilizing in favor of the "resistant" resident cells that begin clonal expansion and may progress to metastatic forms. Stromal components, making up the extracellular matrix and basement membrane, are also not the most innocuous: CAFs embedded throughout secrete proteases that allow the onset of one of the most invasive processes-angiogenesis-through destruction of the ECM and the basement membrane. Vasculature formation, because of angiogenesis, is the largest invader of the TME and the reason metastasis happens. Vasculature is so sporadic and omnipresent in the TME that most drug therapies are mainly focused on stopping this uncontrollable process. As the tumor continues to grow, different processes are constantly supplying it with the ingredients favorable for tumor progression and eventual metastasis. For example, angiogenesis promotes blood vessel formation that will allow the bona fide escape of tumor cells to take place. Another process like hypoxia will present itself in several forms throughout the tumor (mild or acute, cycling or permanent), starting mechanisms such as epithelial to mesenchymal transitions (EMT) of resident cells and inadvertently placing the cells in such a stressful condition that production of ROS and DNA damage is unavoidable. DNA damage can induce mutagenicity while allowing resistant cells to survive. This is where drugs and treatments can subsequently suffer in effectiveness. Finally, another molecule has just surfaced as being a very important player in the TME: nitric oxide. Often overlooked and equated with ROS and initially assigned in the category of pathogenic molecules, nitric oxide can definitely do some damage by causing metabolic reprogramming and promotion of immunosuppressive phenotypes at low concentrations. However, its actions seem to be extremely dose-dependent, and this issue has become a hot target of current treatment goals. Shockingly, nitric oxide, although omnipresent in the TME, can have a positive effect on targeting the TME broadly. Thus, while the TME is a myriad of cellular phenotypes and a combination of different tumor-promoting processes, each process is interconnected into one whole: the tumor microenvironment.
Collapse
Affiliation(s)
- Anastasia Vedenko
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Kush Panara
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Gabriella Goldstein
- College of Health Professions and Sciences, University of Central Florida, Orlando, FL, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Himanshu Arora
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
20
|
Xing S, Tian Z, Zheng W, Yang W, Du N, Gu Y, Yin J, Liu H, Jia X, Huang D, Liu W, Deng M. Hypoxia downregulated miR-4521 suppresses gastric carcinoma progression through regulation of IGF2 and FOXM1. Mol Cancer 2021; 20:9. [PMID: 33407516 PMCID: PMC7786912 DOI: 10.1186/s12943-020-01295-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) show considerable promise as therapeutic agents to improve tumor treatment, as they have been revealed as crucial modulators in tumor progression. However, our understanding of their roles in gastric carcinoma (GC) metastasis is limited. Here, we aimed to identify novel miRNAs involved in GC metastasis and explored their regulatory mechanisms and therapeutic significance in GC. METHODS The microRNA expression profiles of GC tumors at different stages and at different metastasis statuses were compared respectively using the stomach adenocarcinoma (STAD) miRNASeq dataset in TCGA. Using the above method, miR-4521 was picked out for further study. miR-4521 expression in GC tissues was examined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and in situ hybridization (ISH). Highly and lowly invasive cell sublines were established using a repetitive transwell assay. Gain-of-function and loss-of-function analyses were performed to investigate the functions of miR-4521 and its upstream and downstream regulatory mechanisms in vitro and in vivo. Moreover, we investigated the therapeutic role of miR-4521 in a mouse xenograft model. RESULTS In this study, we found that miR-4521 expression was downregulated in GC tissues compared with adjacent normal tissues and that its downregulation was positively correlated with advanced clinical stage, metastasis status and poor patient prognosis. Functional experiments revealed that miR-4521 inhibited GC cell invasion and metastasis in vitro and in vivo. Further studies showed that hypoxia repressed miR-4521 expression via inducing ETS1 and miR-4521 mitigated hypoxia-mediated metastasis, while miR-4521 inactivated the AKT/GSK3β/Snai1 pathway by targeting IGF2 and FOXM1, thereby inhibiting the epithelial-mesenchymal transition (EMT) process and metastasis. In addition, we demonstrated that therapeutic delivery of synthetic miR-4521 suppressed gastric carcinoma progression in vivo. CONCLUSIONS Our results suggest an important role for miR-4521 in regulating GC metastasis and hypoxic response of tumor cells as well as the therapeutic significance of this miRNA in GC.
Collapse
Affiliation(s)
- Shan Xing
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhi Tian
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Wenying Zheng
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China
| | - Wenjuan Yang
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China
| | - Nan Du
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yixue Gu
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China
| | - Jiang Yin
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China
| | - Hao Liu
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China
| | - Xiaoting Jia
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China
| | - Donglan Huang
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China.
| | - Wanli Liu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Min Deng
- Affiliated Cancer Hosipital & Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", No.78, Hengzhigang Road, Guangzhou, 510095, China.
| |
Collapse
|
21
|
Bader SB, Dewhirst MW, Hammond EM. Cyclic Hypoxia: An Update on Its Characteristics, Methods to Measure It and Biological Implications in Cancer. Cancers (Basel) 2020; 13:E23. [PMID: 33374581 PMCID: PMC7793090 DOI: 10.3390/cancers13010023] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Regions of hypoxia occur in most if not all solid cancers. Although the presence of tumor hypoxia is a common occurrence, the levels of hypoxia and proportion of the tumor that are hypoxic vary significantly. Importantly, even within tumors, oxygen levels fluctuate due to changes in red blood cell flux, vascular remodeling and thermoregulation. Together, this leads to cyclic or intermittent hypoxia. Tumor hypoxia predicts for poor patient outcome, in part due to increased resistance to all standard therapies. However, it is less clear how cyclic hypoxia impacts therapy response. Here, we discuss the causes of cyclic hypoxia and, importantly, which imaging modalities are best suited to detecting cyclic vs. chronic hypoxia. In addition, we provide a comparison of the biological response to chronic and cyclic hypoxia, including how the levels of reactive oxygen species and HIF-1 are likely impacted. Together, we highlight the importance of remembering that tumor hypoxia is not a static condition and that the fluctuations in oxygen levels have significant biological consequences.
Collapse
Affiliation(s)
- Samuel B. Bader
- Department of Oncology, The Oxford Institute for Radiation Oncology, Oxford University, Oxford OX3 7DQ, UK;
| | - Mark W. Dewhirst
- Radiation Oncology Department, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ester M. Hammond
- Department of Oncology, The Oxford Institute for Radiation Oncology, Oxford University, Oxford OX3 7DQ, UK;
| |
Collapse
|
22
|
Ardaševa A, Gatenby RA, Anderson ARA, Byrne HM, Maini PK, Lorenzi T. A Mathematical Dissection of the Adaptation of Cell Populations to Fluctuating Oxygen Levels. Bull Math Biol 2020; 82:81. [PMID: 32556703 DOI: 10.1007/s11538-020-00754-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/27/2020] [Indexed: 12/17/2022]
Abstract
The disordered network of blood vessels that arises from tumour angiogenesis results in variations in the delivery of oxygen into the tumour tissue. This brings about regions of chronic hypoxia (i.e. sustained low oxygen levels) and regions with alternating periods of low and relatively higher oxygen levels, and makes it necessary for cancer cells to adapt to fluctuating environmental conditions. We use a phenotype-structured model to dissect the evolutionary dynamics of cell populations exposed to fluctuating oxygen levels. In this model, the phenotypic state of every cell is described by a continuous variable that provides a simple representation of its metabolic phenotype, ranging from fully oxidative to fully glycolytic, and cells are grouped into two competing populations that undergo heritable, spontaneous phenotypic variations at different rates. Model simulations indicate that, depending on the rate at which oxygen is consumed by the cells, dynamic nonlinear interactions between cells and oxygen can stimulate chronic hypoxia and cycling hypoxia. Moreover, the model supports the idea that under chronic-hypoxic conditions lower rates of phenotypic variation lead to a competitive advantage, whereas higher rates of phenotypic variation can confer a competitive advantage under cycling-hypoxic conditions. In the latter case, the numerical results obtained show that bet-hedging evolutionary strategies, whereby cells switch between oxidative and glycolytic phenotypes, can spontaneously emerge. We explain how these results can shed light on the evolutionary process that may underpin the emergence of phenotypic heterogeneity in vascularised tumours.
Collapse
Affiliation(s)
- Aleksandra Ardaševa
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Alexander R A Anderson
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Tommaso Lorenzi
- School of Mathematics and Statistics, University of St Andrews, St Andrews, KY16 9SS, UK. .,Department of Mathematical Sciences "G. L. Lagrange", Dipartimento di Eccellenza, 2018-2022, Politecnico di Torino, 10129, Turin, Italy.
| |
Collapse
|
23
|
Wang J, Tannous BA, Poznansky MC, Chen H. CXCR4 antagonist AMD3100 (plerixafor): From an impurity to a therapeutic agent. Pharmacol Res 2020; 159:105010. [PMID: 32544428 DOI: 10.1016/j.phrs.2020.105010] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/22/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
Abstract
AMD3100 (plerixafor), a CXCR4 antagonist, has opened a variety of avenues for potential therapeutic approaches in different refractory diseases. The CXCL12/CXCR4 axis and its signaling pathways are involved in diverse disorders including HIV-1 infection, tumor development, non-Hodgkin lymphoma, multiple myeloma, WHIM Syndrome, and so on. The mechanisms of action of AMD3100 may relate to mobilizing hematopoietic stem cells, blocking infection of X4 HIV-1, increasing circulating neutrophils, lymphocytes and monocytes, reducing myeloid-derived suppressor cells, and enhancing cytotoxic T-cell infiltration in tumors. Here, we first revisit the pharmacological discovery of AMD3100. We then review monotherapy of AMD3100 and combination use of AMD3100 with other agents in various diseases. Among those, we highlight the perspective of AMD3100 as an immunomodulator to regulate immune responses particularly in the tumor microenvironment and synergize with other therapeutics. All the pre-clinical studies support the clinical testing of the monotherapy and combination therapies with AMD3100 and further development for use in humans.
Collapse
Affiliation(s)
- Jingzhe Wang
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Huabiao Chen
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA; Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Perucho JAU, Chiu KWH, Wong EMF, Tse KY, Chu MMY, Chan LWC, Pang H, Khong PL, Lee EYP. Diffusion-weighted magnetic resonance imaging of primary cervical cancer in the detection of sub-centimetre metastatic lymph nodes. Cancer Imaging 2020; 20:27. [PMID: 32252829 PMCID: PMC7137185 DOI: 10.1186/s40644-020-00303-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 03/20/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Magnetic resonance imaging (MRI) has limited accuracy in detecting pelvic lymph node (PLN) metastasis. This study aimed to examine the use of intravoxel incoherent motion (IVIM) in classifying pelvic lymph node (PLN) involvement in cervical cancer patients. METHODS Fifty cervical cancer patients with pre-treatment magnetic resonance imaging (MRI) were examined for PLN involvement by one subspecialist and one non-subspecialist radiologist. PLN status was confirmed by positron emission tomography or histology. The tumours were then segmented by both radiologists. Kruskal-Wallis tests were used to test for differences between diffusion tumour volume (DTV), apparent diffusion coefficient (ADC), pure diffusion coefficient (D), and perfusion fraction (f) in patients with no malignant PLN involvement, those with sub-centimetre and size-significant PLN metastases. These parameters were then considered as classifiers for PLN involvement, and were compared with the accuracies of radiologists. RESULTS Twenty-one patients had PLN involvement of which 10 had sub-centimetre metastatic PLNs. DTV increased (p = 0.013) while ADC (p = 0.015), and f (p = 0.006) decreased as the nodal status progressed from no malignant involvement to sub-centimetre and then size-significant PLN metastases. In determining PLN involvement, a classification model (DTV + f) had similar accuracies (80%) as the non-subspecialist (76%; p = 0.73) and subspecialist (90%; p = 0.31). However, in identifying patients with sub-centimetre PLN metastasis, the model had higher accuracy (90%) than the non-subspecialist (30%; p = 0.01) but had similar accuracy with the subspecialist (90%, p = 1.00). Interobserver variability in tumour delineation did not significantly affect the performance of the classification model. CONCLUSION IVIM is useful in determining PLN involvement but the added value decreases with reader experience.
Collapse
Affiliation(s)
- Jose Angelo Udal Perucho
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Pok Fu Lam, Hong Kong
| | - Keith Wan Hang Chiu
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Pok Fu Lam, Hong Kong
| | - Esther Man Fung Wong
- Department of Radiology, Pamela Youde Nethersole Eastern Hospital, 3 Lok Man Road, Chai Wan, Hong Kong
| | - Ka Yu Tse
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 6/F, Professorial Block, Queen Mary Hospital, Pok Fu Lam Road, Pok Fu Lam, Hong Kong
| | - Mandy Man Yee Chu
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 6/F, Professorial Block, Queen Mary Hospital, Pok Fu Lam Road, Pok Fu Lam, Hong Kong
| | - Lawrence Wing Chi Chan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Room Y934, 9/F, Lee Shau Kee Building, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Herbert Pang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, G/F, Patrick Manson Building (North Wing), 7 Sassoon Road, Pok Fu Lam, Hong Kong
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Pok Fu Lam, Hong Kong
| | - Elaine Yuen Phin Lee
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Pok Fu Lam, Hong Kong
| |
Collapse
|
25
|
Zhen S, Lu J, Liu YH, Chen W, Li X. Synergistic antitumor effect on cervical cancer by rational combination of PD1 blockade and CRISPR-Cas9-mediated HPV knockout. Cancer Gene Ther 2020; 27:168-178. [PMID: 31455836 DOI: 10.1038/s41417-019-0131-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/03/2019] [Accepted: 08/06/2019] [Indexed: 11/08/2022]
Abstract
Targeted therapy results in objective responses in cervical cancer. However, the responses are short. In contrast, treatment with immune checkpoint inhibitors results in a lower responses rate, but the responses tend to be more durable. Based on these findings, we hypothesized that HPV16 E6/E7-targeted therapy may synergize with the PD-1 pathway blockade to enhance antitumor activity. To test hypothesis, we described for the first time the effects of the CRISPR/Cas9 that was targeted to the HPV and PD1 in vitro and in vivo. Our data showed that gRNA/cas9 targeted HPV16 E6/E7 induced cervical cancer cell SiHa apoptosis, and suggested that overexpression of PD-L1, induced by HPV16 E6/E7, may be responsible for lymphocyte dysfunction. In established SiHa cell- xenografted humanized SCID mice, Administration of gRNA-PD-1 together with gRNA-HPV16 E6/E7 treatment improved the survival and suppressed the tumor growth obviously. In addition, combination treatment increased the population of dendritic cells, CD8+ and CD4+ T lymphocyte cells. According, it enhanced the expression of Th1-associated immune-stimulating genes while reducing the transcription of regulatory/suppressive immune genes, reshaping tumor microenvironment from an immunosuppressive to a stimulatory state. These results demonstrate potent synergistic effects of combination therapy using HPV16 E6/E7-targeted therapy and immune checkpoint blockade PD1, supporting a direct translation of this combination strategy in clinic for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Shuai Zhen
- Center for Translational Medicine, Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Center of Medical Genetics, Northwest Women and Children's Hospital, Xi'an, China
| | - Jiaojiao Lu
- Center for Translational Medicine, Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Center of Medical Genetics, Northwest Women and Children's Hospital, Xi'an, China
| | - Yun-Hui Liu
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wei Chen
- Center of Laboratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xu Li
- Center for Translational Medicine, Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Center of Medical Genetics, Northwest Women and Children's Hospital, Xi'an, China.
| |
Collapse
|
26
|
Delprat V, Tellier C, Demazy C, Raes M, Feron O, Michiels C. Cycling hypoxia promotes a pro-inflammatory phenotype in macrophages via JNK/p65 signaling pathway. Sci Rep 2020; 10:882. [PMID: 31964911 PMCID: PMC6972721 DOI: 10.1038/s41598-020-57677-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023] Open
Abstract
Cycling hypoxia (cyH), also called intermittent hypoxia, occurs in solid tumors and affects different cell types in the tumor microenvironment and in particular the tumor-associated macrophages (TAMs). As cyH and TAMs both favor tumor progression, we investigated whether cyH could drive the pro-tumoral phenotype of macrophages. Here, the effects of cyH on human THP-1 macrophages and murine bone marrow-derived macrophages (BMDM), either unpolarized M0, or polarized in M1 or M2 phenotype were studied. In M0 macrophages, cyH induced a pro-inflammatory phenotype characterized by an increase in TNFα and IL-8/MIP-2 secretion. CyH amplified the pro-inflammatory phenotype of M1 macrophages evidenced by an increased pro-inflammatory cytokine secretion and pro-inflammatory gene expression. Furthermore, cyH increased c-jun activation in human M0 macrophages and highly increased c-jun and NF-κB activation in M1 macrophages. C-jun and p65 are implicated in the effects of cyH on M0 and M1 macrophages since inhibition of their activation prevented the cyH pro-inflammatory effects. In conclusion, we demonstrated that cyH induces or amplifies a pro-inflammatory phenotype in M0 and M1 macrophages by activating JNK/p65 signaling pathway. These results highlight a specific role of cyH in the amplification of tumor-related inflammation by modulating the inflammatory phenotype of macrophages.
Collapse
Affiliation(s)
- Victor Delprat
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium
| | - Céline Tellier
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium
| | - Catherine Demazy
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium
| | - Martine Raes
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH 5349), Institut de recherche expérimentale et clinique, UCLouvain, 57 Avenue Hippocrate, B-1200, Brussels, Belgium
| | - Carine Michiels
- Unit of Biochemistry and Cellular Biology (URBC), Namur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium.
| |
Collapse
|
27
|
Wu H, Song S, Yan A, Guo X, Chang L, Xu L, Hu L, Kuang M, Liu B, He D, Zhao R, Wang L, Wu X, Gu J, Ruan Y. RACK1 promotes the invasive activities and lymph node metastasis of cervical cancer via galectin-1. Cancer Lett 2020; 469:287-300. [DOI: 10.1016/j.canlet.2019.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/19/2022]
|
28
|
Paul T, Basu S. Vascular onco-therapies targeting continuous and intermittent intra-tumor hypoxia. JOURNAL OF CANCER RESEARCH AND PRACTICE 2020. [DOI: 10.4103/jcrp.jcrp_9_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
29
|
Huang W, Wang K, An Y, Meng H, Gao Y, Xiong Z, Yan H, Wang Q, Cai X, Yang X, Zhang B, Chen Q, Yang X, Tian J, Zhang S. In vivo three-dimensional evaluation of tumour hypoxia in nasopharyngeal carcinomas using FMT-CT and MSOT. Eur J Nucl Med Mol Imaging 2019; 47:1027-1038. [PMID: 31705175 PMCID: PMC7101302 DOI: 10.1007/s00259-019-04526-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 09/05/2019] [Indexed: 11/26/2022]
Abstract
Purpose Accurate evaluation of hypoxia is particularly important in patients with nasopharyngeal carcinoma (NPC) undergoing radiotherapy. The aim of this study was to propose a novel imaging strategy for quantitative three-dimensional (3D) evaluation of hypoxia in a small animal model of NPC. Methods A carbonic anhydrase IX (CAIX)-specific molecular probe (CAIX-800) was developed for imaging of hypoxia. Mouse models of subcutaneous, orthotopic, and spontaneous lymph node metastasis from NPC (5 mice per group) were established to assess the imaging strategy. A multi-modality imaging method that consisted of a hybrid combination of fluorescence molecular tomography-computed tomography (FMT-CT) and multispectral optoacoustic tomography (MSOT) was used for 3D quantitative evaluation of tumour hypoxia. Magnetic resonance imaging, histological examination, and immunohistochemical analysis were used as references for comparison and validation. Results In the early stage of NPC (2 weeks after implantation), FMT-CT enabled precise 3D localisation of the hypoxia biomarker with high sensitivity. At the advanced stage (6 weeks after implantation), MSOT allowed multispectral analysis of the biomarker and haemoglobin molecules with high resolution. The combination of high sensitivity and high resolution from FMT-CT and MSOT could not only detect hypoxia in small-sized NPCs but also visualise the heterogeneity of hypoxia in 3D. Conclusions Integration of FMT-CT and MSOT could allow comprehensive and quantifiable evaluation of hypoxia in NPC. These findings may potentially benefit patients with NPC undergoing radiotherapy in the future. A novel multimodality imaging strategy for three-dimensional evaluation of tumour hypoxia in an orthotopic model of nasopharyngeal carcinoma. ![]()
Electronic supplementary material The online version of this article (10.1007/s00259-019-04526-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenhui Huang
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, No. 163, Huangpu West Road, Tianhe District, Guangzhou, Guangdong, 510632, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Haidian District, Beijing, 100190, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Haidian District, Beijing, 100190, China
| | - Yu An
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Haidian District, Beijing, 100190, China
| | - Hui Meng
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Haidian District, Beijing, 100190, China
| | - Yuan Gao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Haidian District, Beijing, 100190, China
| | - Zhiyuan Xiong
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, No. 163, Huangpu West Road, Tianhe District, Guangzhou, Guangdong, 510632, China.,Department of Chemical and Bio-molecular Engineering, The university of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Hao Yan
- Engineering Laboratory for Functionalized Carbon Materials, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Qian Wang
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xuekang Cai
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Road, Xicheng District, Beijing, 100034, China
| | - Xin Yang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Haidian District, Beijing, 100190, China
| | - Bin Zhang
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, No. 163, Huangpu West Road, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Qiuying Chen
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, No. 163, Huangpu West Road, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Road, Xicheng District, Beijing, 100034, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Haidian District, Beijing, 100190, China. .,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China.
| | - Shuixing Zhang
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, No. 163, Huangpu West Road, Tianhe District, Guangzhou, Guangdong, 510632, China.
| |
Collapse
|
30
|
Muz B, Bandara N, Mpoy C, Sun J, Alhallak K, Azab F, Rogers BE, Azab AK. CXCR4-targeted PET imaging using 64Cu-AMD3100 for detection of Waldenström Macroglobulinemia. Cancer Biol Ther 2019; 21:52-60. [PMID: 31571524 DOI: 10.1080/15384047.2019.1665405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Objective: Waldenström Macroglobulinemia (WM) is a rare B-cell malignancy characterized by secretion of immunoglobulin M and cancer infiltration in the bone marrow. Chemokine receptor such as CXCR4 and hypoxic condition in the bone marrow play crucial roles in cancer cell trafficking, homing, adhesion, proliferation, survival, and drug resistance. Herein, we aimed to use CXCR4 as a potential biomarker to detect hypoxic-metastatic WM cells in the bone marrow and in the circulation by using CXCR4-detecting radiopharmaceutical.Methods: We radiolabeled a CXCR4-inhibitor (AMD3100) with 64Cu and tested its binding to WM cells with different levels of CXCR4 expression using gamma counter in vitro. The accumulation of this radiopharmaceutical tracer was tested in vivo in subcutaneous and intratibial models using PET/CT scan. In addition, PBMCs spiked with different amounts of WM cells ex vivo were detected using gamma counting.Results: In vitro, 64Cu-AMD3100 binding to WM cell lines demonstrated a direct correlation with the level of CXCR4 expression, which was increased in cells cultured in hypoxia with elevated levels of CXCR4, and decreased in cells with CXCR4 and HIF-1α knockout. Moreover, 64Cu-AMD3100 detected localized and circulating CXCR4high WM cells with high metastatic potential.Conclusions: In conclusion, we developed a molecularly targeted system, 64Cu-AMD3100, which binds to CXCR4 and specifically detects WM cells with hypoxic phenotype and metastatic potential in the subcutaneous and intratibial models. These preliminary findings using CXCR4-detecting PET radiopharmaceutical tracer indicate a potential technology to predict high-risk patients for the progression to WM due to metastatic potential.
Collapse
Affiliation(s)
- Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nilantha Bandara
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Kinan Alhallak
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Feda Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Buck E Rogers
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
31
|
Hypoxia Downregulates LPP3 and Promotes the Spatial Segregation of ATX and LPP1 During Cancer Cell Invasion. Cancers (Basel) 2019; 11:cancers11091403. [PMID: 31546971 PMCID: PMC6769543 DOI: 10.3390/cancers11091403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/07/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022] Open
Abstract
Hypoxia is a common characteristic of advanced solid tumors and a potent driver of tumor invasion and metastasis. Recent evidence suggests the involvement of autotaxin (ATX) and lysophosphatidic acid receptors (LPARs) in cancer cell invasion promoted by the hypoxic tumor microenvironment; however, the transcriptional and/or spatiotemporal control of this process remain unexplored. Herein, we investigated whether hypoxia promotes cell invasion by affecting the main enzymes involved in its production (ATX) and degradation (lipid phosphate phosphatases, LPP1 and LPP3). We report that hypoxia not only modulates the expression levels of lysophosphatidic acid (LPA) regulatory enzymes but also induces their significant spatial segregation in a variety of cancers. While LPP3 expression was downregulated by hypoxia, ATX and LPP1 were asymmetrically redistributed to the leading edge and to the trailing edge, respectively. This was associated with the opposing roles of ATX and LPPs in cell invasion. The regulated expression and compartmentalization of these enzymes of opposing function can provide an effective way to control the generation of an LPA gradient that drives cellular invasion and migration in the hypoxic zones of tumors.
Collapse
|
32
|
Saxena K, Jolly MK. Acute vs. Chronic vs. Cyclic Hypoxia: Their Differential Dynamics, Molecular Mechanisms, and Effects on Tumor Progression. Biomolecules 2019; 9:E339. [PMID: 31382593 PMCID: PMC6722594 DOI: 10.3390/biom9080339] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Hypoxia has been shown to increase the aggressiveness and severity of tumor progression. Along with chronic and acute hypoxic regions, solid tumors contain regions of cycling hypoxia (also called intermittent hypoxia or IH). Cyclic hypoxia is mimicked in vitro and in vivo by periodic exposure to cycles of hypoxia and reoxygenation (H-R cycles). Compared to chronic hypoxia, cyclic hypoxia has been shown to augment various hallmarks of cancer to a greater extent: angiogenesis, immune evasion, metastasis, survival etc. Cycling hypoxia has also been shown to be the major contributing factor in increasing the risk of cancer in obstructive sleep apnea (OSA) patients. Here, we first compare and contrast the effects of acute, chronic and intermittent hypoxia in terms of molecular pathways activated and the cellular processes affected. We highlight the underlying complexity of these differential effects and emphasize the need to investigate various combinations of factors impacting cellular adaptation to hypoxia: total duration of hypoxia, concentration of oxygen (O2), and the presence of and frequency of H-R cycles. Finally, we summarize the effects of cycling hypoxia on various hallmarks of cancer highlighting their dependence on the abovementioned factors. We conclude with a call for an integrative and rigorous analysis of the effects of varying extents and durations of hypoxia on cells, including tools such as mechanism-based mathematical modelling and microfluidic setups.
Collapse
Affiliation(s)
- Kritika Saxena
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
33
|
Ali M, Kowkuntla S, Delloro DJ, Galambos C, Hathi D, Janz S, Shokeen M, Tripathi C, Xu H, Yuk J, Zhan F, Tomasson MH, Bates ML. Chronic intermittent hypoxia enhances disease progression in myeloma-resistant mice. Am J Physiol Regul Integr Comp Physiol 2019; 316:R678-R686. [PMID: 30892915 DOI: 10.1152/ajpregu.00388.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Obesity is the only known modifiable risk factor for multiple myeloma (MM), an incurable cancer of bone marrow plasma cells. The mechanism linking the two is unknown. Obesity is associated with an increased risk of sleep apnea, which results in chronic intermittent hypoxia (CIH), and drives solid tumor aggressiveness. Given the link between CIH and solid tumor progression, we tested the hypothesis that CIH drives the proliferation of MM cells in culture and their engraftment and progression in vivo. Malignant mouse 5TGM1 cells were cultured in CIH, static hypoxia, or normoxia as a control in custom, gas-permeable plates. Typically MM-resistant C57BL/6J mice were exposed to 10 h/day CIH (AHI = 12/h), static hypoxia, or normoxia for 7 days, followed by injection with 5TGM1 cells and an additional 28 days of exposure. CIH and static hypoxia slowed the growth of 5TGM1 cells in culture. CIH-exposed mice developed significantly more MM than controls (67 vs. 12%, P = 0.005), evidenced by hindlimb paralysis, gammopathy, bone lesions, and bone tumor formation. Static hypoxia was not a significant driver of MM progression and did not reduce survival (P = 0.117). Interestingly, 5TGM1 cells preferentially engrafted in the bone marrow and promoted terminal disease in CIH mice, despite a lower tumor burden, compared with the positive controls. These first experiments in the context of hematological cancer demonstrate that CIH promotes MM through mechanisms distinct from solid tumors and that sleep apnea may be a targetable risk factor in patients with or at risk for blood cancer.
Collapse
Affiliation(s)
- Mahmoud Ali
- Department of Internal Medicine, Hematology and Oncology Division, University of Iowa , Iowa City, Iowa
| | - Sandeep Kowkuntla
- Department of Health and Human Physiology, University of Iowa , Iowa City, Iowa
| | - Derick J Delloro
- Department of Health and Human Physiology, University of Iowa , Iowa City, Iowa
| | - Csaba Galambos
- Department of Pathology and Laboratory Medicine, University of Colorado School of Medicine and Children's Hospital Colorado , Aurora, Colorado
| | - Deep Hathi
- Department of Radiology, Washington University , St. Louis, Missouri
| | - Siegfried Janz
- Department of Pathology, University of Iowa , Iowa City, Iowa
| | - Monica Shokeen
- Department of Radiology, Washington University , St. Louis, Missouri
| | - Chakrapani Tripathi
- Department of Internal Medicine, Hematology and Oncology Division, University of Iowa , Iowa City, Iowa
| | - Hongwei Xu
- Department of Internal Medicine, Hematology and Oncology Division, University of Iowa , Iowa City, Iowa
| | - Jisung Yuk
- Department of Health and Human Physiology, University of Iowa , Iowa City, Iowa
| | - Fenghuang Zhan
- Department of Internal Medicine, Hematology and Oncology Division, University of Iowa , Iowa City, Iowa
| | - Michael H Tomasson
- Department of Internal Medicine, Hematology and Oncology Division, University of Iowa , Iowa City, Iowa
| | - Melissa L Bates
- Department of Internal Medicine, Hematology and Oncology Division, University of Iowa , Iowa City, Iowa.,Stead Family Department of Pediatrics, University of Iowa , Iowa City, Iowa
| |
Collapse
|
34
|
Minassian LM, Cotechini T, Huitema E, Graham CH. Hypoxia-Induced Resistance to Chemotherapy in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:123-139. [PMID: 31201721 DOI: 10.1007/978-3-030-12734-3_9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A major barrier to the successful management of cancer is the development of resistance to therapy. Chemotherapy resistance can either be an intrinsic property of malignant cells developed prior to therapy, or acquired following exposure to anti-cancer drugs. Given the impact of drug resistance to the overall poor survival of cancer patients, there is an urgent need to better understand the molecular pathways regulating this malignant phenotype. In this chapter we describe some of the molecular pathways that contribute to drug resistance in cancer, the role of a microenvironment deficient in oxygen (hypoxia) in malignant progression, and how hypoxia can be a significant factor in the development of drug resistance. We conclude by proposing potential therapeutic approaches that take advantage of a hypoxic microenvironment to chemosensitize therapy-resistant tumours.
Collapse
Affiliation(s)
- Lori M Minassian
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Erin Huitema
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
35
|
Zhen S, Lu J, Hua L, Liu YH, Chen W, Li X. WITHDRAWN: CRISPR/Cas9 mediated HPV and PD1 inhibition produces a synergistic anti-tumor effect on cervical cancer. Arch Biochem Biophys 2018:S0003-9861(18)30611-8. [PMID: 30315769 DOI: 10.1016/j.abb.2018.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 10/28/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Shuai Zhen
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaojiao Lu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ling Hua
- Rongchang Campus Southwest University, Chongqing, China
| | - Yun-Hui Liu
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wei Chen
- Center of Laboratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
36
|
Zhou CF, Ma J, Huang L, Yi HY, Zhang YM, Wu XG, Yan RM, Liang L, Zhong M, Yu YH, Wu S, Wang W. Cervical squamous cell carcinoma-secreted exosomal miR-221-3p promotes lymphangiogenesis and lymphatic metastasis by targeting VASH1. Oncogene 2018; 38:1256-1268. [PMID: 30254211 PMCID: PMC6363643 DOI: 10.1038/s41388-018-0511-x] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/03/2018] [Accepted: 07/31/2018] [Indexed: 12/20/2022]
Abstract
Cancer-secreted exosomal miRNAs are emerging mediators of cancer-stromal cross-talk in the tumor environment. Our previous miRNAs array of cervical squamous cell carcinoma (CSCC) clinical specimens identified upregulation of miR-221-3p. Here, we show that miR-221-3p is closely correlated with peritumoral lymphangiogenesis and lymph node (LN) metastasis in CSCC. More importantly, miR-221-3p is characteristically enriched in and transferred by CSCC-secreted exosomes into human lymphatic endothelial cells (HLECs) to promote HLECs migration and tube formation in vitro, and facilitate lymphangiogenesis and LN metastasis in vivo according to both gain-of-function and loss-of-function experiments. Furthermore, we identify vasohibin-1 (VASH1) as a novel direct target of miR-221-3p through bioinformatic target prediction and luciferase reporter assay. Re-expression and knockdown of VASH1 could respectively rescue and simulate the effects induced by exosomal miR-221-3p. Importantly, the miR-221-3p-VASH1 axis activates the ERK/AKT pathway in HLECs independent of VEGF-C. Finally, circulating exosomal miR-221-3p levels also have biological function in promoting HLECs sprouting in vitro and are closely associated with tumor miR-221-3p expression, lymphatic VASH1 expression, lymphangiogenesis, and LN metastasis in CSCC patients. In conclusion, CSCC-secreted exosomal miR-221-3p transfers into HLECs to promote lymphangiogenesis and lymphatic metastasis via downregulation of VASH1 and may represent a novel diagnostic biomarker and therapeutic target for metastatic CSCC patients in early stages.
Collapse
Affiliation(s)
- Chen-Fei Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.,Department of Obstetrics and Gynecology, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jing Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.,Department of Obstetrics and Gynecology, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lei Huang
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-Upon-Tyne, NE2 4HH, UK
| | - Hong-Yan Yi
- Department of Obstetrics and Gynecology, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yan-Mei Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangdong Provincial Key Laboratory of Proteomic, Guangzhou, 510515, China
| | - Xiang-Guang Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Rui-Ming Yan
- Department of Obstetrics and Gynecology, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Li Liang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yan-Hong Yu
- Department of Obstetrics and Gynecology, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangdong Provincial Key Laboratory of Proteomic, Guangzhou, 510515, China.
| | - Wei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China. .,Department of Obstetrics and Gynecology, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
37
|
Gillies RJ, Brown JS, Anderson ARA, Gatenby RA. Eco-evolutionary causes and consequences of temporal changes in intratumoural blood flow. Nat Rev Cancer 2018; 18:576-585. [PMID: 29891961 PMCID: PMC6441333 DOI: 10.1038/s41568-018-0030-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Temporal changes in blood flow are commonly observed in malignant tumours, but the evolutionary causes and consequences are rarely considered. We propose that stochastic temporal variations in blood flow and microenvironmental conditions arise from the eco-evolutionary dynamics of tumour angiogenesis in which cancer cells, as individual units of selection, can influence and respond only to local environmental conditions. This leads to new vessels arising from the closest available vascular structure regardless of the size or capacity of this parental vessel. These dynamics produce unstable vascular networks with unpredictable spatial and temporal variations in blood flow and microenvironmental conditions. Adaptations of evolving populations to temporally varying environments in nature include increased diversity, greater motility and invasiveness, and highly plastic phenotypes, allowing for broad metabolic adaptability and rapid shifts to high rates of proliferation and profound quiescence. These adaptive strategies, when adopted in cancer cells, promote many commonly observed phenotypic properties including those found in the stem phenotype and in epithelial-to-mesenchymal transition. Temporal variations in intratumoural blood flow, which occur through the promotion of cancer cell phenotypes that facilitate both metastatic spread and resistance to therapy, may have substantial clinical consequences.
Collapse
Affiliation(s)
- Robert J Gillies
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Joel S Brown
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Robert A Gatenby
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
38
|
Araos J, Sleeman JP, Garvalov BK. The role of hypoxic signalling in metastasis: towards translating knowledge of basic biology into novel anti-tumour strategies. Clin Exp Metastasis 2018; 35:563-599. [DOI: 10.1007/s10585-018-9930-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/13/2018] [Indexed: 02/06/2023]
|
39
|
Intermittent hypoxia induces a metastatic phenotype in breast cancer. Oncogene 2018; 37:4214-4225. [DOI: 10.1038/s41388-018-0259-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 03/16/2018] [Accepted: 03/21/2018] [Indexed: 01/07/2023]
|
40
|
Tátrai E, Bartal A, Gacs A, Paku S, Kenessey I, Garay T, Hegedűs B, Molnár E, Cserepes MT, Hegedűs Z, Kucsma N, Szakács G, Tóvári J. Cell type-dependent HIF1 α-mediated effects of hypoxia on proliferation, migration and metastatic potential of human tumor cells. Oncotarget 2018; 8:44498-44510. [PMID: 28562340 PMCID: PMC5546497 DOI: 10.18632/oncotarget.17806] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/24/2017] [Indexed: 12/19/2022] Open
Abstract
Tumor hypoxia promotes neoangiogenesis and contributes to the radio- and chemotherapy resistant and aggressive phenotype of cancer cells. However, the migratory response of tumor cells and the role of small GTPases regulating the organization of cytoskeleton under hypoxic conditions have yet to be established. Accordingly, we measured the proliferation, migration, RhoA activation, the mRNA and protein levels of hypoxia inducible factor-1alpha (HIF-1α) and three small G-proteins, Rac1, cdc42 and RhoA in a panel of five human tumor cell lines under normoxic and hypoxic conditions. Importantly, HT168-M1 human melanoma cells with high baseline migration capacity showed increased HIF-1α and small GTPases expression, RhoA activation and migration under hypoxia. These activities were blocked by anti- HIF-1α shRNA. Moreover, the in vivo metastatic potential was promoted by hypoxia mimicking CoCl2 treatment and reduced upon inhibition of HIF-1α in a spleen to liver colonization experiment. In contrast, HT29 human colon cancer cells with low migration capacity showed limited response to in vitro hypoxia. The expression of the small G-proteins decreased both at mRNA and protein levels and the RhoA activation was reduced. Nevertheless, the number of lung or liver metastatic colonies disseminating from orthotopic HT29 grafts did not change upon CoCl2 or chetomin treatment. Our data demonstrates that the hypoxic environment induces cell-type dependent changes in the levels and activation of small GTPases and results in varying migratory and metastasis promoting responses in different human tumor cell lines.
Collapse
Affiliation(s)
- Enikő Tátrai
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Alexandra Bartal
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary.,Central Pharmacy, National Institute of Oncology, Budapest, Hungary
| | - Alexandra Gacs
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Sándor Paku
- st Institute of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,Tumor Progression Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - István Kenessey
- nd Department of Pathology, Semmelweis University, Budapest, Hungary.,National Cancer Registry, National Institute of Oncology, Budapest, Hungary
| | - Tamás Garay
- nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Balázs Hegedűs
- Tumor Progression Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary.,Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, Essen, Germany
| | - Eszter Molnár
- nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Mihály T Cserepes
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary.,Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Semmelweis University, Budapest, Hungary
| | - Zita Hegedűs
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Nóra Kucsma
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gergely Szakács
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| |
Collapse
|
41
|
Abstract
In 2012, cancer affected 14.1 million people worldwide and was responsible for 8.2 million deaths. The disease predominantly affects aged populations and is one of the leading causes of death in most western countries. In tumors, the aggressive growth of the neoplastic cell population and associated overexpression of pro-angiogenic factors lead to the development of disorganized blood vessel networks that are structurally and functionally different from normal vasculature. A disorganized labyrinth of vessels that are immature, tortuous and hyperpermeable typifies tumor vasculature. Functionally, the ability of the tumor vasculature to deliver nutrients and remove waste products is severely diminished. A critical consequence of the inadequate vascular networks in solid tumors is the development of regions of hypoxia [low oxygen tensions typically defined as oxygen tensions (pO2 values) < 10 mm Hg]. Tumor cells existing in such hypoxic environments have long been known to be resistant to anticancer therapy, display an aggressive phenotype, and promote tumor progression and dissemination. This review discusses the physiological basis of hypoxia, methods of detection, and strategies to overcome the resulting therapy resistance.
Collapse
Affiliation(s)
- Veronica S Hughes
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| | - Jennifer M Wiggins
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| | - Dietmar W Siemann
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| |
Collapse
|
42
|
Lecavalier-Barsoum M, Chaudary N, Han K, Koritzinsky M, Hill R, Milosevic M. Targeting the CXCL12/CXCR4 pathway and myeloid cells to improve radiation treatment of locally advanced cervical cancer. Int J Cancer 2018; 143:1017-1028. [PMID: 29417588 DOI: 10.1002/ijc.31297] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/10/2018] [Accepted: 02/01/2018] [Indexed: 12/17/2022]
Abstract
Cervical cancer is the fourth most commonly diagnosed cancer and the fourth leading cause of cancer death in women worldwide. Approximately half of cervical cancer patients present with locally advanced disease, for which surgery is not an option. These cases are nonetheless potentially curable with radiotherapy and cisplatin chemotherapy. Unfortunately, some tumours are resistant to treatment, and lymph node and distant recurrences are major problems in patients with advanced disease at diagnosis. New targeted treatments that can overcome treatment resistance and reduce metastases are urgently needed. The CXCL12/CXCR4 chemokine pathway is ubiquitously expressed in many normal tissues and cancers, including cervical cancer. Emerging evidence indicates that it plays a central role in cervical cancer pathogenesis, malignant progression, the development of metastases and radiation treatment response. Pre-clinical studies of standard-of-care fractionated radiotherapy and concurrent weekly cisplatin plus the CXCR4 inhibitor Plerixafor (AMD3100) in patient-derived orthotopic cervical cancer xenografts have shown improved primary tumour response and reduced lymph node metastases with no increase in early or late side effects. These studies have pointed the way forward to future clinical trials of radiotherapy/cisplatin plus Plerixafor or other newly emerging CXCL12 or CXCR4 inhibitors in women with cervical cancer.
Collapse
Affiliation(s)
- Magali Lecavalier-Barsoum
- Department of Oncology, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Canada
| | - Naz Chaudary
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Canada
| | - Kathy Han
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Marianne Koritzinsky
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Richard Hill
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Canada.,Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Michael Milosevic
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
43
|
Abstract
Cancer patients with lymph node (LN) metastases have a worse prognosis than those without nodal disease. However, why LN metastases correlate with reduced patient survival is poorly understood. Recent findings provide insight into mechanisms underlying tumor growth in LNs. Tumor cells and their secreted molecules engage stromal, myeloid, and lymphoid cells within primary tumors and in the lymphatic system, decreasing antitumor immunity and promoting tumor growth. Understanding the mechanisms of cancer survival and growth in LNs is key to designing effective therapy for the eradication of LN metastases. In addition, uncovering the implications of LN metastasis for systemic tumor burden will inform treatment decisions. In this review, we discuss the current knowledge of the seeding, growth, and further dissemination of LN metastases.
Collapse
Affiliation(s)
- Dennis Jones
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, MGH Cancer Center, Massachusetts General Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Ethel R Pereira
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, MGH Cancer Center, Massachusetts General Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Timothy P Padera
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, MGH Cancer Center, Massachusetts General Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| |
Collapse
|
44
|
Amelot A, Terrier LM, Mathon B, Cook AR, Mazeron JJ, Valery CA, Cornu P, Leveque M, Carpentier A. Can anticancer chemotherapy promote the progression of brain metastases? Med Oncol 2018; 35:35. [PMID: 29427159 DOI: 10.1007/s12032-018-1097-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 02/05/2018] [Indexed: 12/31/2022]
Abstract
Brain metastases natural history from one primary tumor type might be accelerated or favored by using certain systemic chemotherapy. A great deal was described in mice and suggested in human with antiangiogenic drugs, but little is known about the metastatic progression generated by the perverse effect of anticancer drugs. A total of 413 patients who underwent treatment for brain metastasis (2013-2016) were included. The identification of all previous anticancer drugs received by patients from primary tumor diagnosis to brain metastases diagnosis was collated. The median value for the time of first appearance of brain metastasis in all patients was 13.1 months (SD 1.77). The values of brain metastasis-free survival (bMFS) for each primary cancer were: 50.9 months (SD 8.8) for breast, 28.5 months (SD 11.4) for digestive, 27.7 months (SD 18.3) for melanoma, 12.3 months (SD 8.3) for kidney, 1.5 months (SD 0.1) for lung and 26.9 months (SD 18.3) for others (p < 0.009). Through Cox multivariate proportional hazard model, we identified that the only independent factors associated with short bMFS were: lung primary tumor [odd ratio (OR) 0.234, CI 95% 0.16-0.42; p < 0.0001] and mitotic spindle inhibitor (taxanes) chemotherapy [OR 0.609, CI 95% 0.50-0.93; p < 0.001]. Contrariwise, breast primary tumor [odd ratio (OR) 2.372, CI 95% 1.29-4.3; p < 0.005] was an independent factor that proved a significantly longer bMFS. We suggest that anticancer drugs, especially taxane and its derivatives, could promote brain metastases, decreasing free survival. Mechanisms are discussed but still need to be determined.
Collapse
Affiliation(s)
- Aymeric Amelot
- Department of Neurosurgery, Groupe Hospitalier Pitié-Salpétrière, APHP, 47-83 Boulevard de l'Hôpital, Batiment Babinski, 75013, Paris, France. .,Université Paris VI - Pierre et Marie Curie, Paris, France.
| | | | - Bertrand Mathon
- Department of Neurosurgery, Groupe Hospitalier Pitié-Salpétrière, APHP, 47-83 Boulevard de l'Hôpital, Batiment Babinski, 75013, Paris, France.,Université Paris VI - Pierre et Marie Curie, Paris, France
| | - Ann-Rose Cook
- Department of Neurosurgery, Hopital Bretonneau, Tours, France
| | - Jean-Jacques Mazeron
- Université Paris VI - Pierre et Marie Curie, Paris, France.,Department of Radiotherapy, Groupe Hospitalier Pitié-Salpétrière, APHP, Paris, France
| | - Charles-Ambroise Valery
- Department of Neurosurgery, Groupe Hospitalier Pitié-Salpétrière, APHP, 47-83 Boulevard de l'Hôpital, Batiment Babinski, 75013, Paris, France.,Université Paris VI - Pierre et Marie Curie, Paris, France
| | - Philippe Cornu
- Department of Neurosurgery, Groupe Hospitalier Pitié-Salpétrière, APHP, 47-83 Boulevard de l'Hôpital, Batiment Babinski, 75013, Paris, France.,Université Paris VI - Pierre et Marie Curie, Paris, France
| | - Marc Leveque
- Department of Neurosurgery, Groupe Hospitalier Pitié-Salpétrière, APHP, 47-83 Boulevard de l'Hôpital, Batiment Babinski, 75013, Paris, France.,Université Paris VI - Pierre et Marie Curie, Paris, France
| | - Alexandre Carpentier
- Department of Neurosurgery, Groupe Hospitalier Pitié-Salpétrière, APHP, 47-83 Boulevard de l'Hôpital, Batiment Babinski, 75013, Paris, France.,Université Paris VI - Pierre et Marie Curie, Paris, France
| |
Collapse
|
45
|
Shang C, Wang W, Liao Y, Chen Y, Liu T, Du Q, Huang J, Liang Y, Liu J, Zhao Y, Guo L, Hu Z, Yao S. LNMICC Promotes Nodal Metastasis of Cervical Cancer by Reprogramming Fatty Acid Metabolism. Cancer Res 2017; 78:877-890. [PMID: 29229603 DOI: 10.1158/0008-5472.can-17-2356] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/16/2017] [Accepted: 12/05/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Chunliang Shang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Wei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yuandong Liao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yili Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Tianyu Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Qiqiao Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Jiaming Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yanchun Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Junxiu Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yunhe Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Luyan Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Zheng Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China.
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China.
| |
Collapse
|
46
|
Wei WF, Han LF, Liu D, Wu LF, Chen XJ, Yi HY, Wu XG, Zhong M, Yu YH, Liang L, Wang W. Orthotopic Xenograft Mouse Model of Cervical Cancer for Studying the Role of MicroRNA-21 in Promoting Lymph Node Metastasis. Int J Gynecol Cancer 2017; 27:1587-1595. [PMID: 28945212 DOI: 10.1097/igc.0000000000001059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cervical cancer is the most frequent cause of gynecologic cancer-associated death worldwide. Animal models that demonstrate metastatic patterns consistent with the clinical course of cervical cancer are urgently needed to conduct studies focused on understanding the mechanisms of the disease and identifying optimal treatments. To address this, we established an orthotopic xenograft model of cervical cancer in female NOD-SCID mice using SiHa and ME180 cell lines stably expressing green fluorescent protein to evaluate the role of microRNA-21 (miR-21) in spontaneous lymph node metastasis in vivo. In this case, SiHa and ME180 cells were transduced by lentivirus to stably express green fluorescent protein and miR-21. Overexpression of miR-21 promoted proliferation, migration, and invasion of SiHa and ME180 cells in vitro. Finally, an orthotopic xenograft model of human cervical cancer was successfully established in NOD-SCID mice. Using this model, we confirmed that overexpression of miR-21 resulted in an increase in the size of primary tumors and in the frequency of spontaneous lymph node metastasis at the time of excision. Therefore, the use of the orthotopic xenograft model should allow for the investigation of novel factors that affect metastasis of cervical cancer and presents an opportunity to evaluate potential therapeutic agents that may inhibit the spread of the disease.
Collapse
Affiliation(s)
- Wen-Fei Wei
- *Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong; †Department of Minimally Invasive Gynecologic Surgery, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai; ‡Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan; and §Department of Obstetrics and Gynecology, Third Affiliated Hospital, and ∥Department of Pathology, Nanfang Hospital, Southern Medical University; and ¶Department of Obstetrics and Gynecology, Shenzhen Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Maley CC, Aktipis A, Graham TA, Sottoriva A, Boddy AM, Janiszewska M, Silva AS, Gerlinger M, Yuan Y, Pienta KJ, Anderson KS, Gatenby R, Swanton C, Posada D, Wu CI, Schiffman JD, Hwang ES, Polyak K, Anderson ARA, Brown JS, Greaves M, Shibata D. Classifying the evolutionary and ecological features of neoplasms. Nat Rev Cancer 2017; 17:605-619. [PMID: 28912577 PMCID: PMC5811185 DOI: 10.1038/nrc.2017.69] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neoplasms change over time through a process of cell-level evolution, driven by genetic and epigenetic alterations. However, the ecology of the microenvironment of a neoplastic cell determines which changes provide adaptive benefits. There is widespread recognition of the importance of these evolutionary and ecological processes in cancer, but to date, no system has been proposed for drawing clinically relevant distinctions between how different tumours are evolving. On the basis of a consensus conference of experts in the fields of cancer evolution and cancer ecology, we propose a framework for classifying tumours that is based on four relevant components. These are the diversity of neoplastic cells (intratumoural heterogeneity) and changes over time in that diversity, which make up an evolutionary index (Evo-index), as well as the hazards to neoplastic cell survival and the resources available to neoplastic cells, which make up an ecological index (Eco-index). We review evidence demonstrating the importance of each of these factors and describe multiple methods that can be used to measure them. Development of this classification system holds promise for enabling clinicians to personalize optimal interventions based on the evolvability of the patient's tumour. The Evo- and Eco-indices provide a common lexicon for communicating about how neoplasms change in response to interventions, with potential implications for clinical trials, personalized medicine and basic cancer research.
Collapse
Affiliation(s)
- Carlo C Maley
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 1001 S. McAllister Ave, Tempe, Arizona 85287, USA
| | - Athena Aktipis
- Department of Psychology, Center for Evolution and Medicine, Arizona State University, 651 E. University Drive, Tempe, Arizona 85287, USA
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Amy M Boddy
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Michalina Janiszewska
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue D740C, Boston, Massachusetts 02215, USA
| | - Ariosto S Silva
- Department of Cancer Imaging and Metabolism, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Marco Gerlinger
- Centre for Evolution and Cancer, Division of Molecular Pathology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Kenneth J Pienta
- Brady Urological Institute, The Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, Maryland 21287, USA
| | - Karen S Anderson
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 1001 S. McAllister Ave, Tempe, Arizona 85287, USA
| | - Robert Gatenby
- Cancer Biology and Evolution Program, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - David Posada
- Department of Biochemistry, Genetics and Immunology and Biomedical Research Center (CINBIO), University of Vigo, Spain; Galicia Sur Health Research Institute, Vigo, 36310, Spain
| | - Chung-I Wu
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois 60637, USA
| | - Joshua D Schiffman
- Departments of Pediatrics and Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, Utah 84108, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University and Duke Cancer Institute, 465 Seeley Mudd Building, Durham, North Carolina 27710, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue D740C, Boston, Massachusetts 02215, USA
| | - Alexander R A Anderson
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Joel S Brown
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Darryl Shibata
- Department of Pathology, Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, NOR2424, Los Angeles, California 90033, USA
| |
Collapse
|
48
|
Interleukin-8 mRNA Expression in Locally Advanced Colorectal Cancer Patients. CURRENT HEALTH SCIENCES JOURNAL 2017; 43:209-213. [PMID: 30595877 PMCID: PMC6284832 DOI: 10.12865/chsj.43.03.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/16/2017] [Indexed: 11/18/2022]
Abstract
Purpose: Interleukin-8 (IL-8) has been proven to promote progression of malignant tumours and control angiogenesis processes. We aim to determine and compare interleukin-8 (IL-8) gene expression level in colorectal tumors (CCR) and peritumoral samples obtained through endoscopic biopsy. Material and methods: Total mRNA was obtained from both tumoral and peritumoral tissue samples collected from patients diagnosed with colorectal cancer. Through Quantitative Real Time PCR, IL-8 gene expression was assessed in both pathologic tissue and adjacent normal mucosa. Results: In our cohort, IL8 expression was higher in adjacent normal mucosa than in tumoral tissue, in all the samples. Further studies on larger groups are required to validate our results.
Collapse
|
49
|
Lyng H, Malinen E. Hypoxia in cervical cancer: from biology to imaging. Clin Transl Imaging 2017; 5:373-388. [PMID: 28804704 PMCID: PMC5532411 DOI: 10.1007/s40336-017-0238-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 06/24/2017] [Indexed: 12/14/2022]
Abstract
PURPOSE Hypoxia imaging may improve identification of cervical cancer patients at risk of treatment failure and be utilized in treatment planning and monitoring, but its clinical potential is far from fully realized. Here, we briefly describe the biology of hypoxia in cervix tumors of relevance for imaging, and evaluate positron emission tomography (PET) and magnetic resonance imaging (MRI) techniques that have shown promise for assessing hypoxia in a clinical setting. We further discuss emerging imaging approaches, and how imaging can play a role in future treatment strategies to target hypoxia. METHODS We performed a PubMed literature search, using keywords related to imaging and hypoxia in cervical cancer, with a particular emphasis on studies correlating imaging with other hypoxia measures and treatment outcome. RESULTS Only a few and rather small studies have utilized PET with tracers specific for hypoxia, and no firm conclusions regarding preferred tracer or clinical potential can be drawn so far. Most studies address indirect hypoxia imaging with dynamic contrast-enhanced techniques. Strong evidences for a role of these techniques in hypoxia imaging have been presented. Pre-treatment images have shown significant association to outcome in several studies, and images acquired during fractionated radiotherapy may further improve risk stratification. Multiparametric MRI and multimodality PET/MRI enable combined imaging of factors of relevance for tumor hypoxia and warrant further investigation. CONCLUSIONS Several imaging approaches have shown promise for hypoxia imaging in cervical cancer. Evaluation in large clinical trials is required to decide upon the optimal modality and approach.
Collapse
Affiliation(s)
- Heidi Lyng
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Medical Physics, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
50
|
Gutsche K, Randi EB, Blank V, Fink D, Wenger RH, Leo C, Scholz CC. Intermittent hypoxia confers pro-metastatic gene expression selectively through NF-κB in inflammatory breast cancer cells. Free Radic Biol Med 2016; 101:129-142. [PMID: 27717868 DOI: 10.1016/j.freeradbiomed.2016.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/14/2016] [Accepted: 10/02/2016] [Indexed: 01/02/2023]
Abstract
Inflammatory breast cancer (IBC) is the most aggressive form of breast cancer. Treatment options are limited and the mechanisms underlying its aggressiveness are poorly understood. Intermittent hypoxia (IH) causes oxidative stress and is emerging as important regulator of tumor metastasis. Vessels in IBC tumors have been shown to be immature, which is a primary cause of IH. We therefore investigated the relevance of IH for the modulation of gene expression in IBC cells in order to assess IH as potential regulator of IBC aggressiveness. Gene array analysis of IBC cells following chronic IH (45-60 days) demonstrated increased expression of pro-metastatic genes of the extracellular matrix, such as tenascin-C (TNC; an essential factor of the metastatic niche) and matrix metalloproteinase 9 (MMP9), and of pro-inflammatory processes, such as cyclooxygenase-2 (COX-2). Investigating the oxidative stress-dependent regulation of TNC, we found a gradual sensitivity on mRNA and protein levels. Oxidative stress activated NF-E2-related factor 2 (Nrf2), c-Jun N-terminal kinase (JNK), c-Jun and nuclear factor κB (NF-κB), but TNC upregulation was only dependent on NF-κB activation. Pharmacological inhibition of inhibitor of NF-κB α (IκBα) phosphorylation as well as overexpression of IκBα prevented TNC, MMP9 and COX-2 induction, whereas the pro-inflammatory cytokine interleukin-1β (IL-1β) increased their expression levels. Analysis of the gene array data showed NF-κB binding sites for 64% of all upregulated genes, linking NF-κB with IH-dependent regulation of pro-metastatic gene expression in IBC cells. Our results provide a first link between intermittent hypoxia and pro-metastatic gene expression in IBC cells, revealing a putative novel mechanism for the high metastatic potential of IBC.
Collapse
Affiliation(s)
- Katrin Gutsche
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland; Department of Gynecology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Elisa B Randi
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Volker Blank
- Lady Davis Institute for Medical Research, Department of Medicine & Department of Physiology, McGill University, Montreal, Quebec, Canada H3T 1E2
| | - Daniel Fink
- Department of Gynecology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Cornelia Leo
- Department Women and Children, Cantonal Hospital Baden, 5404 Baden, Switzerland.
| | - Carsten C Scholz
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|