1
|
Qiu SQ, He XF, Liang XL, Shi GY, Zhao ML, Li F, Wu ZY, Tian J, Zhai TT, Du Y. GLUT1 as a generic biomarker enables near-infrared fluorescence molecular imaging guided precise intraoperative tumor detection in breast cancer. Eur J Nucl Med Mol Imaging 2025; 52:2171-2186. [PMID: 39833507 DOI: 10.1007/s00259-025-07095-4] [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: 10/08/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE Precise tumor excision is important but challenging in breast-conserving surgery (BCS). Tumor-specific fluorescence imaging may be used for intraoperative tumor detection and, therefore, to guide precise tumor excision. The aims of this study are to develop a glucose transporter 1 (GLUT1)-targeted near-infrared fluorescence tracer and evaluate its accuracy for breast cancer detection using fresh surgical breast specimens. METHODS Bioinformatic analysis was performed to compare GLUT1 expression between breast cancer and normal breast tissues. A GLUT1-targeted fluorescence imaging tracer WZB117-CY7.5 was developed. In combination with fluorescence imaging (FMI), its binding specificity to GLUT1 was examined in in vitro breast cancer cell experiments, in vivo 4T1 breast tumor-bearing mouse models, and 60 freshly resected human breast tumor tissues. The diagnostic accuracy of WZB117-CY7.5, was evaluated in fresh specimens derived from 60 patients diagnosed with breast cancer. RESULTS GLUT1 expression is higher in breast cancer tissues compared with normal tissues. WZB117-CY7.5 specifically bound to breast cancer cells in in vitro cell experiments and accumulated in tumor areas in a 4T1 tumor-bearing mice after intravenous injection by FMI. Moreover, WZB117-CY7.5 specifically bound to freshly resected human breast cancer and demonstrated excellent diagnostic performance in discriminating breast cancer, irrespective of cancer subtype, from normal breast tissue on fresh surgically resected breast tissues. CONCLUSIONS WZB117-CY7.5 showed high accuracy in intraoperative breast cancer detection, irrespective of the cancer subtype. This highlights its potential for clinical applications as a generic tracer for fluorescence image-guided surgery (FIGS) in BCS and fluorescence image-guided pathology for tissue sampling.
Collapse
Affiliation(s)
- Si-Qi Qiu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, No.114 Waima Road, Shantou, 515041, Shantou, China
- Clinical Research Center, Shantou Central Hospital, Shantou, 515041, China
| | - Xiao-Feng He
- Shantou University Medical College, Shantou, 515041, China
| | - Xiao-Long Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Guang-Yuan Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China
| | - Meng-Long Zhao
- Clinical Research Center, Shantou Central Hospital, Shantou, 515041, China
| | - Fan Li
- Biobank, Shantou Central Hospital, Shantou, 515041, China
| | - Zhi-Yong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, No.114 Waima Road, Shantou, 515041, Shantou, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China.
- Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100191, China.
| | - Tian-Tian Zhai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, No.7 Raoping Road, Jinping District, Shantou, 515041, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China.
- The University of Chinese Academy of Sciences, Beijing, 100080, China.
| |
Collapse
|
2
|
Carbonic Anhydrase IX (CAIX) Expressing Hypoxic Micro-environment Hampers CD8+ Immune Cell Infiltrate in Breast Carcinoma. Appl Immunohistochem Mol Morphol 2023; 31:26-32. [PMID: 36476599 DOI: 10.1097/pai.0000000000001082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/28/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hypoxia and necrosis are common features of invasive cancer. The dynamic upregulation of carbonic anhydrase IX (CAIX), triggered by hypoxia-inducible factor 1 (HIF-1) is 1 of the mechanisms supporting cellular adaptation to hypoxia in solid tumors, including breast carcinoma. CAIX activity results in extracellular acidosis and in a profound reorganization of the tumor micro-environment, influencing biological behavior and prognosis. The main focus of our study was to evaluate the mass and distribution of the immune infiltrate, more specifically of CD8+ effector T-cells, in relation with tumoral CAIX expression. MATERIALS AND METHODS Formalin-fixed and paraffin-embedded breast carcinoma sections were analyzed following double immunohistochemical staining for CAIX and CD8. Scanned digital slides were evaluated for both labelings, and CD8-related signal was determined within and outside CAIX-positive tumor areas using the HistoQuant (3DHistech) image analysis software. Statistical analysis was performed using GraphPad Prism software. RESULTS Of the 34 breast carcinomas, 18 tested partially positive for CAIX. The remaining 16 cases were used as the CAIX-negative control group. Necrotic foci were generally associated with CAIX overexpression, and tumors exhibiting signs of necrosis had a significantly higher rate of relative CAIX expression compared with samples without necrosis (11.47±5.505 vs. without necrosis 3.765±3.5 P-value=0.0216). On the other hand, no statistically significant difference was found when comparing relative CD8+ lymphocyte counts in cases with necrosis as opposed to those where necrosis was absent (134.7±55.7 vs. 97.70±57.25; P value=0.1579). No difference in gross CD8+ T-lymphocyte infiltrate could be measured between CAIX positive and negative samples (98.48±37.32 vs. 95.99±50 P value=0.5928). However, in CAIX-expressing tumors a statistical correlation between the CD8+ T-lymphocyte infiltrate and the extent of CAIX-positive areas was observed. Within the same tumor, CD8+ T-lymphocyte counts showed a significant difference betweeen CAIX+ and CAIX- areas (13.06±9.4 vs. 135.6±62.2 P value <0.0001). CONCLUSION Our measurements demonstrate for the first time that tumor areas with CAIX expression potentially hamper CD8+ T-lymphocyte infiltration in breast carcinoma. The hypoxia-driven adaptive micro-environment likely interferes with the specific response to biological and immune therapies requiring intact effector T-cell response.
Collapse
|
3
|
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
|
4
|
Kciuk M, Gielecińska A, Mujwar S, Mojzych M, Marciniak B, Drozda R, Kontek R. Targeting carbonic anhydrase IX and XII isoforms with small molecule inhibitors and monoclonal antibodies. J Enzyme Inhib Med Chem 2022; 37:1278-1298. [PMID: 35506234 PMCID: PMC9090362 DOI: 10.1080/14756366.2022.2052868] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carbonic anhydrases IX and CAXII (CAIX/CAXII) are transmembrane zinc metalloproteins that catalyze a very basic but crucial physiological reaction: the conversion of carbon dioxide into bicarbonate with a release of the proton. CA, especially CAIX and CAXII isoforms gained the attention of many researchers interested in anticancer drug design due to pivotal functions of enzymes in the cancer cell metastasis and response to hypoxia, and their expression restricted to malignant cells. This offers an opportunity to develop new targeted therapies with fewer side effects. Continuous efforts led to the discovery of a series of diverse compounds with the most abundant sulphonamide derivatives. Here we review current knowledge considering small molecule and antibody-based targeting of CAIX/CAXII in cancer.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland.,Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Somdutt Mujwar
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| |
Collapse
|
5
|
Miripour ZS, Abbasvandi F, Aghaee P, Shojaeian F, Faramarzpour M, Mohaghegh P, Hoseinpour P, Namdar N, Hassanpour Amiri M, Ghafari H, Parniani M, Kaviani A, Alamdar S, NajafiKhoshnoo S, Sanati H, Mapar M, Sadeghian N, Akbari ME, Yunesian M, Abdolahad M. Human study on cancer diagnostic probe (CDP) for real-time excising of breast positive cavity side margins based on tracing hypoxia glycolysis; checking diagnostic accuracy in non-neoadjuvant cases. Cancer Med 2022; 11:1630-1645. [PMID: 35224879 PMCID: PMC8986141 DOI: 10.1002/cam4.4503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/25/2021] [Accepted: 11/18/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cancer diagnostic probe (CDP) had been developed to detect involved breast cavity side margins in real-time (Miripour et al. Bioeng Transl Med. e10236.). Here, we presented the results of the in vivo human model CDP studies on non-neoadjuvant cases. METHODS This study is a prospective, blind comparison to a gold standard, and the medical group recruited patients. CDP and frozen data were achieved before the permanent pathology experiment. The main outcome of the study is surgical margin status. From November 2018 to April 2020, 202 patients were registered, and 188 were assigned for the study. Breast-conserving surgery at any age or gender, re-surgery due to re-currency, or involved margins are acceptable. Patients must be non-neoadjuvant. The reliability of CDP scoring had been evaluated by the pathology of the scored IMs. Then, three models of the study were designed to compare CDP with the frozen sections. Receiver operating characteristic (ROC) curves and AUC were measured based on the permanent postoperative pathology gold standard. RESULTS A matched clinical diagnostic categorization between the pathological results of the tested IMs and response peaks of CDP on 113 cases, was reported (sensitivity = 97%, specificity = 89.3%, accuracy = 92%, positive predictive value (PPV) = 84.2%, and negative predictive value (NPV) = 98%). Study A showed the independent ability of CDP for IM scoring (sensitivity = 80%, specificity = 90%, accuracy = 90%, PPV = 22.2%, and NPV = 99.2%). Study B showed the complementary role of CDP to cover the missed lesions of frozen sections (sensitivity = 93.8%, specificity = 91%, accuracy = 91%, PPV = 55.6%, and NPV = 99.2%). Study C showed the ability of CDP in helping the pathologist to reduce his/her frozen miss judgment (specificity = 92%, accuracy = 93%, PPV = 42.1%, and NPV = 100%). Results were reported based on the post-surgical permanent pathology gold standard. CONCLUSION CDP scoring ability in intra-operative margin detection was verified on non-neoadjuvant breast cancer patients. Non-invasive real-time diagnosis of IMs with pathological values may make CDP a distinct tool with handheld equipment to increase the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Zohreh Sadat Miripour
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Fereshteh Abbasvandi
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- ATMP DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
- Cancer Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Parisa Aghaee
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Fatemeh Shojaeian
- Cancer Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Mahsa Faramarzpour
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Pooneh Mohaghegh
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | | | - Naser Namdar
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Morteza Hassanpour Amiri
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Hadi Ghafari
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Mohammad Parniani
- Pathology DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
| | - Ahmad Kaviani
- Institute of CancerImam Khomeini HospitalTehran University of Medical SciencesTehranIran
| | - Sedigheh Alamdar
- Department of PathobiologyIran University of Medical SciencesShahid Hemmat HighwayTehranIran
| | - Sahar NajafiKhoshnoo
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Hassan Sanati
- ATMP DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
| | - Mahna Mapar
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Nastaran Sadeghian
- ATMP DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
| | | | - Masud Yunesian
- Department of Environmental HealthSchool of Public HealthTehran University of Medical SciencesTehranIran
- Department of Research Methodology and Data AnalysisInstitute for Environmental ResearchTehran University of Medical SciencesTehranIran
| | - Mohammad Abdolahad
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Institute of CancerImam Khomeini HospitalTehran University of Medical SciencesTehranIran
- UT&TUMS Cancer Electronics Research CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
6
|
Wadsworth BJ, Decotret LR, Villamil C, Yapp D, Wilson D, Benard F, McKenzie M, Bennewith KL. Evaluation of 18F-EF5 for detection of hypoxia in localized adenocarcinoma of the prostate. Acta Oncol 2021; 60:1489-1498. [PMID: 34379579 DOI: 10.1080/0284186x.2021.1959636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND A common feature of solid tumours that are resistant to therapy is the presence of regions with low oxygen content (i.e., hypoxia). Oxygen electrode studies suggest that localized prostate adenocarcinoma is commonly hypoxic, although conflicting data have been reported between immunohistochemical detection of hypoxia-induced proteins in biopsy specimens and positron emission tomography (PET) imaging of 18F-labeled hypoxia reporters. Although the 2-nitroimidazole 18F-EF5 is well-established to label hypoxic tumour cells in pre-clinical tumour models and clinical trials of multiple primary tumour sites, it has yet to be tested in prostate cancer. The purpose of this study was to evaluate the feasibility of using 18F-EF5 to detect hypoxia in clinical prostate tumours. MATERIAL AND METHODS Patients with localized adenocarcinoma of the prostate were recruited for pre-treatment 18F-EF5 PET scans. Immunohistochemistry was conducted on diagnostic biopsies to assess the expression of glucose transporter 1 (GLUT1), osteopontin (OPN), and carbonic anhydrase IX (CAIX). Immunoreactivity scores of staining intensity and frequency were used to indicate the presence of tumour hypoxia. RESULTS We found low tumour-to-muscle ratios of 18F-EF5 uptake that were not consistent with tumour hypoxia, causing early termination of the study. However, we observed GLUT1 and OPN expression in all prostate tumour biopsies, indicating the presence of hypoxia in all tumours. CONCLUSION Our data do not support the use of 18F-EF5 PET to detect hypoxia in prostate adenocarcinoma, and suggest the use of immunohistochemistry to quantify expression of the hypoxia-inducible proteins GLUT1 and OPN as indications of prostate tumour hypoxia.
Collapse
Affiliation(s)
- Brennan J. Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Lisa R. Decotret
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Donald Yapp
- Experimental Therapeutics, BC Cancer, Vancouver, Canada
| | - Don Wilson
- Functional Imaging, BC Cancer, Vancouver, Canada
| | - Francois Benard
- Functional Imaging, BC Cancer, Vancouver, Canada
- Molecular Oncology, BC Cancer, Vancouver, Canada
| | | | - Kevin L. Bennewith
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
7
|
de Heer EC, Jalving M, Harris AL. HIFs, angiogenesis, and metabolism: elusive enemies in breast cancer. J Clin Invest 2020; 130:5074-5087. [PMID: 32870818 PMCID: PMC7524491 DOI: 10.1172/jci137552] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-inducible factors (HIFs) and the HIF-dependent cancer hallmarks angiogenesis and metabolic rewiring are well-established drivers of breast cancer aggressiveness, therapy resistance, and poor prognosis. Targeting of HIF and its downstream targets in angiogenesis and metabolism has been unsuccessful so far in the breast cancer clinical setting, with major unresolved challenges residing in target selection, development of robust biomarkers for response prediction, and understanding and harnessing of escape mechanisms. This Review discusses the pathophysiological role of HIFs, angiogenesis, and metabolism in breast cancer and the challenges of targeting these features in patients with breast cancer. Rational therapeutic combinations, especially with immunotherapy and endocrine therapy, seem most promising in the clinical exploitation of the intricate interplay of HIFs, angiogenesis, and metabolism in breast cancer cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Ellen C. de Heer
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, Netherlands
| | - Mathilde Jalving
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, Netherlands
| | - Adrian L. Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Monti E, Marras E, Prini P, Gariboldi MB. Luteolin impairs hypoxia adaptation and progression in human breast and colon cancer cells. Eur J Pharmacol 2020; 881:173210. [PMID: 32526242 DOI: 10.1016/j.ejphar.2020.173210] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Hypoxia-inducible factors (HIFs) are the force which drives hypoxic cancer cells to a more aggressive and resistant phenotype in a number of solid tumors, including colorectal and breast cancer. Results from recent studies suggest a role for HIF-1 in immune evasion and cancer stem cell phenotype promotion, establishing HIF-1 as a potential therapeutic target. Thus, identifying new compounds that might inhibit HIF1 activity, or at least exert antiproliferative effects that are unaffected by HIF1-dependent adaptations, is an attractive goal for the management of hypoxic tumors. Here we show that the flavonoid luteolin exerts a significant cytotoxic effect on the colon cancer cell line HCT116 and the breast adenocarcinoma cell line MDA-MB231, by inducing both apoptotic and necrotic cell death, and that this effect is not impaired by HIF-1 activation. In these cells, luteolin also stimulates autophagy; however this seems to be part of a protective response, rather than contribute to the cytotoxic effect. Interestingly, luteolin induces a decrease in HIF-1 transcriptional activity. This is accompanied by a decrease in the levels of protein markers of stemness and invasion, and by a reduction of migratory capacity of the cells. Taken together, our results suggest that luteolin could be developed into a useful therapeutic agent aimed at hypoxic tumors.
Collapse
Affiliation(s)
- Elena Monti
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via J.H. Dunant 3, 21100, Varese, VA, Italy
| | - Emanuela Marras
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via J.H. Dunant 3, 21100, Varese, VA, Italy
| | - Pamela Prini
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via J.H. Dunant 3, 21100, Varese, VA, Italy
| | - Marzia Bruna Gariboldi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via J.H. Dunant 3, 21100, Varese, VA, Italy.
| |
Collapse
|
9
|
CAIX forms a transport metabolon with monocarboxylate transporters in human breast cancer cells. Oncogene 2019; 39:1710-1723. [PMID: 31723238 DOI: 10.1038/s41388-019-1098-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
Abstract
Tumor cells rely on glycolysis to meet their elevated demand for energy. Thereby they produce significant amounts of lactate and protons, which are exported via monocarboxylate transporters (MCTs), supporting the formation of an acidic microenvironment. The present study demonstrates that carbonic anhydrase IX (CAIX), one of the major acid/base regulators in cancer cells, forms a protein complex with MCT1 and MCT4 in tissue samples from human breast cancer patients, but not healthy breast tissue. Formation of this transport metabolon requires binding of CAIX to the Ig1 domain of the MCT1/4 chaperon CD147 and is required for CAIX-mediated facilitation of MCT1/4 activity. Application of an antibody, directed against the CD147-Ig1 domain, displaces CAIX from the transporter and suppresses CAIX-mediated facilitation of proton-coupled lactate transport. In cancer cells, this "metabolon disruption" results in a decrease in lactate transport, reduced glycolysis, and ultimately reduced cell proliferation. Taken together, the study shows that carbonic anhydrases form transport metabolons with acid/base transporters in human tumor tissue and that these interactions can be exploited to interfere with tumor metabolism and proliferation.
Collapse
|
10
|
Yu M, Yongzhi H, Chen S, Luo X, Lin Y, Zhou Y, Jin H, Hou B, Deng Y, Tu L, Jian Z. The prognostic value of GLUT1 in cancers: a systematic review and meta-analysis. Oncotarget 2018; 8:43356-43367. [PMID: 28498810 PMCID: PMC5522151 DOI: 10.18632/oncotarget.17445] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Increased glycolysis is one of the hallmarks of cancer. The abnormal expression of glucose transporter 1 (GLUT1) was reported to be associated with resistance to current therapy and poor prognosis. Numerous studies have investigated the correlation between GLUT1 expression and prognosis in cancers, but the conclusions are still controversial. Here, we conducted a meta-analysis to explore the association between GLUT1 and survival in human cancers. PubMed, Springer, Medline, and Cochrane Library were searched carefully to identify eligible studies evaluating prognostic value of GLUT1 in cancers. Twenty-seven studies with 4079 patients were included in the present study. Our pooled results identified that increased expression of GLUT1 was associated with unfavorable overall survival (HR = 1.780, 95% CI = 1.574–.013, p < 0.001)) and poorer disease-free survival (HR = 1.95, 95% CI = 1.229–3.095, p = 0.003). Furthermore, overexpression of GLUT1 linked with poor differentiated tumors (RR = 1.380, 95% CI = 1.086–1.755, p = 0.009; I2 = 72.0%, p < 0.001), positive lymph node metastasis (RR = 1.395, 95% CI = 1.082–1.799, p = 0.010; I2 = 70.8%, p = 0.002) and larger tumor size (RR = 1.405, 95% CI = 1.231–1.603, p < 0.001; I2 = 37.3%, p = 0.093). This systematic review and meta-analysis indicated that the GLUT1 may serve as an ideal prognostic biomarker in various cancers.
Collapse
Affiliation(s)
- Min Yu
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Han Yongzhi
- Department of Dermatology, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Shengying Chen
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaodan Luo
- Department of Infectious Diseases, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Ye Lin
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu Zhou
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Haosheng Jin
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Baohua Hou
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanying Deng
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lei Tu
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhixiang Jian
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
11
|
Acetazolamide-based [ 18 F]-PET tracer: In vivo validation of carbonic anhydrase IX as a sole target for imaging of CA-IX expressing hypoxic solid tumors. Bioorg Med Chem Lett 2018; 28:915-921. [DOI: 10.1016/j.bmcl.2018.01.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/05/2023]
|
12
|
Abstract
In vivo imaging, which enables us to peer deeply within living subjects, is producing tremendous opportunities both for clinical diagnostics and as a research tool. Contrast material is often required to clearly visualize the functional architecture of physiological structures. Recent advances in nanomaterials are becoming pivotal to generate the high-resolution, high-contrast images needed for accurate, precision diagnostics. Nanomaterials are playing major roles in imaging by delivering large imaging payloads, yielding improved sensitivity, multiplexing capacity, and modularity of design. Indeed, for several imaging modalities, nanomaterials are now not simply ancillary contrast entities, but are instead the original and sole source of image signal that make possible the modality's existence. We address the physicochemical makeup/design of nanomaterials through the lens of the physical properties that produce contrast signal for the cognate imaging modality-we stratify nanomaterials on the basis of their (i) magnetic, (ii) optical, (iii) acoustic, and/or (iv) nuclear properties. We evaluate them for their ability to provide relevant information under preclinical and clinical circumstances, their in vivo safety profiles (which are being incorporated into their chemical design), their modularity in being fused to create multimodal nanomaterials (spanning multiple different physical imaging modalities and therapeutic/theranostic capabilities), their key properties, and critically their likelihood to be clinically translated.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Stanford University , 3155 Porter Drive, #1214, Palo Alto, California 94304-5483, United States
| | - Sanjiv Sam Gambhir
- The James H. Clark Center , 318 Campus Drive, First Floor, E-150A, Stanford, California 94305-5427, United States
| |
Collapse
|
13
|
van Kuijk SJA, Yaromina A, Houben R, Niemans R, Lambin P, Dubois LJ. Prognostic Significance of Carbonic Anhydrase IX Expression in Cancer Patients: A Meta-Analysis. Front Oncol 2016; 6:69. [PMID: 27066453 PMCID: PMC4810028 DOI: 10.3389/fonc.2016.00069] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/08/2016] [Indexed: 01/08/2023] Open
Abstract
Hypoxia is a characteristic of many solid tumors and an adverse prognostic factor for treatment outcome. Hypoxia increases the expression of carbonic anhydrase IX (CAIX), an enzyme that is predominantly found on tumor cells and is involved in maintaining the cellular pH balance. Many clinical studies investigated the prognostic value of CAIX expression, but most have been inconclusive, partly due to small numbers of patients included. The present meta-analysis was therefore performed utilizing the results of all clinical studies to determine the prognostic value of CAIX expression in solid tumors. Renal cell carcinoma was excluded from this meta-analysis due to an alternative mechanism of upregulation. 958 papers were identified from a literature search performed in PubMed and Embase. These papers were independently evaluated by two reviewers and 147 studies were included in the analysis. The meta-analysis revealed strong significant associations between CAIX expression and all endpoints: overall survival [hazard ratio (HR) = 1.76, 95% confidence interval (95%CI) 1.58–1.98], disease-free survival (HR = 1.87, 95%CI 1.62–2.16), locoregional control (HR = 1.54, 95%CI 1.22–1.93), disease-specific survival (HR = 1.78, 95%CI 1.41–2.25), metastasis-free survival (HR = 1.82, 95%CI 1.33–2.50), and progression-free survival (HR = 1.58, 95%CI 1.27–1.96). Subgroup analyses revealed similar associations in the majority of tumor sites and types. In conclusion, these results show that patients having tumors with high CAIX expression have higher risk of locoregional failure, disease progression, and higher risk to develop metastases, independent of tumor type or site. The results of this meta-analysis further support the development of a clinical test to determine patient prognosis based on CAIX expression and may have important implications for the development of new treatment strategies.
Collapse
Affiliation(s)
- Simon J A van Kuijk
- Department of Radiation Oncology (MAASTRO Lab), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , Netherlands
| | - Ala Yaromina
- Department of Radiation Oncology (MAASTRO Lab), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , Netherlands
| | - Ruud Houben
- Department of Radiation Oncology, MAASTRO Clinic , Maastricht , Netherlands
| | - Raymon Niemans
- Department of Radiation Oncology (MAASTRO Lab), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO Lab), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , Netherlands
| | - Ludwig J Dubois
- Department of Radiation Oncology (MAASTRO Lab), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , Netherlands
| |
Collapse
|
14
|
Mascini NE, Cheng M, Jiang L, Rizwan A, Podmore H, Bhandari DR, Römpp A, Glunde K, Heeren RMA. Mass Spectrometry Imaging of the Hypoxia Marker Pimonidazole in a Breast Tumor Model. Anal Chem 2016; 88:3107-14. [PMID: 26891127 DOI: 10.1021/acs.analchem.5b04032] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although tumor hypoxia is associated with tumor aggressiveness and resistance to cancer treatment, many details of hypoxia-induced changes in tumors remain to be elucidated. Mass spectrometry imaging (MSI) is a technique that is well suited to study the biomolecular composition of specific tissue regions, such as hypoxic tumor regions. Here, we investigate the use of pimonidazole as an exogenous hypoxia marker for matrix-assisted laser desorption/ionization (MALDI) MSI. In hypoxic cells, pimonidazole is reduced and forms reactive products that bind to thiol groups in proteins, peptides, and amino acids. We show that a reductively activated pimonidazole metabolite can be imaged by MALDI-MSI in a breast tumor xenograft model. Immunohistochemical detection of pimonidazole adducts on adjacent tissue sections confirmed that this metabolite is localized to hypoxic tissue regions. We used this metabolite to image hypoxic tissue regions and their associated lipid and small molecule distributions with MALDI-MSI. We identified a heterogeneous distribution of 1-methylnicotinamide and acetylcarnitine, which mostly colocalized with hypoxic tumor regions. As pimonidazole is a widely used immunohistochemical marker of tissue hypoxia, it is likely that the presented direct MALDI-MSI approach is also applicable to other tissues from pimonidazole-injected animals or humans.
Collapse
Affiliation(s)
| | - Menglin Cheng
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Lu Jiang
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Asif Rizwan
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Helen Podmore
- Thermo Fisher Scientific , Stafford House, 1 Boundary Park, Hemel Hempstead HP2 7GE, Herts, United Kingdom
| | - Dhaka R Bhandari
- TransMIT GmbH · TransMIT Center for Mass Spectrometric Developments , Schubertstrasse 60, 35392 Giessen, Germany
| | - Andreas Römpp
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen , Schubertstrasse 60, 35392 Giessen, Germany
| | - Kristine Glunde
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21231, United States
| | - Ron M A Heeren
- FOM Institute AMOLF , 1098 XG Amsterdam, The Netherlands.,The Maastricht Multimodal Molecular Imaging institute (M4I) , 6229 ER Maastricht, The Netherlands
| |
Collapse
|
15
|
Carbonic anhydrase 1 is a promising biomarker for early detection of non-small cell lung cancer. Tumour Biol 2015; 37:553-9. [PMID: 26232327 DOI: 10.1007/s13277-015-3834-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/21/2015] [Indexed: 10/23/2022] Open
Abstract
This study aimed to identify candidate biomarkers associated with stage I non-small cell lung cancer (NSCLC). Sera from three groups, a lung cancer group (n = 11), benign control group (n = 12), and normal control group (n = 10), were collected and pooled. Protein expression profiles were analyzed by a combination of two-dimensional electrophoresis (2DE) and matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS). These methods were used to separate, screen, and identify proteins that were differentially expressed between stage I NSCLC and controls. Differentially expressed proteins were validated by both Western blot and ELISA in an expanded sample size (22, 18, and 18 in three groups, respectively). MALDI-MS identified 12 differentially expressed proteins in the lung cancer group compared to the two control groups. Expression of carbonic anhydrase 1 (CA1) was validated by Western blot. CA1 was significantly elevated in the lung cancer group compared to controls. ELISA results confirmed that CA1 in the lung cancer group (3.18 ± 1.27 ng/mL, n = 22) was highly expressed in stage I NSCLC patients compared to those in the benign control group (2.21 ± 0.71 ng/mL, n = 18) and the normal control group (2.04 ± 0.63 ng/mL, n = 18) (P = 0.001). In conclusion, we provide evidence that CA1 is highly expressed in the sera of stage I NSCLC patients. Additionally, CA1 might serve as a novel biomarker for early detection of NSCLC.
Collapse
|
16
|
Li J, Zhang G, Wang X, Li XF. Is carbonic anhydrase IX a validated target for molecular imaging of cancer and hypoxia? Future Oncol 2015; 11:1531-41. [PMID: 25963430 PMCID: PMC4976829 DOI: 10.2217/fon.15.11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The presence of hypoxia is a general feature of most solid malignancies, and hypoxia is considered as one of major factors for anticancer therapy failure. Carbonic anhydrase IX (CAIX) has been reported to be an endogenous hypoxia marker, CAIX monoclonal antibodies, their segments and inhibitors are developed for CAIX imaging. However, growing evidence indicates that CAIX expression under hypoxia condition may be cancer cell lines or cancer-type dependent. Here we review the current literature on CAIX and discuss the advantage and limitation of CAIX as a target for tumor hypoxia imaging. Accordingly, CAIX would be unreliable as a universal target for cancer and tumor hypoxia visualization.
Collapse
Affiliation(s)
- Jianbo Li
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| | - Guojian Zhang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| | - Xuemei Wang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Xiao-Feng Li
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| |
Collapse
|